Skip to main content
Top
Published in: BMC Medicine 1/2020

01-12-2020 | Schizophrenia | Review

Endothelial dysfunction in neuroprogressive disorders—causes and suggested treatments

Authors: Gerwyn Morris, Basant K. Puri, Lisa Olive, Andre Carvalho, Michael Berk, Ken Walder, Lise Tuset Gustad, Michael Maes

Published in: BMC Medicine | Issue 1/2020

Login to get access

Abstract

Background

Potential routes whereby systemic inflammation, oxidative stress and mitochondrial dysfunction may drive the development of endothelial dysfunction and atherosclerosis, even in an environment of low cholesterol, are examined.

Main text

Key molecular players involved in the regulation of endothelial cell function are described, including PECAM-1, VE-cadherin, VEGFRs, SFK, Rho GEF TRIO, RAC-1, ITAM, SHP-2, MAPK/ERK, STAT-3, NF-κB, PI3K/AKT, eNOS, nitric oxide, miRNAs, KLF-4 and KLF-2. The key roles of platelet activation, xanthene oxidase and myeloperoxidase in the genesis of endothelial cell dysfunction and activation are detailed. The following roles of circulating reactive oxygen species (ROS), reactive nitrogen species and pro-inflammatory cytokines in the development of endothelial cell dysfunction are then described: paracrine signalling by circulating hydrogen peroxide, inhibition of eNOS and increased levels of mitochondrial ROS, including compromised mitochondrial dynamics, loss of calcium ion homeostasis and inactivation of SIRT-1-mediated signalling pathways. Next, loss of cellular redox homeostasis is considered, including further aspects of the roles of hydrogen peroxide signalling, the pathological consequences of elevated NF-κB, compromised S-nitrosylation and the development of hypernitrosylation and increased transcription of atherogenic miRNAs. These molecular aspects are then applied to neuroprogressive disorders by considering the following potential generators of endothelial dysfunction and activation in major depressive disorder, bipolar disorder and schizophrenia: NF-κB; platelet activation; atherogenic miRs; myeloperoxidase; xanthene oxidase and uric acid; and inflammation, oxidative stress, nitrosative stress and mitochondrial dysfunction.

Conclusions

Finally, on the basis of the above molecular mechanisms, details are given of potential treatment options for mitigating endothelial cell dysfunction and activation in neuroprogressive disorders.
Literature
1.
go back to reference Wu Q, Kling JM. Depression and the risk of myocardial infarction and coronary death: a meta-analysis of prospective cohort studies. Medicine (Baltimore). 2016;95(6):e2815.CrossRef Wu Q, Kling JM. Depression and the risk of myocardial infarction and coronary death: a meta-analysis of prospective cohort studies. Medicine (Baltimore). 2016;95(6):e2815.CrossRef
2.
go back to reference Gan Y, Gong Y, Tong X, Sun H, Cong Y, Dong X, Wang Y, Xu X, Yin X, Deng J, et al. Depression and the risk of coronary heart disease: a meta-analysis of prospective cohort studies. BMC Psychiatry. 2014;14:371.PubMedPubMedCentralCrossRef Gan Y, Gong Y, Tong X, Sun H, Cong Y, Dong X, Wang Y, Xu X, Yin X, Deng J, et al. Depression and the risk of coronary heart disease: a meta-analysis of prospective cohort studies. BMC Psychiatry. 2014;14:371.PubMedPubMedCentralCrossRef
3.
go back to reference Correll CU, Solmi M, Veronese N, Bortolato B, Rosson S, Santonastaso P, Thapa-Chhetri N, Fornaro M, Gallicchio D, Collantoni E, et al. Prevalence, incidence and mortality from cardiovascular disease in patients with pooled and specific severe mental illness: a large-scale meta-analysis of 3,211,768 patients and 113,383,368 controls. World Psychiatry. 2017;16(2):163–80.PubMedPubMedCentralCrossRef Correll CU, Solmi M, Veronese N, Bortolato B, Rosson S, Santonastaso P, Thapa-Chhetri N, Fornaro M, Gallicchio D, Collantoni E, et al. Prevalence, incidence and mortality from cardiovascular disease in patients with pooled and specific severe mental illness: a large-scale meta-analysis of 3,211,768 patients and 113,383,368 controls. World Psychiatry. 2017;16(2):163–80.PubMedPubMedCentralCrossRef
4.
5.
go back to reference Hayes JF, Miles J, Walters K, King M, Osborn DP. A systematic review and meta-analysis of premature mortality in bipolar affective disorder. Acta Psychiatr Scand. 2015;131(6):417–25.PubMedPubMedCentralCrossRef Hayes JF, Miles J, Walters K, King M, Osborn DP. A systematic review and meta-analysis of premature mortality in bipolar affective disorder. Acta Psychiatr Scand. 2015;131(6):417–25.PubMedPubMedCentralCrossRef
6.
go back to reference Goldstein BI, Carnethon MR, Matthews KA, McIntyre RS, Miller GE, Raghuveer G, Stoney CM, Wasiak H, McCrindle BW. Major depressive disorder and bipolar disorder predispose youth to accelerated atherosclerosis and early cardiovascular disease: a scientific statement from the American Heart Association. Circulation. 2015;132(10):965–86.PubMedCrossRef Goldstein BI, Carnethon MR, Matthews KA, McIntyre RS, Miller GE, Raghuveer G, Stoney CM, Wasiak H, McCrindle BW. Major depressive disorder and bipolar disorder predispose youth to accelerated atherosclerosis and early cardiovascular disease: a scientific statement from the American Heart Association. Circulation. 2015;132(10):965–86.PubMedCrossRef
7.
go back to reference Crump C, Sundquist K, Winkleby MA, Sundquist J. Comorbidities and mortality in bipolar disorder: a Swedish national cohort study. JAMA Psychiatry. 2013;70(9):931–9.PubMedCrossRef Crump C, Sundquist K, Winkleby MA, Sundquist J. Comorbidities and mortality in bipolar disorder: a Swedish national cohort study. JAMA Psychiatry. 2013;70(9):931–9.PubMedCrossRef
8.
go back to reference Gale CR, Batty GD, Osborn DP, Tynelius P, Rasmussen F. Mental disorders across the adult life course and future coronary heart disease: evidence for general susceptibility. Circulation. 2014;129(2):186–93.PubMedCrossRef Gale CR, Batty GD, Osborn DP, Tynelius P, Rasmussen F. Mental disorders across the adult life course and future coronary heart disease: evidence for general susceptibility. Circulation. 2014;129(2):186–93.PubMedCrossRef
9.
go back to reference Hayes JF, Marston L, Walters K, King MB, Osborn DPJ. Mortality gap for people with bipolar disorder and schizophrenia: UK-based cohort study 2000-2014. Br J Psychiatry. 2017;211(3):175–81.PubMedPubMedCentralCrossRef Hayes JF, Marston L, Walters K, King MB, Osborn DPJ. Mortality gap for people with bipolar disorder and schizophrenia: UK-based cohort study 2000-2014. Br J Psychiatry. 2017;211(3):175–81.PubMedPubMedCentralCrossRef
10.
go back to reference Aksay SS, Bumb JM, Janke C, Biemann R, Borucki K, Lederbogen F, Deuschle M, Sartorius A, Kranaster L. Serum lipid profile changes after successful treatment with electroconvulsive therapy in major depression: a prospective pilot trial. J Affect Disord. 2016;189:85–8.PubMedCrossRef Aksay SS, Bumb JM, Janke C, Biemann R, Borucki K, Lederbogen F, Deuschle M, Sartorius A, Kranaster L. Serum lipid profile changes after successful treatment with electroconvulsive therapy in major depression: a prospective pilot trial. J Affect Disord. 2016;189:85–8.PubMedCrossRef
11.
go back to reference Hummel J, Westphal S, Weber-Hamann B, Gilles M, Lederbogen F, Angermeier T, Luley C, Deuschle M, Kopf D. Serum lipoproteins improve after successful pharmacologic antidepressant treatment: a randomized open-label prospective trial. J Clin Psychiatry. 2011;72(7):885–91.PubMedCrossRef Hummel J, Westphal S, Weber-Hamann B, Gilles M, Lederbogen F, Angermeier T, Luley C, Deuschle M, Kopf D. Serum lipoproteins improve after successful pharmacologic antidepressant treatment: a randomized open-label prospective trial. J Clin Psychiatry. 2011;72(7):885–91.PubMedCrossRef
12.
go back to reference Ong KL, Morris MJ, McClelland RL, Maniam J, Allison MA, Rye KA. Lipids, lipoprotein distribution and depressive symptoms: the Multi-Ethnic Study of Atherosclerosis. Transl Psychiatry. 2016;6(11):e962.PubMedPubMedCentralCrossRef Ong KL, Morris MJ, McClelland RL, Maniam J, Allison MA, Rye KA. Lipids, lipoprotein distribution and depressive symptoms: the Multi-Ethnic Study of Atherosclerosis. Transl Psychiatry. 2016;6(11):e962.PubMedPubMedCentralCrossRef
13.
go back to reference Lehto SM, Niskanen L, Tolmunen T, Hintikka J, Viinamaki H, Heiskanen T, Honkalampi K, Kokkonen M, Koivumaa-Honkanen H. Low serum HDL-cholesterol levels are associated with long symptom duration in patients with major depressive disorder. Psychiatry Clin Neurosci. 2010;64(3):279–83.PubMedCrossRef Lehto SM, Niskanen L, Tolmunen T, Hintikka J, Viinamaki H, Heiskanen T, Honkalampi K, Kokkonen M, Koivumaa-Honkanen H. Low serum HDL-cholesterol levels are associated with long symptom duration in patients with major depressive disorder. Psychiatry Clin Neurosci. 2010;64(3):279–83.PubMedCrossRef
14.
go back to reference Parekh A, Smeeth D, Milner Y, Thure S. The role of lipid biomarkers in major depression. Healthcare (Basel). 2017;5(1):5.CrossRef Parekh A, Smeeth D, Milner Y, Thure S. The role of lipid biomarkers in major depression. Healthcare (Basel). 2017;5(1):5.CrossRef
15.
16.
go back to reference van Reedt Dortland AK, Giltay EJ, van Veen T, van Pelt J, Zitman FG, Penninx BW. Associations between serum lipids and major depressive disorder: results from the Netherlands Study of Depression and Anxiety (NESDA). J Clin Psychiatry. 2010;71(6):729–36.PubMedCrossRef van Reedt Dortland AK, Giltay EJ, van Veen T, van Pelt J, Zitman FG, Penninx BW. Associations between serum lipids and major depressive disorder: results from the Netherlands Study of Depression and Anxiety (NESDA). J Clin Psychiatry. 2010;71(6):729–36.PubMedCrossRef
17.
go back to reference Peterfalvi A, Nemeth N, Herczeg R, Tenyi T, Miseta A, Czeh B, Simon M. Examining the influence of early life stress on serum lipid profiles and cognitive functioning in depressed patients. Front Psychol. 2019;10:1798.PubMedPubMedCentralCrossRef Peterfalvi A, Nemeth N, Herczeg R, Tenyi T, Miseta A, Czeh B, Simon M. Examining the influence of early life stress on serum lipid profiles and cognitive functioning in depressed patients. Front Psychol. 2019;10:1798.PubMedPubMedCentralCrossRef
18.
go back to reference Huang YJ, Tsai SY, Chung KH, Chen PH, Huang SH, Kuo CJ. State-dependent alterations of lipid profiles in patients with bipolar disorder. Int J Psychiatry Med. 2018;53(4):273–81.PubMedCrossRef Huang YJ, Tsai SY, Chung KH, Chen PH, Huang SH, Kuo CJ. State-dependent alterations of lipid profiles in patients with bipolar disorder. Int J Psychiatry Med. 2018;53(4):273–81.PubMedCrossRef
19.
go back to reference Wysokinski A, Strzelecki D, Kloszewska I. Levels of triglycerides, cholesterol, LDL, HDL and glucose in patients with schizophrenia, unipolar depression and bipolar disorder. Diab Metabol Syndrome. 2015;9(3):168–76.CrossRef Wysokinski A, Strzelecki D, Kloszewska I. Levels of triglycerides, cholesterol, LDL, HDL and glucose in patients with schizophrenia, unipolar depression and bipolar disorder. Diab Metabol Syndrome. 2015;9(3):168–76.CrossRef
20.
go back to reference Su M, Li E, Tang C, Zhao Y, Liu R, Gao K. Comparison of blood lipid profile/thyroid function markers between unipolar and bipolar depressed patients and in depressed patients with anhedonia or suicidal thoughts. Mol Med. 2019;25(1):51.PubMedPubMedCentralCrossRef Su M, Li E, Tang C, Zhao Y, Liu R, Gao K. Comparison of blood lipid profile/thyroid function markers between unipolar and bipolar depressed patients and in depressed patients with anhedonia or suicidal thoughts. Mol Med. 2019;25(1):51.PubMedPubMedCentralCrossRef
21.
go back to reference Huang JT, Wang L, Prabakaran S, Wengenroth M, Lockstone HE, Koethe D, Gerth CW, Gross S, Schreiber D, Lilley K, et al. Independent protein-profiling studies show a decrease in apolipoprotein A1 levels in schizophrenia CSF, brain and peripheral tissues. Mol Psychiatry. 2008;13(12):1118–28.PubMedCrossRef Huang JT, Wang L, Prabakaran S, Wengenroth M, Lockstone HE, Koethe D, Gerth CW, Gross S, Schreiber D, Lilley K, et al. Independent protein-profiling studies show a decrease in apolipoprotein A1 levels in schizophrenia CSF, brain and peripheral tissues. Mol Psychiatry. 2008;13(12):1118–28.PubMedCrossRef
22.
go back to reference Misiak B, Stanczykiewicz B, Laczmanski L, Frydecka D. Lipid profile disturbances in antipsychotic-naive patients with first-episode non-affective psychosis: a systematic review and meta-analysis. Schizophr Res. 2017;190:18–27.PubMedCrossRef Misiak B, Stanczykiewicz B, Laczmanski L, Frydecka D. Lipid profile disturbances in antipsychotic-naive patients with first-episode non-affective psychosis: a systematic review and meta-analysis. Schizophr Res. 2017;190:18–27.PubMedCrossRef
23.
go back to reference Pillinger T, Beck K, Stubbs B, Howes OD. Cholesterol and triglyceride levels in first-episode psychosis: systematic review and meta-analysis. Br J Psychiatry. 2017;211(6):339–49.PubMedPubMedCentralCrossRef Pillinger T, Beck K, Stubbs B, Howes OD. Cholesterol and triglyceride levels in first-episode psychosis: systematic review and meta-analysis. Br J Psychiatry. 2017;211(6):339–49.PubMedPubMedCentralCrossRef
24.
go back to reference Zhang J, Chen L, Delzell E, Muntner P, Hillegass WB, Safford MM, Millan IY, Crowson CS, Curtis JR. The association between inflammatory markers, serum lipids and the risk of cardiovascular events in patients with rheumatoid arthritis. Ann Rheum Dis. 2014;73(7):1301–8.PubMedCrossRef Zhang J, Chen L, Delzell E, Muntner P, Hillegass WB, Safford MM, Millan IY, Crowson CS, Curtis JR. The association between inflammatory markers, serum lipids and the risk of cardiovascular events in patients with rheumatoid arthritis. Ann Rheum Dis. 2014;73(7):1301–8.PubMedCrossRef
25.
go back to reference Myasoedova E, Crowson CS, Kremers HM, Roger VL, Fitz-Gibbon PD, Therneau TM, Gabriel SE. Lipid paradox in rheumatoid arthritis: the impact of serum lipid measures and systemic inflammation on the risk of cardiovascular disease. Ann Rheum Dis. 2011;70(3):482–7.PubMedCrossRef Myasoedova E, Crowson CS, Kremers HM, Roger VL, Fitz-Gibbon PD, Therneau TM, Gabriel SE. Lipid paradox in rheumatoid arthritis: the impact of serum lipid measures and systemic inflammation on the risk of cardiovascular disease. Ann Rheum Dis. 2011;70(3):482–7.PubMedCrossRef
26.
go back to reference Lind L, Lithell H. Impaired glucose and lipid metabolism seen in intensive care patients is related to severity of illness and survival. Clin Intensive Care. 1994;5(3):100–5.PubMed Lind L, Lithell H. Impaired glucose and lipid metabolism seen in intensive care patients is related to severity of illness and survival. Clin Intensive Care. 1994;5(3):100–5.PubMed
27.
go back to reference Gabay C, Kushner I. Acute-phase proteins and other systemic responses to inflammation. N Engl J Med. 1999;340(6):448–54.PubMedCrossRef Gabay C, Kushner I. Acute-phase proteins and other systemic responses to inflammation. N Engl J Med. 1999;340(6):448–54.PubMedCrossRef
28.
go back to reference Popa C, Netea MG, van Riel PL, van der Meer JW, Stalenhoef AF. The role of TNF-alpha in chronic inflammatory conditions, intermediary metabolism, and cardiovascular risk. J Lipid Res. 2007;48(4):751–62.PubMedCrossRef Popa C, Netea MG, van Riel PL, van der Meer JW, Stalenhoef AF. The role of TNF-alpha in chronic inflammatory conditions, intermediary metabolism, and cardiovascular risk. J Lipid Res. 2007;48(4):751–62.PubMedCrossRef
29.
go back to reference Feingold KR, Grunfeld C. Effect of inflammation on HDL structure and function. Curr Opin Lipidol. 2016;27(5):521–30.PubMedCrossRef Feingold KR, Grunfeld C. Effect of inflammation on HDL structure and function. Curr Opin Lipidol. 2016;27(5):521–30.PubMedCrossRef
30.
go back to reference Khovidhunkit W, Kim MS, Memon RA, Shigenaga JK, Moser AH, Feingold KR, Grunfeld C. Effects of infection and inflammation on lipid and lipoprotein metabolism: mechanisms and consequences to the host. J Lipid Res. 2004;45(7):1169–96.PubMedCrossRef Khovidhunkit W, Kim MS, Memon RA, Shigenaga JK, Moser AH, Feingold KR, Grunfeld C. Effects of infection and inflammation on lipid and lipoprotein metabolism: mechanisms and consequences to the host. J Lipid Res. 2004;45(7):1169–96.PubMedCrossRef
31.
go back to reference Fillman SG, Weickert TW, Lenroot RK, Catts SV, Bruggemann JM, Catts VS, Weickert CS. Elevated peripheral cytokines characterize a subgroup of people with schizophrenia displaying poor verbal fluency and reduced Broca's area volume. Mol Psychiatry. 2016;21(8):1090–8.PubMedCrossRef Fillman SG, Weickert TW, Lenroot RK, Catts SV, Bruggemann JM, Catts VS, Weickert CS. Elevated peripheral cytokines characterize a subgroup of people with schizophrenia displaying poor verbal fluency and reduced Broca's area volume. Mol Psychiatry. 2016;21(8):1090–8.PubMedCrossRef
32.
go back to reference Boerrigter D, Weickert TW, Lenroot R, O'Donnell M, Galletly C, Liu D, Burgess M, Cadiz R, Jacomb I, Catts VS, et al. Using blood cytokine measures to define high inflammatory biotype of schizophrenia and schizoaffective disorder. J Neuroinflammation. 2017;14(1):188.PubMedPubMedCentralCrossRef Boerrigter D, Weickert TW, Lenroot R, O'Donnell M, Galletly C, Liu D, Burgess M, Cadiz R, Jacomb I, Catts VS, et al. Using blood cytokine measures to define high inflammatory biotype of schizophrenia and schizoaffective disorder. J Neuroinflammation. 2017;14(1):188.PubMedPubMedCentralCrossRef
33.
go back to reference Momtazmanesh S, Zare-Shahabadi A, Rezaei N. Cytokine alterations in schizophrenia: an updated review. Front Psychiatry. 2019;10(892). Momtazmanesh S, Zare-Shahabadi A, Rezaei N. Cytokine alterations in schizophrenia: an updated review. Front Psychiatry. 2019;10(892).
34.
go back to reference Modabbernia A, Taslimi S, Brietzke E, Ashrafi M. Cytokine alterations in bipolar disorder: a meta-analysis of 30 studies. Biol Psychiatry. 2013;74(1):15–25.PubMedCrossRef Modabbernia A, Taslimi S, Brietzke E, Ashrafi M. Cytokine alterations in bipolar disorder: a meta-analysis of 30 studies. Biol Psychiatry. 2013;74(1):15–25.PubMedCrossRef
35.
go back to reference Brietzke E, Stertz L, Fernandes BS, Kauer-Sant'anna M, Mascarenhas M, Escosteguy Vargas A, Chies JA, Kapczinski F. Comparison of cytokine levels in depressed, manic and euthymic patients with bipolar disorder. J Affect Disord. 2009;116(3):214–7.PubMedCrossRef Brietzke E, Stertz L, Fernandes BS, Kauer-Sant'anna M, Mascarenhas M, Escosteguy Vargas A, Chies JA, Kapczinski F. Comparison of cytokine levels in depressed, manic and euthymic patients with bipolar disorder. J Affect Disord. 2009;116(3):214–7.PubMedCrossRef
36.
go back to reference Munkholm K, Vinberg M, Vedel Kessing L. Cytokines in bipolar disorder: a systematic review and meta-analysis. J Affect Disord. 2013;144(1–2):16–27.PubMedCrossRef Munkholm K, Vinberg M, Vedel Kessing L. Cytokines in bipolar disorder: a systematic review and meta-analysis. J Affect Disord. 2013;144(1–2):16–27.PubMedCrossRef
37.
go back to reference Goldsmith DR, Rapaport MH, Miller BJ. A meta-analysis of blood cytokine network alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder and depression. Mol Psychiatry. 2016;21(12):1696–709.PubMedPubMedCentralCrossRef Goldsmith DR, Rapaport MH, Miller BJ. A meta-analysis of blood cytokine network alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder and depression. Mol Psychiatry. 2016;21(12):1696–709.PubMedPubMedCentralCrossRef
38.
go back to reference Maes M, Berk M, Goehler L, Song C, Anderson G, Galecki P, Leonard B. Depression and sickness behavior are Janus-faced responses to shared inflammatory pathways. BMC Med. 2012;10:66.PubMedPubMedCentralCrossRef Maes M, Berk M, Goehler L, Song C, Anderson G, Galecki P, Leonard B. Depression and sickness behavior are Janus-faced responses to shared inflammatory pathways. BMC Med. 2012;10:66.PubMedPubMedCentralCrossRef
39.
go back to reference Berk M, Williams LJ, Jacka FN, O’Neil A, Pasco JA, Moylan S, Allen NB, Stuart AL, Hayley AC, Byrne ML, et al. So depression is an inflammatory disease, but where does the inflammation come from? BMC Med. 2013;11(1):200.PubMedPubMedCentralCrossRef Berk M, Williams LJ, Jacka FN, O’Neil A, Pasco JA, Moylan S, Allen NB, Stuart AL, Hayley AC, Byrne ML, et al. So depression is an inflammatory disease, but where does the inflammation come from? BMC Med. 2013;11(1):200.PubMedPubMedCentralCrossRef
40.
go back to reference Chen LP, Dai HY, Dai ZZ, Xu CT, Wu RH. Anterior cingulate cortex and cerebellar hemisphere neurometabolite changes in depression treatment: a 1H magnetic resonance spectroscopy study. Psychiatry Clin Neurosci. 2014;68(5):357–64.PubMedCrossRef Chen LP, Dai HY, Dai ZZ, Xu CT, Wu RH. Anterior cingulate cortex and cerebellar hemisphere neurometabolite changes in depression treatment: a 1H magnetic resonance spectroscopy study. Psychiatry Clin Neurosci. 2014;68(5):357–64.PubMedCrossRef
41.
go back to reference Zou W, Feng R, Yang Y. Changes in the serum levels of inflammatory cytokines in antidepressant drug-naive patients with major depression. PLoS One. 2018;13(6):e0197267.PubMedPubMedCentralCrossRef Zou W, Feng R, Yang Y. Changes in the serum levels of inflammatory cytokines in antidepressant drug-naive patients with major depression. PLoS One. 2018;13(6):e0197267.PubMedPubMedCentralCrossRef
42.
go back to reference Taleb S. Inflammation in atherosclerosis. Arch Cardiovas Dis. 2016;109(12):708–15.CrossRef Taleb S. Inflammation in atherosclerosis. Arch Cardiovas Dis. 2016;109(12):708–15.CrossRef
43.
go back to reference Bäck M, Yurdagul A, Tabas I, Öörni K, Kovanen PT. Inflammation and its resolution in atherosclerosis: mediators and therapeutic opportunities. Nat Rev Cardiol. 2019;16(7):389–406.PubMedPubMedCentral Bäck M, Yurdagul A, Tabas I, Öörni K, Kovanen PT. Inflammation and its resolution in atherosclerosis: mediators and therapeutic opportunities. Nat Rev Cardiol. 2019;16(7):389–406.PubMedPubMedCentral
44.
go back to reference Liu Y, Tie L. Apolipoprotein M and sphingosine-1-phosphate complex alleviates TNF-alpha-induced endothelial cell injury and inflammation through PI3K/AKT signaling pathway. BMC Cardiovasc Disord. 2019;19(1):279.PubMedPubMedCentralCrossRef Liu Y, Tie L. Apolipoprotein M and sphingosine-1-phosphate complex alleviates TNF-alpha-induced endothelial cell injury and inflammation through PI3K/AKT signaling pathway. BMC Cardiovasc Disord. 2019;19(1):279.PubMedPubMedCentralCrossRef
45.
go back to reference Panchal SK, Brown L. Cholesterol versus inflammation as cause of chronic diseases. Nutrients. 2019;11(10). Panchal SK, Brown L. Cholesterol versus inflammation as cause of chronic diseases. Nutrients. 2019;11(10).
46.
go back to reference Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377(12):1119–31.PubMedCrossRef Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377(12):1119–31.PubMedCrossRef
47.
48.
go back to reference Yang X, Li Y, Li Y, Ren X, Zhang X, Hu D, Gao Y, Xing Y, Shang H. Oxidative stress-mediated atherosclerosis: mechanisms and therapies. Front Physiol. 2017;8:600.PubMedPubMedCentralCrossRef Yang X, Li Y, Li Y, Ren X, Zhang X, Hu D, Gao Y, Xing Y, Shang H. Oxidative stress-mediated atherosclerosis: mechanisms and therapies. Front Physiol. 2017;8:600.PubMedPubMedCentralCrossRef
49.
go back to reference Kattoor AJ, Pothineni NVK, Palagiri D, Mehta JL. Oxidative stress in atherosclerosis. Curr Atheroscler Rep. 2017;19(11):42.PubMedCrossRef Kattoor AJ, Pothineni NVK, Palagiri D, Mehta JL. Oxidative stress in atherosclerosis. Curr Atheroscler Rep. 2017;19(11):42.PubMedCrossRef
50.
go back to reference Marchio P, Guerra-Ojeda S, Vila JM, Aldasoro M, Victor VM, Mauricio MD. Targeting early atherosclerosis: a focus on oxidative stress and inflammation. Oxidative Med Cell Longev. 2019;2019:32.CrossRef Marchio P, Guerra-Ojeda S, Vila JM, Aldasoro M, Victor VM, Mauricio MD. Targeting early atherosclerosis: a focus on oxidative stress and inflammation. Oxidative Med Cell Longev. 2019;2019:32.CrossRef
51.
go back to reference Madamanchi NR, Runge MS. Mitochondrial dysfunction in atherosclerosis. Circ Res. 2007;100(4):460–73.PubMedCrossRef Madamanchi NR, Runge MS. Mitochondrial dysfunction in atherosclerosis. Circ Res. 2007;100(4):460–73.PubMedCrossRef
52.
go back to reference Peng W, Cai G, Xia Y, Chen J, Wu P, Wang Z, Li G, Wei D. Mitochondrial dysfunction in atherosclerosis. DNA Cell Biol. 2019;38(7):597–606.PubMedCrossRef Peng W, Cai G, Xia Y, Chen J, Wu P, Wang Z, Li G, Wei D. Mitochondrial dysfunction in atherosclerosis. DNA Cell Biol. 2019;38(7):597–606.PubMedCrossRef
53.
go back to reference Yu EPK, Reinhold J, Yu H, Starks L, Uryga AK, Foote K, Finigan A, Figg N, Pung YF, Logan A, et al. Mitochondrial respiration is reduced in atherosclerosis, promoting necrotic core formation and reducing relative fibrous cap thickness. Arterioscler Thromb Vasc Biol. 2017;37(12):2322–32.PubMedPubMedCentralCrossRef Yu EPK, Reinhold J, Yu H, Starks L, Uryga AK, Foote K, Finigan A, Figg N, Pung YF, Logan A, et al. Mitochondrial respiration is reduced in atherosclerosis, promoting necrotic core formation and reducing relative fibrous cap thickness. Arterioscler Thromb Vasc Biol. 2017;37(12):2322–32.PubMedPubMedCentralCrossRef
55.
56.
go back to reference Morris G, Puri BK, Walker AJ, Maes M, Carvalho AF, Walder K, Berk M. Shared pathways for neuroprogression and somatoprogression in neuropsychiatric disorders. Neurosci Biobehav Rev. 2019;107:862–882. Morris G, Puri BK, Walker AJ, Maes M, Carvalho AF, Walder K, Berk M. Shared pathways for neuroprogression and somatoprogression in neuropsychiatric disorders. Neurosci Biobehav Rev. 2019;107:862–882.
57.
go back to reference Morris G, Stubbs B, Kohler CA, Walder K, Slyepchenko A, Berk M, Carvalho AF. The putative role of oxidative stress and inflammation in the pathophysiology of sleep dysfunction across neuropsychiatric disorders: focus on chronic fatigue syndrome, bipolar disorder and multiple sclerosis. Sleep Med Rev. 2018;41:255–65.PubMedCrossRef Morris G, Stubbs B, Kohler CA, Walder K, Slyepchenko A, Berk M, Carvalho AF. The putative role of oxidative stress and inflammation in the pathophysiology of sleep dysfunction across neuropsychiatric disorders: focus on chronic fatigue syndrome, bipolar disorder and multiple sclerosis. Sleep Med Rev. 2018;41:255–65.PubMedCrossRef
58.
go back to reference Morris G, Walder K, McGee SL, Dean OM, Tye SJ, Maes M, Berk M. A model of the mitochondrial basis of bipolar disorder. Neurosci Biobehav Rev. 2017;74(Pt A):1–20.PubMedCrossRef Morris G, Walder K, McGee SL, Dean OM, Tye SJ, Maes M, Berk M. A model of the mitochondrial basis of bipolar disorder. Neurosci Biobehav Rev. 2017;74(Pt A):1–20.PubMedCrossRef
59.
go back to reference Pei L, Wallace DC. Mitochondrial etiology of neuropsychiatric disorders. Biol Psychiatry. 2018;83(9):722–30.PubMedCrossRef Pei L, Wallace DC. Mitochondrial etiology of neuropsychiatric disorders. Biol Psychiatry. 2018;83(9):722–30.PubMedCrossRef
61.
go back to reference Callaly E, Walder K, Morris G, Maes M, Debnath M, Berk M. Mitochondrial dysfunction in the pathophysiology of bipolar disorder: effects of pharmacotherapy. Mini Rev Medi Chemi. 2015;15(5):355–65.CrossRef Callaly E, Walder K, Morris G, Maes M, Debnath M, Berk M. Mitochondrial dysfunction in the pathophysiology of bipolar disorder: effects of pharmacotherapy. Mini Rev Medi Chemi. 2015;15(5):355–65.CrossRef
62.
go back to reference Szewczyk A, Jarmuszkiewicz W, Koziel A, Sobieraj I, Nobik W, Lukasiak A, Skup A, Bednarczyk P, Drabarek B, Dymkowska D, et al. Mitochondrial mechanisms of endothelial dysfunction. Pharmacol Rep. 2015;67(4):704–10.PubMedCrossRef Szewczyk A, Jarmuszkiewicz W, Koziel A, Sobieraj I, Nobik W, Lukasiak A, Skup A, Bednarczyk P, Drabarek B, Dymkowska D, et al. Mitochondrial mechanisms of endothelial dysfunction. Pharmacol Rep. 2015;67(4):704–10.PubMedCrossRef
65.
go back to reference Incalza MA, D'Oria R, Natalicchio A, Perrini S, Laviola L, Giorgino F. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vasc Pharmacol. 2018;100:1–19.CrossRef Incalza MA, D'Oria R, Natalicchio A, Perrini S, Laviola L, Giorgino F. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vasc Pharmacol. 2018;100:1–19.CrossRef
66.
go back to reference Haybar H, Shahrabi S, Rezaeeyan H, Shirzad R, Saki N. Endothelial cells: from dysfunction mechanism to pharmacological effect in cardiovascular disease. Cardiovasc Toxicol. 2019;19(1):13–22.PubMedCrossRef Haybar H, Shahrabi S, Rezaeeyan H, Shirzad R, Saki N. Endothelial cells: from dysfunction mechanism to pharmacological effect in cardiovascular disease. Cardiovasc Toxicol. 2019;19(1):13–22.PubMedCrossRef
67.
go back to reference Szmitko PE, Wang C-H, Weisel RD, JRd A, Anderson TJ, Verma S. New markers of inflammation and endothelial cell activation. Circulation. 2003;108(16):1917–23.PubMedCrossRef Szmitko PE, Wang C-H, Weisel RD, JRd A, Anderson TJ, Verma S. New markers of inflammation and endothelial cell activation. Circulation. 2003;108(16):1917–23.PubMedCrossRef
68.
go back to reference Teixeira BC, Lopes AL, Macedo RCO, Correa CS, Ramis TR, Ribeiro JL, Reischak-Oliveira A. Inflammatory markers, endothelial function and cardiovascular risk. J Vasc Brasileiro. 2014;13:108–15.CrossRef Teixeira BC, Lopes AL, Macedo RCO, Correa CS, Ramis TR, Ribeiro JL, Reischak-Oliveira A. Inflammatory markers, endothelial function and cardiovascular risk. J Vasc Brasileiro. 2014;13:108–15.CrossRef
71.
go back to reference Khan SY, Awad EM, Oszwald A, Mayr M, Yin X, Waltenberger B, Stuppner H, Lipovac M, Uhrin P, Breuss JM. Premature senescence of endothelial cells upon chronic exposure to TNFα can be prevented by N-acetyl cysteine and plumericin. Sci Rep. 2017;7(1):39501.PubMedPubMedCentralCrossRef Khan SY, Awad EM, Oszwald A, Mayr M, Yin X, Waltenberger B, Stuppner H, Lipovac M, Uhrin P, Breuss JM. Premature senescence of endothelial cells upon chronic exposure to TNFα can be prevented by N-acetyl cysteine and plumericin. Sci Rep. 2017;7(1):39501.PubMedPubMedCentralCrossRef
72.
go back to reference Widlansky ME, Gokce N, Keaney JF Jr, Vita JA. The clinical implications of endothelial dysfunction. J Am Coll Cardiol. 2003;42(7):1149–60.PubMedCrossRef Widlansky ME, Gokce N, Keaney JF Jr, Vita JA. The clinical implications of endothelial dysfunction. J Am Coll Cardiol. 2003;42(7):1149–60.PubMedCrossRef
74.
go back to reference Yeboah J, Folsom AR, Burke GL, Johnson C, Polak JF, Post W, Lima JA, Crouse JR, Herrington DM. Predictive value of brachial flow-mediated dilation for incident cardiovascular events in a population-based study: the multi-ethnic study of atherosclerosis. Circulation. 2009;120(6):502–9.PubMedPubMedCentralCrossRef Yeboah J, Folsom AR, Burke GL, Johnson C, Polak JF, Post W, Lima JA, Crouse JR, Herrington DM. Predictive value of brachial flow-mediated dilation for incident cardiovascular events in a population-based study: the multi-ethnic study of atherosclerosis. Circulation. 2009;120(6):502–9.PubMedPubMedCentralCrossRef
75.
go back to reference Rubinshtein R, Kuvin JT, Soffler M, Lennon RJ, Lavi S, Nelson RE, Pumper GM, Lerman LO, Lerman A. Assessment of endothelial function by non-invasive peripheral arterial tonometry predicts late cardiovascular adverse events. Eur Heart J. 2010;31(9):1142–8.PubMedCrossRef Rubinshtein R, Kuvin JT, Soffler M, Lennon RJ, Lavi S, Nelson RE, Pumper GM, Lerman LO, Lerman A. Assessment of endothelial function by non-invasive peripheral arterial tonometry predicts late cardiovascular adverse events. Eur Heart J. 2010;31(9):1142–8.PubMedCrossRef
76.
go back to reference Widmer RJ, Lerman A. Endothelial dysfunction and cardiovascular disease. Glob Cardiol Sci Pract. 2014;2014(3):291–308.PubMedPubMedCentral Widmer RJ, Lerman A. Endothelial dysfunction and cardiovascular disease. Glob Cardiol Sci Pract. 2014;2014(3):291–308.PubMedPubMedCentral
77.
go back to reference Shechter M, Shechter A, Koren-Morag N, Feinberg MS, Hiersch L. Usefulness of brachial artery flow-mediated dilation to predict long-term cardiovascular events in subjects without heart disease. Am J Cardiol. 2014;113(1):162–7.PubMedCrossRef Shechter M, Shechter A, Koren-Morag N, Feinberg MS, Hiersch L. Usefulness of brachial artery flow-mediated dilation to predict long-term cardiovascular events in subjects without heart disease. Am J Cardiol. 2014;113(1):162–7.PubMedCrossRef
78.
go back to reference Oral E, Halici Z, Cinar I, Ozcan E, Kutlu Z. Evaluation of endothelial dysfunction in bipolar affective disorders: serum endocan and urotensin-II levels. Clin Psychopharmacol Neurosci. 2019;17(2):211–21.PubMedPubMedCentralCrossRef Oral E, Halici Z, Cinar I, Ozcan E, Kutlu Z. Evaluation of endothelial dysfunction in bipolar affective disorders: serum endocan and urotensin-II levels. Clin Psychopharmacol Neurosci. 2019;17(2):211–21.PubMedPubMedCentralCrossRef
79.
go back to reference Goldstein BI, Young LT. Toward clinically applicable biomarkers in bipolar disorder: focus on BDNF, inflammatory markers, and endothelial function. Curr Psychiatry Rep. 2013;15(12):425.PubMedPubMedCentralCrossRef Goldstein BI, Young LT. Toward clinically applicable biomarkers in bipolar disorder: focus on BDNF, inflammatory markers, and endothelial function. Curr Psychiatry Rep. 2013;15(12):425.PubMedPubMedCentralCrossRef
80.
go back to reference Vetter MW, Martin B-J, Fung M, Pajevic M, Anderson TJ, Raedler TJ. Microvascular dysfunction in schizophrenia: a case–control study. NPJ Schizophr. 2015;1(1):15023.PubMedPubMedCentralCrossRef Vetter MW, Martin B-J, Fung M, Pajevic M, Anderson TJ, Raedler TJ. Microvascular dysfunction in schizophrenia: a case–control study. NPJ Schizophr. 2015;1(1):15023.PubMedPubMedCentralCrossRef
81.
go back to reference Nguyen TT, Dev SI, Chen G, Liou SC, Martin AS, Irwin MR, Carroll JE, Tu X, Jeste DV, Eyler LT. Abnormal levels of vascular endothelial biomarkers in schizophrenia. Eur Arch Psychiatry Clin Neurosci. 2018;268(8):849–60.PubMedCrossRef Nguyen TT, Dev SI, Chen G, Liou SC, Martin AS, Irwin MR, Carroll JE, Tu X, Jeste DV, Eyler LT. Abnormal levels of vascular endothelial biomarkers in schizophrenia. Eur Arch Psychiatry Clin Neurosci. 2018;268(8):849–60.PubMedCrossRef
82.
go back to reference Rybakowski JK, Wykretowicz A, Heymann-Szlachcinska A, Wysocki H. Impairment of endothelial function in unipolar and bipolar depression. Biol Psychiatry. 2006;60(8):889–91.PubMedCrossRef Rybakowski JK, Wykretowicz A, Heymann-Szlachcinska A, Wysocki H. Impairment of endothelial function in unipolar and bipolar depression. Biol Psychiatry. 2006;60(8):889–91.PubMedCrossRef
83.
84.
go back to reference Higgins P, Dawson J, Lees KR, McArthur K, Quinn TJ, Walters MR. Xanthine oxidase inhibition for the treatment of cardiovascular disease: a systematic review and meta-analysis. Cardiovasc Ther. 2012;30(4):217–26.PubMedCrossRef Higgins P, Dawson J, Lees KR, McArthur K, Quinn TJ, Walters MR. Xanthine oxidase inhibition for the treatment of cardiovascular disease: a systematic review and meta-analysis. Cardiovasc Ther. 2012;30(4):217–26.PubMedCrossRef
85.
go back to reference Nussbaum C, Klinke A, Adam M, Baldus S, Sperandio M. Myeloperoxidase: a leukocyte-derived protagonist of inflammation and cardiovascular disease. Antioxid Redox Signal. 2013;18(6):692–713.PubMedCrossRef Nussbaum C, Klinke A, Adam M, Baldus S, Sperandio M. Myeloperoxidase: a leukocyte-derived protagonist of inflammation and cardiovascular disease. Antioxid Redox Signal. 2013;18(6):692–713.PubMedCrossRef
86.
go back to reference Rajendran P, Rengarajan T, Thangavel J, Nishigaki Y, Sakthisekaran D, Sethi G, Nishigaki I. The vascular endothelium and human diseases. Int J Biol Sci. 2013;9(10):1057–69.PubMedPubMedCentralCrossRef Rajendran P, Rengarajan T, Thangavel J, Nishigaki Y, Sakthisekaran D, Sethi G, Nishigaki I. The vascular endothelium and human diseases. Int J Biol Sci. 2013;9(10):1057–69.PubMedPubMedCentralCrossRef
87.
go back to reference Fledderus JO, Boon RA, Volger OL, Hurttila H, Yla-Herttuala S, Pannekoek H, Levonen AL, Horrevoets AJ. KLF2 primes the antioxidant transcription factor Nrf2 for activation in endothelial cells. Arterioscler Thromb Vasc Biol. 2008;28(7):1339–46.PubMedCrossRef Fledderus JO, Boon RA, Volger OL, Hurttila H, Yla-Herttuala S, Pannekoek H, Levonen AL, Horrevoets AJ. KLF2 primes the antioxidant transcription factor Nrf2 for activation in endothelial cells. Arterioscler Thromb Vasc Biol. 2008;28(7):1339–46.PubMedCrossRef
88.
go back to reference Nayak L, Lin Z, Jain MK. “Go with the flow”: how Krüppel-like factor 2 regulates the vasoprotective effects of shear stress. Antioxid Redox Signal. 2011;15(5):1449–61.PubMedPubMedCentralCrossRef Nayak L, Lin Z, Jain MK. “Go with the flow”: how Krüppel-like factor 2 regulates the vasoprotective effects of shear stress. Antioxid Redox Signal. 2011;15(5):1449–61.PubMedPubMedCentralCrossRef
89.
go back to reference Chiplunkar AR, Curtis BC, Eades GL, Kane MS, Fox SJ, Haar JL, Lloyd JA. The Kruppel-like factor 2 and Kruppel-like factor 4 genes interact to maintain endothelial integrity in mouse embryonic vasculogenesis. BMC Dev Biol. 2013;13:40.PubMedPubMedCentralCrossRef Chiplunkar AR, Curtis BC, Eades GL, Kane MS, Fox SJ, Haar JL, Lloyd JA. The Kruppel-like factor 2 and Kruppel-like factor 4 genes interact to maintain endothelial integrity in mouse embryonic vasculogenesis. BMC Dev Biol. 2013;13:40.PubMedPubMedCentralCrossRef
90.
go back to reference Young A, Wu W, Sun W, Benjamin Larman H, Wang N, Li YS, Shyy JY, Chien S, Garcia-Cardena G. Flow activation of AMP-activated protein kinase in vascular endothelium leads to Kruppel-like factor 2 expression. Arterioscler Thromb Vasc Biol. 2009;29(11):1902–8.PubMedPubMedCentralCrossRef Young A, Wu W, Sun W, Benjamin Larman H, Wang N, Li YS, Shyy JY, Chien S, Garcia-Cardena G. Flow activation of AMP-activated protein kinase in vascular endothelium leads to Kruppel-like factor 2 expression. Arterioscler Thromb Vasc Biol. 2009;29(11):1902–8.PubMedPubMedCentralCrossRef
91.
go back to reference Zhong F, Lee K, He JC. Role of Krüppel-like factor-2 in kidney disease. Nephrology. 2018;23(S4):53–6.PubMedCrossRef Zhong F, Lee K, He JC. Role of Krüppel-like factor-2 in kidney disease. Nephrology. 2018;23(S4):53–6.PubMedCrossRef
92.
go back to reference Doddaballapur A, Michalik KM, Manavski Y, Lucas T, Houtkooper RH, You X, Chen W, Zeiher AM, Potente M, Dimmeler S, et al. Laminar shear stress inhibits endothelial cell metabolism via KLF2-mediated repression of PFKFB3. Arterioscler Thromb Vasc Biol. 2015;35(1):137–45.PubMedCrossRef Doddaballapur A, Michalik KM, Manavski Y, Lucas T, Houtkooper RH, You X, Chen W, Zeiher AM, Potente M, Dimmeler S, et al. Laminar shear stress inhibits endothelial cell metabolism via KLF2-mediated repression of PFKFB3. Arterioscler Thromb Vasc Biol. 2015;35(1):137–45.PubMedCrossRef
93.
go back to reference Harding IC, Mitra R, Mensah SA, Nersesyan A, Bal NN, Ebong EE. Endothelial barrier reinforcement relies on flow-regulated glycocalyx, a potential therapeutic target. Biorheology. 2019;56(2–3):131–49.PubMedPubMedCentralCrossRef Harding IC, Mitra R, Mensah SA, Nersesyan A, Bal NN, Ebong EE. Endothelial barrier reinforcement relies on flow-regulated glycocalyx, a potential therapeutic target. Biorheology. 2019;56(2–3):131–49.PubMedPubMedCentralCrossRef
94.
95.
go back to reference Ganguli A, Persson L, Palmer IR, Evans I, Yang L, Smallwood R, Black R, Qwarnstrom EE. Distinct NF-kappaB regulation by shear stress through Ras-dependent IkappaBalpha oscillations: real-time analysis of flow-mediated activation in live cells. Circ Res. 2005;96(6):626–34.PubMedCrossRef Ganguli A, Persson L, Palmer IR, Evans I, Yang L, Smallwood R, Black R, Qwarnstrom EE. Distinct NF-kappaB regulation by shear stress through Ras-dependent IkappaBalpha oscillations: real-time analysis of flow-mediated activation in live cells. Circ Res. 2005;96(6):626–34.PubMedCrossRef
96.
go back to reference Mohan S, Koyoma K, Thangasamy A, Nakano H, Glickman RD, Mohan N. Low shear stress preferentially enhances IKK activity through selective sources of ROS for persistent activation of NF-kappaB in endothelial cells. Am J Physiol Cell Physiol. 2007;292(1):C362–71.PubMedCrossRef Mohan S, Koyoma K, Thangasamy A, Nakano H, Glickman RD, Mohan N. Low shear stress preferentially enhances IKK activity through selective sources of ROS for persistent activation of NF-kappaB in endothelial cells. Am J Physiol Cell Physiol. 2007;292(1):C362–71.PubMedCrossRef
97.
go back to reference Kadohama T, Akasaka N, Nishimura K, Hoshino Y, Sasajima T, Sumpio BE. p38 Mitogen-activated protein kinase activation in endothelial cell is implicated in cell alignment and elongation induced by fluid shear stress. Endothelium. 2006;13(1):43–50.PubMedCrossRef Kadohama T, Akasaka N, Nishimura K, Hoshino Y, Sasajima T, Sumpio BE. p38 Mitogen-activated protein kinase activation in endothelial cell is implicated in cell alignment and elongation induced by fluid shear stress. Endothelium. 2006;13(1):43–50.PubMedCrossRef
98.
go back to reference Cuhlmann S, Van der Heiden K, Saliba D, Tremoleda JL, Khalil M, Zakkar M, Chaudhury H, Luong le A, Mason JC, Udalova I, et al. Disturbed blood flow induces RelA expression via c-Jun N-terminal kinase 1: a novel mode of NF-kappaB regulation that promotes arterial inflammation. Circ Res. 2011;108(8):950–9.PubMedCrossRef Cuhlmann S, Van der Heiden K, Saliba D, Tremoleda JL, Khalil M, Zakkar M, Chaudhury H, Luong le A, Mason JC, Udalova I, et al. Disturbed blood flow induces RelA expression via c-Jun N-terminal kinase 1: a novel mode of NF-kappaB regulation that promotes arterial inflammation. Circ Res. 2011;108(8):950–9.PubMedCrossRef
99.
go back to reference Amaya R, Pierides A, Tarbell JM. The interaction between fluid wall shear stress and solid circumferential strain affects endothelial gene expression. PLoS One. 2015;10(7):e0129952.PubMedPubMedCentralCrossRef Amaya R, Pierides A, Tarbell JM. The interaction between fluid wall shear stress and solid circumferential strain affects endothelial gene expression. PLoS One. 2015;10(7):e0129952.PubMedPubMedCentralCrossRef
100.
go back to reference Baeriswyl DC, Prionisti I, Peach T, Tsolkas G, Chooi KY, Vardakis J, Morel S, Diagbouga MR, Bijlenga P, Cuhlmann S, et al. Disturbed flow induces a sustained, stochastic NF-κB activation which may support intracranial aneurysm growth in vivo. Sci Rep. 2019;9(1):4738.PubMedPubMedCentralCrossRef Baeriswyl DC, Prionisti I, Peach T, Tsolkas G, Chooi KY, Vardakis J, Morel S, Diagbouga MR, Bijlenga P, Cuhlmann S, et al. Disturbed flow induces a sustained, stochastic NF-κB activation which may support intracranial aneurysm growth in vivo. Sci Rep. 2019;9(1):4738.PubMedPubMedCentralCrossRef
101.
go back to reference Pan L, Hong Z, Yu L, Gao Y, Zhang R, Feng H, Su L, Wang G. Shear stress induces human aortic endothelial cell apoptosis via interleukin1 receptorassociated kinase 2induced endoplasmic reticulum stress. Mol Med Rep. 2017;16(5):7205.PubMedPubMedCentralCrossRef Pan L, Hong Z, Yu L, Gao Y, Zhang R, Feng H, Su L, Wang G. Shear stress induces human aortic endothelial cell apoptosis via interleukin1 receptorassociated kinase 2induced endoplasmic reticulum stress. Mol Med Rep. 2017;16(5):7205.PubMedPubMedCentralCrossRef
102.
go back to reference Bailey KA, Haj FG, Simon SI, Passerini AG. Atherosusceptible shear stress activates endoplasmic reticulum stress to promote endothelial inflammation. Sci Rep. 2017;7(1):8196.PubMedPubMedCentralCrossRef Bailey KA, Haj FG, Simon SI, Passerini AG. Atherosusceptible shear stress activates endoplasmic reticulum stress to promote endothelial inflammation. Sci Rep. 2017;7(1):8196.PubMedPubMedCentralCrossRef
103.
go back to reference Maamoun H, Abdelsalam SS, Zeidan A, Korashy HM, Agouni A. Endoplasmic reticulum stress: a critical molecular driver of endothelial dysfunction and cardiovascular disturbances associated with diabetes. Int J Mol Sci. 2019;20(7):1658.PubMedCentralCrossRef Maamoun H, Abdelsalam SS, Zeidan A, Korashy HM, Agouni A. Endoplasmic reticulum stress: a critical molecular driver of endothelial dysfunction and cardiovascular disturbances associated with diabetes. Int J Mol Sci. 2019;20(7):1658.PubMedCentralCrossRef
105.
go back to reference Warboys CM, de Luca A, Amini N, Luong L, Duckles H, Hsiao S, White A, Biswas S, Khamis R, Chong CK, et al. Disturbed flow promotes endothelial senescence via a p53-dependent pathway. Arterioscler Thromb Vasc Biol. 2014;34(5):985–95.PubMedCrossRef Warboys CM, de Luca A, Amini N, Luong L, Duckles H, Hsiao S, White A, Biswas S, Khamis R, Chong CK, et al. Disturbed flow promotes endothelial senescence via a p53-dependent pathway. Arterioscler Thromb Vasc Biol. 2014;34(5):985–95.PubMedCrossRef
106.
go back to reference Vion A-C, Kheloufi M, Hammoutene A, Poisson J, Lasselin J, Devue C, Pic I, Dupont N, Busse J, Stark K, et al. Autophagy is required for endothelial cell alignment and atheroprotection under physiological blood flow. Proc Natl Acad Sci. 2017;114(41):E8675–84.PubMedCrossRefPubMedCentral Vion A-C, Kheloufi M, Hammoutene A, Poisson J, Lasselin J, Devue C, Pic I, Dupont N, Busse J, Stark K, et al. Autophagy is required for endothelial cell alignment and atheroprotection under physiological blood flow. Proc Natl Acad Sci. 2017;114(41):E8675–84.PubMedCrossRefPubMedCentral
107.
go back to reference Katsuumi G, Shimizu I, Yoshida Y, Minamino T: Vascular senescence in cardiovascular and metabolic diseases. Front Cardiovas Med 2018, 5(18). Katsuumi G, Shimizu I, Yoshida Y, Minamino T: Vascular senescence in cardiovascular and metabolic diseases. Front Cardiovas Med 2018, 5(18).
110.
go back to reference Mitroulis I, Alexaki VI, Kourtzelis I, Ziogas A, Hajishengallis G, Chavakis T. Leukocyte integrins: role in leukocyte recruitment and as therapeutic targets in inflammatory disease. Pharmacol Ther. 2015;147:123–35.PubMedCrossRef Mitroulis I, Alexaki VI, Kourtzelis I, Ziogas A, Hajishengallis G, Chavakis T. Leukocyte integrins: role in leukocyte recruitment and as therapeutic targets in inflammatory disease. Pharmacol Ther. 2015;147:123–35.PubMedCrossRef
111.
go back to reference Čejková S, Králová-Lesná I, Poledne R. Monocyte adhesion to the endothelium is an initial stage of atherosclerosis development. Cor et Vasa. 2016;58(4):e419–25.CrossRef Čejková S, Králová-Lesná I, Poledne R. Monocyte adhesion to the endothelium is an initial stage of atherosclerosis development. Cor et Vasa. 2016;58(4):e419–25.CrossRef
112.
go back to reference Ramji DP, Davies TS. Cytokines in atherosclerosis: key players in all stages of disease and promising therapeutic targets. Cytokine Growth Factor Rev. 2015;26(6):673–85.PubMedPubMedCentralCrossRef Ramji DP, Davies TS. Cytokines in atherosclerosis: key players in all stages of disease and promising therapeutic targets. Cytokine Growth Factor Rev. 2015;26(6):673–85.PubMedPubMedCentralCrossRef
113.
go back to reference Yu XH, Fu YC, Zhang DW, Yin K, Tang CK. Foam cells in atherosclerosis. Clin Chim Acta. 2013;424:245–52.PubMedCrossRef Yu XH, Fu YC, Zhang DW, Yin K, Tang CK. Foam cells in atherosclerosis. Clin Chim Acta. 2013;424:245–52.PubMedCrossRef
114.
go back to reference Chistiakov DA, Melnichenko AA, Myasoedova VA, Grechko AV, Orekhov AN. Mechanisms of foam cell formation in atherosclerosis. J Mol Med. 2017;95(11):1153–65.PubMedCrossRef Chistiakov DA, Melnichenko AA, Myasoedova VA, Grechko AV, Orekhov AN. Mechanisms of foam cell formation in atherosclerosis. J Mol Med. 2017;95(11):1153–65.PubMedCrossRef
115.
go back to reference Chistiakov DA, Bobryshev YV, Orekhov AN. Macrophage-mediated cholesterol handling in atherosclerosis. J Cell Mol Med. 2016;20(1):17–28.PubMedCrossRef Chistiakov DA, Bobryshev YV, Orekhov AN. Macrophage-mediated cholesterol handling in atherosclerosis. J Cell Mol Med. 2016;20(1):17–28.PubMedCrossRef
117.
go back to reference Conway DE, Breckenridge MT, Hinde E, Gratton E, Chen CS, Schwartz MA. Fluid shear stress on endothelial cells modulates mechanical tension across VE-cadherin and PECAM-1. Curr Biol. 2013;23(11):1024–30.PubMedPubMedCentralCrossRef Conway DE, Breckenridge MT, Hinde E, Gratton E, Chen CS, Schwartz MA. Fluid shear stress on endothelial cells modulates mechanical tension across VE-cadherin and PECAM-1. Curr Biol. 2013;23(11):1024–30.PubMedPubMedCentralCrossRef
118.
go back to reference Coon BG, Baeyens N, Han J, Budatha M, Ross TD, Fang JS, Yun S, Thomas JL, Schwartz MA. Intramembrane binding of VE-cadherin to VEGFR2 and VEGFR3 assembles the endothelial mechanosensory complex. J Cell Biol. 2015;208(7):975–86.PubMedPubMedCentralCrossRef Coon BG, Baeyens N, Han J, Budatha M, Ross TD, Fang JS, Yun S, Thomas JL, Schwartz MA. Intramembrane binding of VE-cadherin to VEGFR2 and VEGFR3 assembles the endothelial mechanosensory complex. J Cell Biol. 2015;208(7):975–86.PubMedPubMedCentralCrossRef
119.
go back to reference Conway DE, Schwartz MA. Mechanotransduction of shear stress occurs through changes in VE-cadherin and PECAM-1 tension: implications for cell migration. Cell Adhes Migr. 2015;9(5):335–9.CrossRef Conway DE, Schwartz MA. Mechanotransduction of shear stress occurs through changes in VE-cadherin and PECAM-1 tension: implications for cell migration. Cell Adhes Migr. 2015;9(5):335–9.CrossRef
120.
go back to reference Dejana E, Vestweber D. The role of VE-cadherin in vascular morphogenesis and permeability control. Prog Mol Biol Transl Sci. 2013;116:119–44.PubMedCrossRef Dejana E, Vestweber D. The role of VE-cadherin in vascular morphogenesis and permeability control. Prog Mol Biol Transl Sci. 2013;116:119–44.PubMedCrossRef
121.
go back to reference Conway DE, Coon BG, Budatha M, Arsenovic PT, Orsenigo F, Wessel F, Zhang J, Zhuang Z, Dejana E, Vestweber D, et al. VE-cadherin phosphorylation regulates endothelial fluid shear stress responses through the polarity protein LGN. Curr Biol. 2017;27(14):2219–2225.e2215.PubMedPubMedCentralCrossRef Conway DE, Coon BG, Budatha M, Arsenovic PT, Orsenigo F, Wessel F, Zhang J, Zhuang Z, Dejana E, Vestweber D, et al. VE-cadherin phosphorylation regulates endothelial fluid shear stress responses through the polarity protein LGN. Curr Biol. 2017;27(14):2219–2225.e2215.PubMedPubMedCentralCrossRef
122.
go back to reference Sidibé A, Imhof BA. VE-cadherin phosphorylation decides: vascular permeability or diapedesis. Nat Immunol. 2014;15(3):215–7.PubMedCrossRef Sidibé A, Imhof BA. VE-cadherin phosphorylation decides: vascular permeability or diapedesis. Nat Immunol. 2014;15(3):215–7.PubMedCrossRef
123.
go back to reference Kroon J, Heemskerk N, Kalsbeek MJT, de Waard V, van Rijssel J, van Buul JD. Flow-induced endothelial cell alignment requires the RhoGEF Trio as a scaffold protein to polarize active Rac1 distribution. Mol Biol Cell. 2017;28(13):1745–53.PubMedPubMedCentralCrossRef Kroon J, Heemskerk N, Kalsbeek MJT, de Waard V, van Rijssel J, van Buul JD. Flow-induced endothelial cell alignment requires the RhoGEF Trio as a scaffold protein to polarize active Rac1 distribution. Mol Biol Cell. 2017;28(13):1745–53.PubMedPubMedCentralCrossRef
124.
go back to reference Timmerman I, Heemskerk N, Kroon J, Schaefer A, van Rijssel J, Hoogenboezem M, van Unen J, Goedhart J, Gadella TW Jr, Yin T, et al. A local VE-cadherin and Trio-based signaling complex stabilizes endothelial junctions through Rac1. J Cell Sci. 2015;128(16):3041–54.PubMedCrossRef Timmerman I, Heemskerk N, Kroon J, Schaefer A, van Rijssel J, Hoogenboezem M, van Unen J, Goedhart J, Gadella TW Jr, Yin T, et al. A local VE-cadherin and Trio-based signaling complex stabilizes endothelial junctions through Rac1. J Cell Sci. 2015;128(16):3041–54.PubMedCrossRef
125.
go back to reference Rho SS, Ando K, Fukuhara S. Dynamic regulation of vascular permeability by vascular endothelial cadherin-mediated endothelial cell-cell junctions. J Nippon Med School. 2017;84(4):148–59.CrossRef Rho SS, Ando K, Fukuhara S. Dynamic regulation of vascular permeability by vascular endothelial cadherin-mediated endothelial cell-cell junctions. J Nippon Med School. 2017;84(4):148–59.CrossRef
126.
go back to reference Mandyam CD, Villalpando EG, Steiner NL, Quach LW, Fannon MJ, Somkuwar SS. Platelet endothelial cell adhesion molecule-1 and oligodendrogenesis: significance in alcohol use disorders. Brain Sci. 2017;7(10):131.PubMedCentralCrossRef Mandyam CD, Villalpando EG, Steiner NL, Quach LW, Fannon MJ, Somkuwar SS. Platelet endothelial cell adhesion molecule-1 and oligodendrogenesis: significance in alcohol use disorders. Brain Sci. 2017;7(10):131.PubMedCentralCrossRef
127.
go back to reference Feng YM, Chen XH, Zhang X. Roles of PECAM-1 in cell function and disease progression. Eur Rev Med Pharmacol Sci. 2016;20(19):4082–8.PubMed Feng YM, Chen XH, Zhang X. Roles of PECAM-1 in cell function and disease progression. Eur Rev Med Pharmacol Sci. 2016;20(19):4082–8.PubMed
128.
go back to reference Park S, Sorenson CM, Sheibani N. PECAM-1 isoforms, eNOS and endoglin axis in regulation of angiogenesis. Clin Sci. 2015;129(3):217–34.CrossRef Park S, Sorenson CM, Sheibani N. PECAM-1 isoforms, eNOS and endoglin axis in regulation of angiogenesis. Clin Sci. 2015;129(3):217–34.CrossRef
129.
go back to reference Lertkiatmongkol P, Paddock C, Newman DK, Zhu J, Thomas MJ, Newman PJ. The role of sialylated glycans in human platelet endothelial cell adhesion molecule 1 (PECAM-1)-mediated trans homophilic interactions and endothelial cell barrier function. J Biol Chem. 2016;291(50):26216–25.PubMedPubMedCentralCrossRef Lertkiatmongkol P, Paddock C, Newman DK, Zhu J, Thomas MJ, Newman PJ. The role of sialylated glycans in human platelet endothelial cell adhesion molecule 1 (PECAM-1)-mediated trans homophilic interactions and endothelial cell barrier function. J Biol Chem. 2016;291(50):26216–25.PubMedPubMedCentralCrossRef
130.
go back to reference Stevens HY, Melchior B, Bell KS, Yun S, Yeh J-C, Frangos JA. PECAM-1 is a critical mediator of atherosclerosis. Dis Models Mech. 2008;1(2–3):175–81.CrossRef Stevens HY, Melchior B, Bell KS, Yun S, Yeh J-C, Frangos JA. PECAM-1 is a critical mediator of atherosclerosis. Dis Models Mech. 2008;1(2–3):175–81.CrossRef
131.
go back to reference Feaver RE, Gelfand BD, Blackman BR. Human haemodynamic frequency harmonics regulate the inflammatory phenotype of vascular endothelial cells. Nat Commun. 2013;4:1525.PubMedCrossRef Feaver RE, Gelfand BD, Blackman BR. Human haemodynamic frequency harmonics regulate the inflammatory phenotype of vascular endothelial cells. Nat Commun. 2013;4:1525.PubMedCrossRef
132.
go back to reference Chistiakov DA, Orekhov AN, Bobryshev YV. Endothelial PECAM-1 and its function in vascular physiology and atherogenic pathology. Exp Mol Pathol. 2016;100(3):409–15.PubMedCrossRef Chistiakov DA, Orekhov AN, Bobryshev YV. Endothelial PECAM-1 and its function in vascular physiology and atherogenic pathology. Exp Mol Pathol. 2016;100(3):409–15.PubMedCrossRef
133.
go back to reference Di Lorenzo A, Lin MI, Murata T, Landskroner-Eiger S, Schleicher M, Kothiya M, Iwakiri Y, Yu J, Huang PL, Sessa WC. eNOS-derived nitric oxide regulates endothelial barrier function through VE-cadherin and Rho GTPases. J Cell Sci. 2013;126(Pt 24):5541–52.PubMedPubMedCentralCrossRef Di Lorenzo A, Lin MI, Murata T, Landskroner-Eiger S, Schleicher M, Kothiya M, Iwakiri Y, Yu J, Huang PL, Sessa WC. eNOS-derived nitric oxide regulates endothelial barrier function through VE-cadherin and Rho GTPases. J Cell Sci. 2013;126(Pt 24):5541–52.PubMedPubMedCentralCrossRef
134.
go back to reference Zhuang Y, Peng H, Mastej V, Chen W. MicroRNA regulation of endothelial junction proteins and clinical consequence. Mediat Inflamm. 2016;2016:5078627.CrossRef Zhuang Y, Peng H, Mastej V, Chen W. MicroRNA regulation of endothelial junction proteins and clinical consequence. Mediat Inflamm. 2016;2016:5078627.CrossRef
135.
136.
go back to reference Sun X, Belkin N, Feinberg MW. Endothelial microRNAs and atherosclerosis. Curr Atheroscler Rep. 2013;15(12):372.PubMedCrossRef Sun X, Belkin N, Feinberg MW. Endothelial microRNAs and atherosclerosis. Curr Atheroscler Rep. 2013;15(12):372.PubMedCrossRef
137.
go back to reference Kumar S, Kim CW, Simmons RD, Jo H. Role of flow-sensitive microRNAs in endothelial dysfunction and atherosclerosis: mechanosensitive athero-miRs. Arterioscler Thromb Vasc Biol. 2014;34(10):2206–16.PubMedPubMedCentralCrossRef Kumar S, Kim CW, Simmons RD, Jo H. Role of flow-sensitive microRNAs in endothelial dysfunction and atherosclerosis: mechanosensitive athero-miRs. Arterioscler Thromb Vasc Biol. 2014;34(10):2206–16.PubMedPubMedCentralCrossRef
138.
go back to reference Kumar S, Williams D, Sur S, Wang JY, Jo H. Role of flow-sensitive microRNAs and long noncoding RNAs in vascular dysfunction and atherosclerosis. Vasc Pharmacol. 2019;114:76–92.CrossRef Kumar S, Williams D, Sur S, Wang JY, Jo H. Role of flow-sensitive microRNAs and long noncoding RNAs in vascular dysfunction and atherosclerosis. Vasc Pharmacol. 2019;114:76–92.CrossRef
139.
go back to reference Marin T, Gongol B, Chen Z, Woo B, Subramaniam S, Chien S, Shyy JY. Mechanosensitive microRNAs-role in endothelial responses to shear stress and redox state. Free Radic Biol Med. 2013;64:61–8.PubMedPubMedCentralCrossRef Marin T, Gongol B, Chen Z, Woo B, Subramaniam S, Chien S, Shyy JY. Mechanosensitive microRNAs-role in endothelial responses to shear stress and redox state. Free Radic Biol Med. 2013;64:61–8.PubMedPubMedCentralCrossRef
141.
go back to reference Gou L, Zhao L, Song W, Wang L, Liu J, Zhang H, Huang Y, Lau CW, Yao X, Tian XY, et al. Inhibition of miR-92a suppresses oxidative stress and improves endothelial function by upregulating heme oxygenase-1 in db/db mice. Antioxid Redox Signal. 2018;28(5):358–70.PubMedCrossRef Gou L, Zhao L, Song W, Wang L, Liu J, Zhang H, Huang Y, Lau CW, Yao X, Tian XY, et al. Inhibition of miR-92a suppresses oxidative stress and improves endothelial function by upregulating heme oxygenase-1 in db/db mice. Antioxid Redox Signal. 2018;28(5):358–70.PubMedCrossRef
142.
go back to reference Loyer X, Potteaux S, Vion AC, Guerin CL, Boulkroun S, Rautou PE, Ramkhelawon B, Esposito B, Dalloz M, Paul JL, et al. Inhibition of microRNA-92a prevents endothelial dysfunction and atherosclerosis in mice. Circ Res. 2014;114(3):434–43.PubMedCrossRef Loyer X, Potteaux S, Vion AC, Guerin CL, Boulkroun S, Rautou PE, Ramkhelawon B, Esposito B, Dalloz M, Paul JL, et al. Inhibition of microRNA-92a prevents endothelial dysfunction and atherosclerosis in mice. Circ Res. 2014;114(3):434–43.PubMedCrossRef
143.
go back to reference Fang Y, Davies PF. Site-specific microRNA-92a regulation of Kruppel-like factors 4 and 2 in atherosusceptible endothelium. Arterioscler Thromb Vasc Biol. 2012;32(4):979–87.PubMedPubMedCentralCrossRef Fang Y, Davies PF. Site-specific microRNA-92a regulation of Kruppel-like factors 4 and 2 in atherosusceptible endothelium. Arterioscler Thromb Vasc Biol. 2012;32(4):979–87.PubMedPubMedCentralCrossRef
144.
go back to reference Wu W, Xiao H, Laguna-Fernandez A, Villarreal G Jr, Wang KC, Geary GG, Zhang Y, Wang WC, Huang HD, Zhou J, et al. Flow-dependent regulation of Kruppel-like factor 2 is mediated by microRNA-92a. Circulation. 2011;124(5):633–41.PubMedCrossRef Wu W, Xiao H, Laguna-Fernandez A, Villarreal G Jr, Wang KC, Geary GG, Zhang Y, Wang WC, Huang HD, Zhou J, et al. Flow-dependent regulation of Kruppel-like factor 2 is mediated by microRNA-92a. Circulation. 2011;124(5):633–41.PubMedCrossRef
145.
go back to reference Qin X, Wang X, Wang Y, Tang Z, Cui Q, Xi J, Li YS, Chien S, Wang N. MicroRNA-19a mediates the suppressive effect of laminar flow on cyclin D1 expression in human umbilical vein endothelial cells. Proc Natl Acad Sci U S A. 2010;107(7):3240–4.PubMedPubMedCentralCrossRef Qin X, Wang X, Wang Y, Tang Z, Cui Q, Xi J, Li YS, Chien S, Wang N. MicroRNA-19a mediates the suppressive effect of laminar flow on cyclin D1 expression in human umbilical vein endothelial cells. Proc Natl Acad Sci U S A. 2010;107(7):3240–4.PubMedPubMedCentralCrossRef
146.
go back to reference Zhong L, Simard MJ, Huot J. Endothelial microRNAs regulating the NF-kappaB pathway and cell adhesion molecules during inflammation. FASEB J. 2018;32(8):4070–84.PubMedCrossRef Zhong L, Simard MJ, Huot J. Endothelial microRNAs regulating the NF-kappaB pathway and cell adhesion molecules during inflammation. FASEB J. 2018;32(8):4070–84.PubMedCrossRef
147.
go back to reference SenBanerjee S, Lin Z, Atkins GB, Greif DM, Rao RM, Kumar A, Feinberg MW, Chen Z, Simon DI, Luscinskas FW, et al. KLF2 is a novel transcriptional regulator of endothelial proinflammatory activation. J Exp Med. 2004;199(10):1305–15.PubMedPubMedCentralCrossRef SenBanerjee S, Lin Z, Atkins GB, Greif DM, Rao RM, Kumar A, Feinberg MW, Chen Z, Simon DI, Luscinskas FW, et al. KLF2 is a novel transcriptional regulator of endothelial proinflammatory activation. J Exp Med. 2004;199(10):1305–15.PubMedPubMedCentralCrossRef
148.
go back to reference Zhou G, Hamik A, Nayak L, Tian H, Shi H, Lu Y, Sharma N, Liao X, Hale A, Boerboom L, et al. Endothelial Kruppel-like factor 4 protects against atherothrombosis in mice. J Clin Invest. 2012;122(12):4727–31.PubMedPubMedCentralCrossRef Zhou G, Hamik A, Nayak L, Tian H, Shi H, Lu Y, Sharma N, Liao X, Hale A, Boerboom L, et al. Endothelial Kruppel-like factor 4 protects against atherothrombosis in mice. J Clin Invest. 2012;122(12):4727–31.PubMedPubMedCentralCrossRef
149.
go back to reference Taganov KD, Boldin MP, Chang KJ, Baltimore D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci U S A. 2006;103(33):12481–6.PubMedPubMedCentralCrossRef Taganov KD, Boldin MP, Chang KJ, Baltimore D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci U S A. 2006;103(33):12481–6.PubMedPubMedCentralCrossRef
151.
go back to reference Bartoszewski R, Serocki M, Janaszak-Jasiecka A, Bartoszewska S, Kochan-Jamrozy K, Piotrowski A, Króliczewski J, Collawn JF. miR-200b downregulates Kruppel Like Factor 2 (KLF2) during acute hypoxia in human endothelial cells. Eur J Cell Biol. 2017;96(8):758–66.PubMedPubMedCentralCrossRef Bartoszewski R, Serocki M, Janaszak-Jasiecka A, Bartoszewska S, Kochan-Jamrozy K, Piotrowski A, Króliczewski J, Collawn JF. miR-200b downregulates Kruppel Like Factor 2 (KLF2) during acute hypoxia in human endothelial cells. Eur J Cell Biol. 2017;96(8):758–66.PubMedPubMedCentralCrossRef
152.
go back to reference Neth P, Nazari-Jahantigh M, Schober A, Weber C. MicroRNAs in flow-dependent vascular remodelling. Cardiovasc Res. 2013;99(2):294–303.PubMedCrossRef Neth P, Nazari-Jahantigh M, Schober A, Weber C. MicroRNAs in flow-dependent vascular remodelling. Cardiovasc Res. 2013;99(2):294–303.PubMedCrossRef
153.
go back to reference Pan Y, Hui X, Hoo RLC, Ye D, Chan CYC, Feng T, Wang Y, Lam KSL, Xu A. Adipocyte-secreted exosomal microRNA-34a inhibits M2 macrophage polarization to promote obesity-induced adipose inflammation. J Clin Invest. 2019;129(2):834–49.PubMedPubMedCentralCrossRef Pan Y, Hui X, Hoo RLC, Ye D, Chan CYC, Feng T, Wang Y, Lam KSL, Xu A. Adipocyte-secreted exosomal microRNA-34a inhibits M2 macrophage polarization to promote obesity-induced adipose inflammation. J Clin Invest. 2019;129(2):834–49.PubMedPubMedCentralCrossRef
154.
go back to reference Chen Q, Li L, Tu Y, Zheng LL, Liu W, Zuo XY, He YM, Zhang SY, Zhu W, Cao JP, et al. MiR-34a regulates apoptosis in liver cells by targeting the KLF4 gene. Cell Mol Biol Lett. 2014;19(1):52–64.PubMedPubMedCentralCrossRef Chen Q, Li L, Tu Y, Zheng LL, Liu W, Zuo XY, He YM, Zhang SY, Zhu W, Cao JP, et al. MiR-34a regulates apoptosis in liver cells by targeting the KLF4 gene. Cell Mol Biol Lett. 2014;19(1):52–64.PubMedPubMedCentralCrossRef
155.
go back to reference Pircher J, Merkle M, Wörnle M, Ribeiro A, Czermak T, Stampnik Y, Mannell H, Niemeyer M, Vielhauer V, Krötz F. Prothrombotic effects of tumor necrosis factor alpha in vivo are amplified by the absence of TNF-alpha receptor subtype 1 and require TNF-alpha receptor subtype 2. Arthritis Res Ther. 2012;14(5):R225.PubMedPubMedCentralCrossRef Pircher J, Merkle M, Wörnle M, Ribeiro A, Czermak T, Stampnik Y, Mannell H, Niemeyer M, Vielhauer V, Krötz F. Prothrombotic effects of tumor necrosis factor alpha in vivo are amplified by the absence of TNF-alpha receptor subtype 1 and require TNF-alpha receptor subtype 2. Arthritis Res Ther. 2012;14(5):R225.PubMedPubMedCentralCrossRef
156.
go back to reference Pignatelli P, De Biase L, Lenti L, Tocci G, Brunelli A, Cangemi R, Riondino S, Grego S, Volpe M, Violi F. Tumor necrosis factor-alpha as trigger of platelet activation in patients with heart failure. Blood. 2005;106(6):1992–4.PubMedCrossRef Pignatelli P, De Biase L, Lenti L, Tocci G, Brunelli A, Cangemi R, Riondino S, Grego S, Volpe M, Violi F. Tumor necrosis factor-alpha as trigger of platelet activation in patients with heart failure. Blood. 2005;106(6):1992–4.PubMedCrossRef
157.
go back to reference Jang JY, Min JH, Chae YH, Baek JY, Wang SB, Park SJ, Oh GT, Lee S-H, Ho Y-S, Chang T-S. Reactive oxygen species play a critical role in collagen-induced platelet activation via SHP-2 oxidation. Antioxid Redox Signal. 2014;20(16):2528–40.PubMedPubMedCentralCrossRef Jang JY, Min JH, Chae YH, Baek JY, Wang SB, Park SJ, Oh GT, Lee S-H, Ho Y-S, Chang T-S. Reactive oxygen species play a critical role in collagen-induced platelet activation via SHP-2 oxidation. Antioxid Redox Signal. 2014;20(16):2528–40.PubMedPubMedCentralCrossRef
158.
go back to reference Qiao J, Arthur JF, Gardiner EE, Andrews RK, Zeng L, Xu K. Regulation of platelet activation and thrombus formation by reactive oxygen species. Redox Biol. 2018;14:126–30.PubMedCrossRef Qiao J, Arthur JF, Gardiner EE, Andrews RK, Zeng L, Xu K. Regulation of platelet activation and thrombus formation by reactive oxygen species. Redox Biol. 2018;14:126–30.PubMedCrossRef
159.
go back to reference Smith TL, Weyrich AS. Platelets as central mediators of systemic inflammatory responses. Thromb Res. 2011;127(5):391–4.PubMedCrossRef Smith TL, Weyrich AS. Platelets as central mediators of systemic inflammatory responses. Thromb Res. 2011;127(5):391–4.PubMedCrossRef
160.
go back to reference Jenne CN, Urrutia R, Kubes P. Platelets: bridging hemostasis, inflammation, and immunity. Int J Lab Hematol. 2013;35(3):254–61.PubMedCrossRef Jenne CN, Urrutia R, Kubes P. Platelets: bridging hemostasis, inflammation, and immunity. Int J Lab Hematol. 2013;35(3):254–61.PubMedCrossRef
161.
162.
go back to reference Violi F, Pignatelli P, Basili S. Nutrition, supplements, and vitamins in platelet function and bleeding. Circulation. 2010;121(8):1033–44.PubMedCrossRef Violi F, Pignatelli P, Basili S. Nutrition, supplements, and vitamins in platelet function and bleeding. Circulation. 2010;121(8):1033–44.PubMedCrossRef
163.
go back to reference El Haouari M. Platelet oxidative stress and its relationship with cardiovascular diseases in type 2 diabetes mellitus patients. Curr Med Chem. 2019;26(22):4145–65.PubMedCrossRef El Haouari M. Platelet oxidative stress and its relationship with cardiovascular diseases in type 2 diabetes mellitus patients. Curr Med Chem. 2019;26(22):4145–65.PubMedCrossRef
164.
go back to reference Hamilos M, Petousis S, Parthenakis F. Interaction between platelets and endothelium: from pathophysiology to new therapeutic options. Cardiovasc Diagn Ther. 2018;8(5):568–80.PubMedPubMedCentralCrossRef Hamilos M, Petousis S, Parthenakis F. Interaction between platelets and endothelium: from pathophysiology to new therapeutic options. Cardiovasc Diagn Ther. 2018;8(5):568–80.PubMedPubMedCentralCrossRef
165.
go back to reference Celik S, Langer H, Stellos K, May AE, Shankar V, Kurz K, Katus HA, Gawaz MP, Dengler TJ. Platelet-associated LIGHT (TNFSF14) mediates adhesion of platelets to human vascular endothelium. Thromb Haemost. 2007;98(4):798–805.PubMed Celik S, Langer H, Stellos K, May AE, Shankar V, Kurz K, Katus HA, Gawaz MP, Dengler TJ. Platelet-associated LIGHT (TNFSF14) mediates adhesion of platelets to human vascular endothelium. Thromb Haemost. 2007;98(4):798–805.PubMed
166.
go back to reference Celik S, Shankar V, Richter A, Hippe HJ, Akhavanpoor M, Bea F, Erbel C, Urban S, Blank N, Wambsganss N, et al. Proinflammatory and prothrombotic effects on human vascular endothelial cells of immune-cell-derived LIGHT. Eur J Med Res. 2009;14(4):147–56.PubMedPubMedCentralCrossRef Celik S, Shankar V, Richter A, Hippe HJ, Akhavanpoor M, Bea F, Erbel C, Urban S, Blank N, Wambsganss N, et al. Proinflammatory and prothrombotic effects on human vascular endothelial cells of immune-cell-derived LIGHT. Eur J Med Res. 2009;14(4):147–56.PubMedPubMedCentralCrossRef
167.
go back to reference Otterdal K, Smith C, Oie E, Pedersen TM, Yndestad A, Stang E, Endresen K, Solum NO, Aukrust P, Damås JK. Platelet-derived LIGHT induces inflammatory responses in endothelial cells and monocytes. Blood. 2006;108(3):928–35.PubMedCrossRef Otterdal K, Smith C, Oie E, Pedersen TM, Yndestad A, Stang E, Endresen K, Solum NO, Aukrust P, Damås JK. Platelet-derived LIGHT induces inflammatory responses in endothelial cells and monocytes. Blood. 2006;108(3):928–35.PubMedCrossRef
168.
go back to reference Gerdes N, Seijkens T, Lievens D, Kuijpers MJE, Winkels H, Projahn D, Hartwig H, Beckers L, Megens RTA, Boon L, et al. Platelet CD40 exacerbates atherosclerosis by transcellular activation of endothelial cells and leukocytes. Arterioscler Thromb Vasc Biol. 2016;36(3):482–90.PubMedCrossRef Gerdes N, Seijkens T, Lievens D, Kuijpers MJE, Winkels H, Projahn D, Hartwig H, Beckers L, Megens RTA, Boon L, et al. Platelet CD40 exacerbates atherosclerosis by transcellular activation of endothelial cells and leukocytes. Arterioscler Thromb Vasc Biol. 2016;36(3):482–90.PubMedCrossRef
169.
go back to reference Michel NA, Zirlik A, Wolf D: CD40L and its receptors in atherothrombosis—an update. Front Cardiovas Med 2017, 4(40). Michel NA, Zirlik A, Wolf D: CD40L and its receptors in atherothrombosis—an update. Front Cardiovas Med 2017, 4(40).
170.
go back to reference Heiss EH, Schachner D, Zimmermann K, Dirsch VM. Glucose availability is a decisive factor for Nrf2-mediated gene expression. Redox Biol. 2013;1(1):359–65.PubMedPubMedCentralCrossRef Heiss EH, Schachner D, Zimmermann K, Dirsch VM. Glucose availability is a decisive factor for Nrf2-mediated gene expression. Redox Biol. 2013;1(1):359–65.PubMedPubMedCentralCrossRef
171.
go back to reference Deshauer C, Morgan AM, Ryan EO, Handel TM, Prestegard JH, Wang X. Interactions of the chemokine CCL5/RANTES with medium-sized chondroitin sulfate ligands. Structure. 2015;23(6):1066–77.PubMedPubMedCentralCrossRef Deshauer C, Morgan AM, Ryan EO, Handel TM, Prestegard JH, Wang X. Interactions of the chemokine CCL5/RANTES with medium-sized chondroitin sulfate ligands. Structure. 2015;23(6):1066–77.PubMedPubMedCentralCrossRef
172.
go back to reference von Hundelshausen P, Schmitt MMN: Platelets and their chemokines in atherosclerosis—clinical applications. Front Physiol 2014, 5(294). von Hundelshausen P, Schmitt MMN: Platelets and their chemokines in atherosclerosis—clinical applications. Front Physiol 2014, 5(294).
174.
go back to reference Øynebråten I, Barois N, Bergeland T, Küchler AM, Bakke O, Haraldsen G. Oligomerized, filamentous surface presentation of RANTES/CCL5 on vascular endothelial cells. Sci Rep. 2015;5(1):9261.PubMedPubMedCentralCrossRef Øynebråten I, Barois N, Bergeland T, Küchler AM, Bakke O, Haraldsen G. Oligomerized, filamentous surface presentation of RANTES/CCL5 on vascular endothelial cells. Sci Rep. 2015;5(1):9261.PubMedPubMedCentralCrossRef
176.
177.
go back to reference Carnevale R, Bartimoccia S, Nocella C, Di Santo S, Loffredo L, Illuminati G, Lombardi E, Boz V, Del Ben M, De Marco L, et al. LDL oxidation by platelets propagates platelet activation via an oxidative stress-mediated mechanism. Atherosclerosis. 2014;237(1):108–16.PubMedCrossRef Carnevale R, Bartimoccia S, Nocella C, Di Santo S, Loffredo L, Illuminati G, Lombardi E, Boz V, Del Ben M, De Marco L, et al. LDL oxidation by platelets propagates platelet activation via an oxidative stress-mediated mechanism. Atherosclerosis. 2014;237(1):108–16.PubMedCrossRef
178.
go back to reference Carnevale R, Pignatelli P, Lenti L, Buchetti B, Sanguigni V, Di Santo S, Violi F. LDL are oxidatively modified by platelets via GP91(phox) and accumulate in human monocytes. FASEB J. 2007;21(3):927–34.PubMedCrossRef Carnevale R, Pignatelli P, Lenti L, Buchetti B, Sanguigni V, Di Santo S, Violi F. LDL are oxidatively modified by platelets via GP91(phox) and accumulate in human monocytes. FASEB J. 2007;21(3):927–34.PubMedCrossRef
179.
go back to reference Sachais BS, Kuo A, Nassar T, Morgan J, Kariko K, Williams KJ, Feldman M, Aviram M, Shah N, Jarett L, et al. Platelet factor 4 binds to low-density lipoprotein receptors and disrupts the endocytic machinery, resulting in retention of low-density lipoprotein on the cell surface. Blood. 2002;99(10):3613–22.PubMedCrossRef Sachais BS, Kuo A, Nassar T, Morgan J, Kariko K, Williams KJ, Feldman M, Aviram M, Shah N, Jarett L, et al. Platelet factor 4 binds to low-density lipoprotein receptors and disrupts the endocytic machinery, resulting in retention of low-density lipoprotein on the cell surface. Blood. 2002;99(10):3613–22.PubMedCrossRef
180.
go back to reference Nassar T, Sachais BS, Se A, Kowalska MA, Bdeir K, Leitersdorf E, Hiss E, Ziporen L, Aviram M, Cines D, et al. Platelet factor 4 enhances the binding of oxidized low-density lipoprotein to vascular wall cells. J Biol Chem. 2003;278(8):6187–93.PubMedCrossRef Nassar T, Sachais BS, Se A, Kowalska MA, Bdeir K, Leitersdorf E, Hiss E, Ziporen L, Aviram M, Cines D, et al. Platelet factor 4 enhances the binding of oxidized low-density lipoprotein to vascular wall cells. J Biol Chem. 2003;278(8):6187–93.PubMedCrossRef
181.
go back to reference Li X, Meng X, Gao X, Pang X, Wang Y, Wu X, Deng X, Zhang Q, Sun C, Li Y. Elevated serum xanthine oxidase activity is associated with the development of type 2 diabetes: a prospective cohort study. Diabetes Care. 2018;41(4):884–90.PubMed Li X, Meng X, Gao X, Pang X, Wang Y, Wu X, Deng X, Zhang Q, Sun C, Li Y. Elevated serum xanthine oxidase activity is associated with the development of type 2 diabetes: a prospective cohort study. Diabetes Care. 2018;41(4):884–90.PubMed
182.
go back to reference Battelli MG, Bortolotti M, Polito L, Bolognesi A. The role of xanthine oxidoreductase and uric acid in metabolic syndrome. Biochim Biophys Acta Mol basis Dis. 2018;1864(8):2557–65.PubMedCrossRef Battelli MG, Bortolotti M, Polito L, Bolognesi A. The role of xanthine oxidoreductase and uric acid in metabolic syndrome. Biochim Biophys Acta Mol basis Dis. 2018;1864(8):2557–65.PubMedCrossRef
183.
go back to reference Battelli MG, Bolognesi A, Polito L. Pathophysiology of circulating xanthine oxidoreductase: new emerging roles for a multi-tasking enzyme. Biochim Biophys Acta (BBA) - Mol Basis Dis. 2014;1842(9):1502–17.CrossRef Battelli MG, Bolognesi A, Polito L. Pathophysiology of circulating xanthine oxidoreductase: new emerging roles for a multi-tasking enzyme. Biochim Biophys Acta (BBA) - Mol Basis Dis. 2014;1842(9):1502–17.CrossRef
184.
go back to reference Schuchardt M, Herrmann J, Tolle M, van der Giet M. Xanthine oxidase and its role as target in cardiovascular disease: cardiovascular protection by enzyme inhibition? Curr Pharm Des. 2017;23(23):3391–404.PubMedCrossRef Schuchardt M, Herrmann J, Tolle M, van der Giet M. Xanthine oxidase and its role as target in cardiovascular disease: cardiovascular protection by enzyme inhibition? Curr Pharm Des. 2017;23(23):3391–404.PubMedCrossRef
185.
go back to reference Alem MM. Allopurinol and endothelial function: a systematic review with meta-analysis of randomized controlled trials. Cardiovasc Ther. 2018;36(4):e12432.PubMedCrossRef Alem MM. Allopurinol and endothelial function: a systematic review with meta-analysis of randomized controlled trials. Cardiovasc Ther. 2018;36(4):e12432.PubMedCrossRef
186.
go back to reference Xin W, Mi S, Lin Z. Allopurinol therapy improves vascular endothelial function in subjects at risk for cardiovascular diseases: a meta-analysis of randomized controlled trials. Cardiovasc Ther. 2016;34(6):441–9.PubMedCrossRef Xin W, Mi S, Lin Z. Allopurinol therapy improves vascular endothelial function in subjects at risk for cardiovascular diseases: a meta-analysis of randomized controlled trials. Cardiovasc Ther. 2016;34(6):441–9.PubMedCrossRef
187.
go back to reference Cicero AFG, Pirro M, Watts GF, Mikhailidis DP, Banach M, Sahebkar A. Effects of allopurinol on endothelial function: a systematic review and meta-analysis of randomized placebo-controlled trials. Drugs. 2018;78(1):99–109.PubMedCrossRef Cicero AFG, Pirro M, Watts GF, Mikhailidis DP, Banach M, Sahebkar A. Effects of allopurinol on endothelial function: a systematic review and meta-analysis of randomized placebo-controlled trials. Drugs. 2018;78(1):99–109.PubMedCrossRef
188.
go back to reference Bredemeier M, Lopes LM, Eisenreich MA, Hickmann S, Bongiorno GK, d’Avila R, Morsch ALB, da Silva Stein F, Campos GGD. Xanthine oxidase inhibitors for prevention of cardiovascular events: a systematic review and meta-analysis of randomized controlled trials. BMC Cardiovasc Disord. 2018;18(1):24.PubMedPubMedCentralCrossRef Bredemeier M, Lopes LM, Eisenreich MA, Hickmann S, Bongiorno GK, d’Avila R, Morsch ALB, da Silva Stein F, Campos GGD. Xanthine oxidase inhibitors for prevention of cardiovascular events: a systematic review and meta-analysis of randomized controlled trials. BMC Cardiovasc Disord. 2018;18(1):24.PubMedPubMedCentralCrossRef
189.
go back to reference Munzel T, Camici GG, Maack C, Bonetti NR, Fuster V, Kovacic JC. Impact of oxidative stress on the heart and vasculature: part 2 of a 3-part series. J Am Coll Cardiol. 2017;70(2):212–29.PubMedPubMedCentralCrossRef Munzel T, Camici GG, Maack C, Bonetti NR, Fuster V, Kovacic JC. Impact of oxidative stress on the heart and vasculature: part 2 of a 3-part series. J Am Coll Cardiol. 2017;70(2):212–29.PubMedPubMedCentralCrossRef
190.
go back to reference George J. Role of urate, xanthine oxidase and the effects of allopurinol in vascular oxidative stress. Vasc Health Risk Manag. 2009:265. George J. Role of urate, xanthine oxidase and the effects of allopurinol in vascular oxidative stress. Vasc Health Risk Manag. 2009:265.
191.
go back to reference Gao X, Zhang H, Belmadani S, Wu J, Xu X, Elford H, Potter BJ, Zhang C. Role of TNF-alpha-induced reactive oxygen species in endothelial dysfunction during reperfusion injury. Am J Phys Heart Circ Phys. 2008;295(6):H2242–9. Gao X, Zhang H, Belmadani S, Wu J, Xu X, Elford H, Potter BJ, Zhang C. Role of TNF-alpha-induced reactive oxygen species in endothelial dysfunction during reperfusion injury. Am J Phys Heart Circ Phys. 2008;295(6):H2242–9.
192.
go back to reference Zhang C, Hein TW, Wang W, Ren Y, Shipley RD, Kuo L. Activation of JNK and xanthine oxidase by TNF-alpha impairs nitric oxide-mediated dilation of coronary arterioles. J Mol Cell Cardiol. 2006;40(2):247–57.PubMedCrossRef Zhang C, Hein TW, Wang W, Ren Y, Shipley RD, Kuo L. Activation of JNK and xanthine oxidase by TNF-alpha impairs nitric oxide-mediated dilation of coronary arterioles. J Mol Cell Cardiol. 2006;40(2):247–57.PubMedCrossRef
193.
go back to reference Ogura J, Kuwayama K, Sasaki S, Kaneko C, Koizumi T, Yabe K, Tsujimoto T, Takeno R, Takaya A, Kobayashi M, et al. Reactive oxygen species derived from xanthine oxidase interrupt dimerization of breast cancer resistance protein, resulting in suppression of uric acid excretion to the intestinal lumen. Biochem Pharmacol. 2015;97(1):89–98.PubMedCrossRef Ogura J, Kuwayama K, Sasaki S, Kaneko C, Koizumi T, Yabe K, Tsujimoto T, Takeno R, Takaya A, Kobayashi M, et al. Reactive oxygen species derived from xanthine oxidase interrupt dimerization of breast cancer resistance protein, resulting in suppression of uric acid excretion to the intestinal lumen. Biochem Pharmacol. 2015;97(1):89–98.PubMedCrossRef
194.
go back to reference Ives A, Nomura J, Martinon F, Roger T, LeRoy D, Miner JN, Simon G, Busso N, So A. Xanthine oxidoreductase regulates macrophage IL1β secretion upon NLRP3 inflammasome activation. Nat Commun. 2015;6(1):6555.PubMedCrossRef Ives A, Nomura J, Martinon F, Roger T, LeRoy D, Miner JN, Simon G, Busso N, So A. Xanthine oxidoreductase regulates macrophage IL1β secretion upon NLRP3 inflammasome activation. Nat Commun. 2015;6(1):6555.PubMedCrossRef
195.
go back to reference Maruhashi T, Hisatome I, Kihara Y, Higashi Y. Hyperuricemia and endothelial function: from molecular background to clinical perspectives. Atherosclerosis. 2018;278:226–31.PubMedCrossRef Maruhashi T, Hisatome I, Kihara Y, Higashi Y. Hyperuricemia and endothelial function: from molecular background to clinical perspectives. Atherosclerosis. 2018;278:226–31.PubMedCrossRef
196.
go back to reference Desco M-C, Asensi M, Márquez R, Martínez-Valls J, Vento M, Pallardó FV, Sastre J, Viña J. Xanthine oxidase is involved in free radical production in type 1 diabetes. Protect Allopurinol. 2002;51(4):1118–24. Desco M-C, Asensi M, Márquez R, Martínez-Valls J, Vento M, Pallardó FV, Sastre J, Viña J. Xanthine oxidase is involved in free radical production in type 1 diabetes. Protect Allopurinol. 2002;51(4):1118–24.
197.
go back to reference Landmesser U, Spiekermann S, Preuss C, Sorrentino S, Fischer D, Manes C, Mueller M, Drexler H. Angiotensin II induces endothelial xanthine oxidase activation. Arterioscler Thromb Vasc Biol. 2007;27(4):943–8.PubMedCrossRef Landmesser U, Spiekermann S, Preuss C, Sorrentino S, Fischer D, Manes C, Mueller M, Drexler H. Angiotensin II induces endothelial xanthine oxidase activation. Arterioscler Thromb Vasc Biol. 2007;27(4):943–8.PubMedCrossRef
198.
go back to reference Houston M, Estevez A, Chumley P, Aslan M, Marklund S, Parks DA, Freeman BA. Binding of xanthine oxidase to vascular endothelium: kinetic characterization and oxidative impairment of nitric oxide-dependent signaling. J Biol Chem. 1999;274(8):4985–94.PubMedCrossRef Houston M, Estevez A, Chumley P, Aslan M, Marklund S, Parks DA, Freeman BA. Binding of xanthine oxidase to vascular endothelium: kinetic characterization and oxidative impairment of nitric oxide-dependent signaling. J Biol Chem. 1999;274(8):4985–94.PubMedCrossRef
199.
go back to reference Tomiyama H, Higashi Y, Takase B, Node K, Sata M, Inoue T, Ishibashi Y, Ueda S, Shimada K, Yamashina A. Relationships among hyperuricemia, metabolic syndrome, and endothelial function. Am J Hypertens. 2011;24(7):770–4.PubMedCrossRef Tomiyama H, Higashi Y, Takase B, Node K, Sata M, Inoue T, Ishibashi Y, Ueda S, Shimada K, Yamashina A. Relationships among hyperuricemia, metabolic syndrome, and endothelial function. Am J Hypertens. 2011;24(7):770–4.PubMedCrossRef
200.
go back to reference Cicero AFG, Rosticci M, Parini A, Baronio C, D'Addato S, Borghi C. Serum uric acid is inversely proportional to estimated stroke volume and cardiac output in a large sample of pharmacologically untreated subjects: data from the Brisighella Heart Study. Intern Emerg Med. 2014;9(6):655–60.PubMedCrossRef Cicero AFG, Rosticci M, Parini A, Baronio C, D'Addato S, Borghi C. Serum uric acid is inversely proportional to estimated stroke volume and cardiac output in a large sample of pharmacologically untreated subjects: data from the Brisighella Heart Study. Intern Emerg Med. 2014;9(6):655–60.PubMedCrossRef
201.
go back to reference Volterrani M, Iellamo F, Sposato B, Romeo F. Uric acid lowering therapy in cardiovascular diseases. Int J Cardiol. 2016;213:20–2.PubMedCrossRef Volterrani M, Iellamo F, Sposato B, Romeo F. Uric acid lowering therapy in cardiovascular diseases. Int J Cardiol. 2016;213:20–2.PubMedCrossRef
202.
204.
go back to reference Rahimi-Sakak F, Maroofi M, Rahmani J, Bellissimo N, Hekmatdoost A. Serum uric acid and risk of cardiovascular mortality: a systematic review and dose-response meta-analysis of cohort studies of over a million participants. BMC Cardiovasc Disord. 2019;19(1):218.PubMedPubMedCentralCrossRef Rahimi-Sakak F, Maroofi M, Rahmani J, Bellissimo N, Hekmatdoost A. Serum uric acid and risk of cardiovascular mortality: a systematic review and dose-response meta-analysis of cohort studies of over a million participants. BMC Cardiovasc Disord. 2019;19(1):218.PubMedPubMedCentralCrossRef
205.
go back to reference Chang C-C, Wu C-H, Liu L-K, Chou R-H, Kuo C-S, Huang P-H, Chen L-K, Lin S-J. Association between serum uric acid and cardiovascular risk in nonhypertensive and nondiabetic individuals: the Taiwan I-Lan Longitudinal Aging Study. Sci Rep. 2018;8(1):5234.PubMedPubMedCentralCrossRef Chang C-C, Wu C-H, Liu L-K, Chou R-H, Kuo C-S, Huang P-H, Chen L-K, Lin S-J. Association between serum uric acid and cardiovascular risk in nonhypertensive and nondiabetic individuals: the Taiwan I-Lan Longitudinal Aging Study. Sci Rep. 2018;8(1):5234.PubMedPubMedCentralCrossRef
206.
go back to reference Kang DH, Han L, Ouyang X, Kahn AM, Kanellis J, Li P, Feng L, Nakagawa T, Watanabe S, Hosoyamada M, et al. Uric acid causes vascular smooth muscle cell proliferation by entering cells via a functional urate transporter. Am J Nephrol. 2005;25(5):425–33.PubMedCrossRef Kang DH, Han L, Ouyang X, Kahn AM, Kanellis J, Li P, Feng L, Nakagawa T, Watanabe S, Hosoyamada M, et al. Uric acid causes vascular smooth muscle cell proliferation by entering cells via a functional urate transporter. Am J Nephrol. 2005;25(5):425–33.PubMedCrossRef
207.
go back to reference Price KL, Sautin YY, Long DA. Human vascular smooth muscle cells express a urate transporter. J Am Soc Nephrol. 2006;17:1791–5.PubMedCrossRef Price KL, Sautin YY, Long DA. Human vascular smooth muscle cells express a urate transporter. J Am Soc Nephrol. 2006;17:1791–5.PubMedCrossRef
208.
go back to reference Liu S, Yuan Y, Zhou Y, Zhao M, Chen Y, Cheng J, Lu Y, Liu J. Phloretin attenuates hyperuricemia-induced endothelial dysfunction through co-inhibiting inflammation and GLUT9-mediated uric acid uptake. J Cell Mol Med. 2017;21(10):2553–62.PubMedPubMedCentralCrossRef Liu S, Yuan Y, Zhou Y, Zhao M, Chen Y, Cheng J, Lu Y, Liu J. Phloretin attenuates hyperuricemia-induced endothelial dysfunction through co-inhibiting inflammation and GLUT9-mediated uric acid uptake. J Cell Mol Med. 2017;21(10):2553–62.PubMedPubMedCentralCrossRef
211.
go back to reference Cai W, Duan X-M, Liu Y, Yu J, Tang Y-L, Liu Z-L, Jiang S, Zhang C-P, Liu J-Y, Xu J-X. Uric acid induces endothelial dysfunction by activating the HMGB1/RAGE signaling pathway. Biomed Res Int. 2017;2017:4391920.PubMedPubMedCentral Cai W, Duan X-M, Liu Y, Yu J, Tang Y-L, Liu Z-L, Jiang S, Zhang C-P, Liu J-Y, Xu J-X. Uric acid induces endothelial dysfunction by activating the HMGB1/RAGE signaling pathway. Biomed Res Int. 2017;2017:4391920.PubMedPubMedCentral
212.
go back to reference Yang X, Gu J, Lv H, Li H, Cheng Y, Liu Y, Jiang Y. Uric acid induced inflammatory responses in endothelial cells via up-regulating(pro)renin receptor. Biomed Pharmacother. 2019;109:1163–70.PubMedCrossRef Yang X, Gu J, Lv H, Li H, Cheng Y, Liu Y, Jiang Y. Uric acid induced inflammatory responses in endothelial cells via up-regulating(pro)renin receptor. Biomed Pharmacother. 2019;109:1163–70.PubMedCrossRef
213.
go back to reference Liang WY, Zhu XY, Zhang JW, Feng XR, Wang YC, Liu ML. Uric acid promotes chemokine and adhesion molecule production in vascular endothelium via nuclear factor-kappa B signaling. Nutr Metabol Cardiovasc Dis. 2015;25(2):187–94.CrossRef Liang WY, Zhu XY, Zhang JW, Feng XR, Wang YC, Liu ML. Uric acid promotes chemokine and adhesion molecule production in vascular endothelium via nuclear factor-kappa B signaling. Nutr Metabol Cardiovasc Dis. 2015;25(2):187–94.CrossRef
214.
go back to reference Rabadi MM, Kuo M-C, Ghaly T, Rabadi SM, Weber M, Goligorsky MS, Ratliff BB. Interaction between uric acid and HMGB1 translocation and release from endothelial cells. Am J Physiol Renal Physiol. 2012;302(6):F730–41.PubMedCrossRef Rabadi MM, Kuo M-C, Ghaly T, Rabadi SM, Weber M, Goligorsky MS, Ratliff BB. Interaction between uric acid and HMGB1 translocation and release from endothelial cells. Am J Physiol Renal Physiol. 2012;302(6):F730–41.PubMedCrossRef
215.
go back to reference Braga TT, Forni MF, Correa-Costa M, Ramos RN, Barbuto JA, Branco P, Castoldi A, Hiyane MI, Davanso MR, Latz E, et al. Soluble uric acid activates the NLRP3 inflammasome. Sci Rep. 2017;7(1):39884.PubMedPubMedCentralCrossRef Braga TT, Forni MF, Correa-Costa M, Ramos RN, Barbuto JA, Branco P, Castoldi A, Hiyane MI, Davanso MR, Latz E, et al. Soluble uric acid activates the NLRP3 inflammasome. Sci Rep. 2017;7(1):39884.PubMedPubMedCentralCrossRef
216.
go back to reference Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature. 2003;425(6957):516–21.PubMedCrossRef Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature. 2003;425(6957):516–21.PubMedCrossRef
217.
go back to reference Park J-H, Jin YM, Hwang S, Cho D-H, Kang D-H, Jo I. Uric acid attenuates nitric oxide production by decreasing the interaction between endothelial nitric oxide synthase and calmodulin in human umbilical vein endothelial cells: a mechanism for uric acid-induced cardiovascular disease development. Nitric oxide. 2013;32:36–42.PubMedCrossRef Park J-H, Jin YM, Hwang S, Cho D-H, Kang D-H, Jo I. Uric acid attenuates nitric oxide production by decreasing the interaction between endothelial nitric oxide synthase and calmodulin in human umbilical vein endothelial cells: a mechanism for uric acid-induced cardiovascular disease development. Nitric oxide. 2013;32:36–42.PubMedCrossRef
218.
go back to reference Verzola D, Ratto E, Villaggio B, Parodi EL, Pontremoli R, Garibotto G, Viazzi F. Uric acid promotes apoptosis in human proximal tubule cells by oxidative stress and the activation of NADPH oxidase NOX 4. PLoS One. 2014;9(12):e115210.PubMedPubMedCentralCrossRef Verzola D, Ratto E, Villaggio B, Parodi EL, Pontremoli R, Garibotto G, Viazzi F. Uric acid promotes apoptosis in human proximal tubule cells by oxidative stress and the activation of NADPH oxidase NOX 4. PLoS One. 2014;9(12):e115210.PubMedPubMedCentralCrossRef
219.
go back to reference K-s L, Z-h P, Cheng W-j, C-f D, Tong H. Endoplasmic reticulum stress-induced apoptosis in the development of reproduction. J Reprod Contracept. 2016;27(1):51–9. K-s L, Z-h P, Cheng W-j, C-f D, Tong H. Endoplasmic reticulum stress-induced apoptosis in the development of reproduction. J Reprod Contracept. 2016;27(1):51–9.
220.
go back to reference Komori H, Yamada K, Tamai I. Hyperuricemia enhances intracellular urate accumulation via down-regulation of cell-surface BCRP/ABCG2 expression in vascular endothelial cells. Biochim Biophys Acta Biomembr. 2018;1860(5):973–80.PubMedCrossRef Komori H, Yamada K, Tamai I. Hyperuricemia enhances intracellular urate accumulation via down-regulation of cell-surface BCRP/ABCG2 expression in vascular endothelial cells. Biochim Biophys Acta Biomembr. 2018;1860(5):973–80.PubMedCrossRef
221.
go back to reference Kong J, Chalcraft K, Mandur TS, Jimenez-Saiz R, Walker TD, Goncharova S, Gordon ME, Naji L, Flader K, Larche M, et al. Comprehensive metabolomics identifies the alarmin uric acid as a critical signal for the induction of peanut allergy. Allergy. 2015;70(5):495–505.PubMedCrossRef Kong J, Chalcraft K, Mandur TS, Jimenez-Saiz R, Walker TD, Goncharova S, Gordon ME, Naji L, Flader K, Larche M, et al. Comprehensive metabolomics identifies the alarmin uric acid as a critical signal for the induction of peanut allergy. Allergy. 2015;70(5):495–505.PubMedCrossRef
222.
go back to reference Kanda A, Ishida S. (Pro)renin receptor: involvement in diabetic retinopathy and development of molecular targeted therapy. J Diab Investig. 2019;10(1):6–17.CrossRef Kanda A, Ishida S. (Pro)renin receptor: involvement in diabetic retinopathy and development of molecular targeted therapy. J Diab Investig. 2019;10(1):6–17.CrossRef
223.
go back to reference Yu M-A, Sánchez-Lozada LG, Johnson RJ, Kang D-H. Oxidative stress with an activation of the renin–angiotensin system in human vascular endothelial cells as a novel mechanism of uric acid-induced endothelial dysfunction. J Hypertens. 2010;28(6):1234–42.PubMed Yu M-A, Sánchez-Lozada LG, Johnson RJ, Kang D-H. Oxidative stress with an activation of the renin–angiotensin system in human vascular endothelial cells as a novel mechanism of uric acid-induced endothelial dysfunction. J Hypertens. 2010;28(6):1234–42.PubMed
224.
go back to reference Corry DB, Eslami P, Yamamoto K, Nyby MD, Makino H, Tuck ML. Uric acid stimulates vascular smooth muscle cell proliferation and oxidative stress via the vascular renin–angiotensin system. J Hypertens. 2008;26(2):269–75.PubMedCrossRef Corry DB, Eslami P, Yamamoto K, Nyby MD, Makino H, Tuck ML. Uric acid stimulates vascular smooth muscle cell proliferation and oxidative stress via the vascular renin–angiotensin system. J Hypertens. 2008;26(2):269–75.PubMedCrossRef
225.
go back to reference Uraoka M, Ikeda K, Nakagawa Y, Koide M, Akakabe Y, Nakano-Kurimoto R, Takahashi T, Matoba S, Yamada H, Okigaki M, et al. Prorenin induces ERK activation in endothelial cells to enhance neovascularization independently of the renin-angiotensin system. Biochem Biophys Res Commun. 2009;390(4):1202–7.PubMedCrossRef Uraoka M, Ikeda K, Nakagawa Y, Koide M, Akakabe Y, Nakano-Kurimoto R, Takahashi T, Matoba S, Yamada H, Okigaki M, et al. Prorenin induces ERK activation in endothelial cells to enhance neovascularization independently of the renin-angiotensin system. Biochem Biophys Res Commun. 2009;390(4):1202–7.PubMedCrossRef
226.
go back to reference Ho E, Karimi Galougahi K, Liu C-C, Bhindi R, Figtree GA. Biological markers of oxidative stress: applications to cardiovascular research and practice. Redox Biol. 2013;1(1):483–91.PubMedPubMedCentralCrossRef Ho E, Karimi Galougahi K, Liu C-C, Bhindi R, Figtree GA. Biological markers of oxidative stress: applications to cardiovascular research and practice. Redox Biol. 2013;1(1):483–91.PubMedPubMedCentralCrossRef
227.
go back to reference Khan AA, Alsahli MA, Rahmani AH. Myeloperoxidase as an active disease biomarker: recent biochemical and pathological perspectives. Med Sci (Basel). 2018;6(2):33. Khan AA, Alsahli MA, Rahmani AH. Myeloperoxidase as an active disease biomarker: recent biochemical and pathological perspectives. Med Sci (Basel). 2018;6(2):33.
228.
go back to reference Aratani Y. Myeloperoxidase: its role for host defense, inflammation, and neutrophil function. Arch Biochem Biophys. 2018;640:47–52.PubMedCrossRef Aratani Y. Myeloperoxidase: its role for host defense, inflammation, and neutrophil function. Arch Biochem Biophys. 2018;640:47–52.PubMedCrossRef
229.
go back to reference Odobasic D, Kitching AR, Holdsworth SR. Neutrophil-mediated regulation of innate and adaptive immunity: the role of myeloperoxidase. J Immunol Res. 2016;2016:2349817.PubMedPubMedCentralCrossRef Odobasic D, Kitching AR, Holdsworth SR. Neutrophil-mediated regulation of innate and adaptive immunity: the role of myeloperoxidase. J Immunol Res. 2016;2016:2349817.PubMedPubMedCentralCrossRef
230.
go back to reference Delporte C, Van Antwerpen P, Vanhamme L, Roumeguere T, Zouaoui Boudjeltia K. Low-density lipoprotein modified by myeloperoxidase in inflammatory pathways and clinical studies. Mediat Inflamm. 2013;2013:971579.CrossRef Delporte C, Van Antwerpen P, Vanhamme L, Roumeguere T, Zouaoui Boudjeltia K. Low-density lipoprotein modified by myeloperoxidase in inflammatory pathways and clinical studies. Mediat Inflamm. 2013;2013:971579.CrossRef
231.
go back to reference Roumeguere T, Zouaoui Boudjeltia K, Babar S, Nuyens V, Rousseau A, Van Antwerpen P, Ducobu J, Wespes E, Vanhaeverbeek M. Effects of phosphodiesterase inhibitors on the inflammatory response of endothelial cells stimulated by myeloperoxidase-modified low-density lipoprotein or tumor necrosis factor alpha. Eur Urol. 2010;57(3):522–8.PubMedCrossRef Roumeguere T, Zouaoui Boudjeltia K, Babar S, Nuyens V, Rousseau A, Van Antwerpen P, Ducobu J, Wespes E, Vanhaeverbeek M. Effects of phosphodiesterase inhibitors on the inflammatory response of endothelial cells stimulated by myeloperoxidase-modified low-density lipoprotein or tumor necrosis factor alpha. Eur Urol. 2010;57(3):522–8.PubMedCrossRef
232.
go back to reference Boudjeltia KZ, Legssyer I, Van Antwerpen P, Kisoka RL, Babar S, Moguilevsky N, Delree P, Ducobu J, Remacle C, Vanhaeverbeek M, et al. Triggering of inflammatory response by myeloperoxidase-oxidized LDL. Biochem Cell Biol. 2006;84(5):805–12.PubMedCrossRef Boudjeltia KZ, Legssyer I, Van Antwerpen P, Kisoka RL, Babar S, Moguilevsky N, Delree P, Ducobu J, Remacle C, Vanhaeverbeek M, et al. Triggering of inflammatory response by myeloperoxidase-oxidized LDL. Biochem Cell Biol. 2006;84(5):805–12.PubMedCrossRef
233.
go back to reference Sokolov AV, Kostevich VA, Runova OL, Gorudko IV, Vasilyev VB, Cherenkevich SN, Panasenko OM. Proatherogenic modification of LDL by surface-bound myeloperoxidase. Chem Phys Lipids. 2014;180:72–80.PubMedCrossRef Sokolov AV, Kostevich VA, Runova OL, Gorudko IV, Vasilyev VB, Cherenkevich SN, Panasenko OM. Proatherogenic modification of LDL by surface-bound myeloperoxidase. Chem Phys Lipids. 2014;180:72–80.PubMedCrossRef
234.
go back to reference Wang G, Qian P, Jackson FR, Qian G, Wu G. Sequential activation of JAKs, STATs and xanthine dehydrogenase/oxidase by hypoxia in lung microvascular endothelial cells. Int J Biochem Cell Biol. 2008;40(3):461–70.PubMedCrossRef Wang G, Qian P, Jackson FR, Qian G, Wu G. Sequential activation of JAKs, STATs and xanthine dehydrogenase/oxidase by hypoxia in lung microvascular endothelial cells. Int J Biochem Cell Biol. 2008;40(3):461–70.PubMedCrossRef
235.
go back to reference Jerke U, Rolle S, Purfürst B, Luft FC, Nauseef WM, Kettritz R. β2 integrin-mediated cell-cell contact transfers active myeloperoxidase from neutrophils to endothelial cells. J Biol Chem. 2013;288(18):12910–9.PubMedPubMedCentralCrossRef Jerke U, Rolle S, Purfürst B, Luft FC, Nauseef WM, Kettritz R. β2 integrin-mediated cell-cell contact transfers active myeloperoxidase from neutrophils to endothelial cells. J Biol Chem. 2013;288(18):12910–9.PubMedPubMedCentralCrossRef
236.
go back to reference Yiginer O, Ozcelik F, Inanc T, Aparci M, Ozmen N, Cingozbay BY, Kardesoglu E, Suleymanoglu S, Sener G, Cebeci BS. Allopurinol improves endothelial function and reduces oxidant-inflammatory enzyme of myeloperoxidase in metabolic syndrome. Clin Res Cardiol. 2008;97(5):334–40.PubMedCrossRef Yiginer O, Ozcelik F, Inanc T, Aparci M, Ozmen N, Cingozbay BY, Kardesoglu E, Suleymanoglu S, Sener G, Cebeci BS. Allopurinol improves endothelial function and reduces oxidant-inflammatory enzyme of myeloperoxidase in metabolic syndrome. Clin Res Cardiol. 2008;97(5):334–40.PubMedCrossRef
237.
go back to reference Stamp LK, Turner R, Khalilova IS, Zhang M, Drake J, Forbes LV, Kettle AJ. Myeloperoxidase and oxidation of uric acid in gout: implications for the clinical consequences of hyperuricaemia. Rheumatology (Oxford). 2014;53(11):1958–65.CrossRef Stamp LK, Turner R, Khalilova IS, Zhang M, Drake J, Forbes LV, Kettle AJ. Myeloperoxidase and oxidation of uric acid in gout: implications for the clinical consequences of hyperuricaemia. Rheumatology (Oxford). 2014;53(11):1958–65.CrossRef
238.
go back to reference Astern JM, Pendergraft WF 3rd, Falk RJ, Jennette JC, Schmaier AH, Mahdi F, Preston GA. Myeloperoxidase interacts with endothelial cell-surface cytokeratin 1 and modulates bradykinin production by the plasma Kallikrein-Kinin system. Am J Pathol. 2007;171(1):349–60.PubMedPubMedCentralCrossRef Astern JM, Pendergraft WF 3rd, Falk RJ, Jennette JC, Schmaier AH, Mahdi F, Preston GA. Myeloperoxidase interacts with endothelial cell-surface cytokeratin 1 and modulates bradykinin production by the plasma Kallikrein-Kinin system. Am J Pathol. 2007;171(1):349–60.PubMedPubMedCentralCrossRef
239.
go back to reference Baldus S, Heitzer T, Eiserich JP, Lau D, Mollnau H, Ortak M, Petri S, Goldmann B, Duchstein H-J, Berger J, et al. Myeloperoxidase enhances nitric oxide catabolism during myocardial ischemia and reperfusion. Free Radic Biol Med. 2004;37(6):902–11.PubMedCrossRef Baldus S, Heitzer T, Eiserich JP, Lau D, Mollnau H, Ortak M, Petri S, Goldmann B, Duchstein H-J, Berger J, et al. Myeloperoxidase enhances nitric oxide catabolism during myocardial ischemia and reperfusion. Free Radic Biol Med. 2004;37(6):902–11.PubMedCrossRef
240.
go back to reference Eiserich JP, Baldus S, Brennan M-L, Ma W, Zhang C, Tousson A, Castro L, Lusis AJ, Nauseef WM, White CR, et al. Myeloperoxidase, a leukocyte-derived vascular NO oxidase. Sci (New York). 2002;296(5577):2391–4.CrossRef Eiserich JP, Baldus S, Brennan M-L, Ma W, Zhang C, Tousson A, Castro L, Lusis AJ, Nauseef WM, White CR, et al. Myeloperoxidase, a leukocyte-derived vascular NO oxidase. Sci (New York). 2002;296(5577):2391–4.CrossRef
241.
go back to reference Maiocchi S, Dang L, Morris J, Thomas S, Rees M. PP28 - inhibition of myeloperoxidase-mediated endothelial dysfunction by nitroxides. Free Radic Biol Med. 2015;86:S28–9.CrossRef Maiocchi S, Dang L, Morris J, Thomas S, Rees M. PP28 - inhibition of myeloperoxidase-mediated endothelial dysfunction by nitroxides. Free Radic Biol Med. 2015;86:S28–9.CrossRef
242.
go back to reference Bienert GP, Moller AL, Kristiansen KA, Schulz A, Moller IM, Schjoerring JK, Jahn TP. Specific aquaporins facilitate the diffusion of hydrogen peroxide across membranes. J Biol Chem. 2007;282(2):1183–92.PubMedCrossRef Bienert GP, Moller AL, Kristiansen KA, Schulz A, Moller IM, Schjoerring JK, Jahn TP. Specific aquaporins facilitate the diffusion of hydrogen peroxide across membranes. J Biol Chem. 2007;282(2):1183–92.PubMedCrossRef
243.
go back to reference Al Ghouleh I, Frazziano G, Rodriguez AI, Csányi G, Maniar S, St Croix CM, Kelley EE, Egaña LA, Song GJ, Bisello A, et al. Aquaporin 1, Nox1, and Ask1 mediate oxidant-induced smooth muscle cell hypertrophy. Cardiovasc Res. 2013;97(1):134–42.PubMedCrossRef Al Ghouleh I, Frazziano G, Rodriguez AI, Csányi G, Maniar S, St Croix CM, Kelley EE, Egaña LA, Song GJ, Bisello A, et al. Aquaporin 1, Nox1, and Ask1 mediate oxidant-induced smooth muscle cell hypertrophy. Cardiovasc Res. 2013;97(1):134–42.PubMedCrossRef
244.
245.
246.
go back to reference Oliveira-Marques V, Marinho HS, Cyrne L, Antunes F. Role of hydrogen peroxide in NF-kappaB activation: from inducer to modulator. Antioxid Redox Signal. 2009;11(9):2223–43.PubMedCrossRef Oliveira-Marques V, Marinho HS, Cyrne L, Antunes F. Role of hydrogen peroxide in NF-kappaB activation: from inducer to modulator. Antioxid Redox Signal. 2009;11(9):2223–43.PubMedCrossRef
247.
go back to reference Brandes RP, Weissmann N, Schroder K. Nox family NADPH oxidases: molecular mechanisms of activation. Free Radic Biol Med. 2014;76:208–26.PubMedCrossRef Brandes RP, Weissmann N, Schroder K. Nox family NADPH oxidases: molecular mechanisms of activation. Free Radic Biol Med. 2014;76:208–26.PubMedCrossRef
248.
go back to reference Chen K, Kirber MT, Xiao H, Yang Y, Keaney JF Jr. Regulation of ROS signal transduction by NADPH oxidase 4 localization. J Cell Biol. 2008;181(7):1129–39.PubMedPubMedCentralCrossRef Chen K, Kirber MT, Xiao H, Yang Y, Keaney JF Jr. Regulation of ROS signal transduction by NADPH oxidase 4 localization. J Cell Biol. 2008;181(7):1129–39.PubMedPubMedCentralCrossRef
250.
go back to reference Daiber A, Di Lisa F, Oelze M, Kroller-Schon S, Steven S, Schulz E, Munzel T. Crosstalk of mitochondria with NADPH oxidase via reactive oxygen and nitrogen species signalling and its role for vascular function. Br J Pharmacol. 2017;174(12):1670–89.PubMedCrossRef Daiber A, Di Lisa F, Oelze M, Kroller-Schon S, Steven S, Schulz E, Munzel T. Crosstalk of mitochondria with NADPH oxidase via reactive oxygen and nitrogen species signalling and its role for vascular function. Br J Pharmacol. 2017;174(12):1670–89.PubMedCrossRef
251.
go back to reference Sartoretto JL, Kalwa H, Pluth MD, Lippard SJ, Michel T. Hydrogen peroxide differentially modulates cardiac myocyte nitric oxide synthesis. Proc Natl Acad Sci. 2011;108(38):15792–7.PubMedCrossRefPubMedCentral Sartoretto JL, Kalwa H, Pluth MD, Lippard SJ, Michel T. Hydrogen peroxide differentially modulates cardiac myocyte nitric oxide synthesis. Proc Natl Acad Sci. 2011;108(38):15792–7.PubMedCrossRefPubMedCentral
252.
go back to reference Silva BR, Pernomian L, Grando MD, Bendhack LM. Phenylephrine activates eNOS Ser1177 phosphorylation and nitric oxide signaling in renal hypertensive rat aorta. Eur J Pharmacol. 2014;738:192–9.PubMedCrossRef Silva BR, Pernomian L, Grando MD, Bendhack LM. Phenylephrine activates eNOS Ser1177 phosphorylation and nitric oxide signaling in renal hypertensive rat aorta. Eur J Pharmacol. 2014;738:192–9.PubMedCrossRef
253.
go back to reference Koju N, Taleb A, Zhou J, Lv G, Yang J, Cao X, Lei H, Ding Q. Pharmacological strategies to lower crosstalk between nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondria. Biomed Pharmacother. 2019;111:1478–98.PubMedCrossRef Koju N, Taleb A, Zhou J, Lv G, Yang J, Cao X, Lei H, Ding Q. Pharmacological strategies to lower crosstalk between nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondria. Biomed Pharmacother. 2019;111:1478–98.PubMedCrossRef
254.
go back to reference Lee DY, Wauquier F, Eid AA, Roman LJ, Ghosh-Choudhury G, Khazim K, Block K, Gorin Y. Nox4 NADPH oxidase mediates peroxynitrite-dependent uncoupling of endothelial nitric-oxide synthase and fibronectin expression in response to angiotensin II: role of mitochondrial reactive oxygen species. J Biol Chem. 2013;288(40):28668–86.PubMedPubMedCentralCrossRef Lee DY, Wauquier F, Eid AA, Roman LJ, Ghosh-Choudhury G, Khazim K, Block K, Gorin Y. Nox4 NADPH oxidase mediates peroxynitrite-dependent uncoupling of endothelial nitric-oxide synthase and fibronectin expression in response to angiotensin II: role of mitochondrial reactive oxygen species. J Biol Chem. 2013;288(40):28668–86.PubMedPubMedCentralCrossRef
255.
go back to reference Förstermann U, Xia N, Li H. Roles of vascular oxidative stress and nitric oxide in the pathogenesis of atherosclerosis. Circ Res. 2017;120(4):713–35.PubMedCrossRef Förstermann U, Xia N, Li H. Roles of vascular oxidative stress and nitric oxide in the pathogenesis of atherosclerosis. Circ Res. 2017;120(4):713–35.PubMedCrossRef
256.
go back to reference Förstermann U, Sessa WC. Nitric oxide synthases: regulation and function. Eur Heart J. 2012;33(7):829–37 837a-837d.PubMedCrossRef Förstermann U, Sessa WC. Nitric oxide synthases: regulation and function. Eur Heart J. 2012;33(7):829–37 837a-837d.PubMedCrossRef
258.
go back to reference Bendall JK, Douglas G, McNeill E, Channon KM, Crabtree MJ. Tetrahydrobiopterin in cardiovascular health and disease. Antioxid Redox Signal. 2014;20(18):3040–77.PubMedPubMedCentralCrossRef Bendall JK, Douglas G, McNeill E, Channon KM, Crabtree MJ. Tetrahydrobiopterin in cardiovascular health and disease. Antioxid Redox Signal. 2014;20(18):3040–77.PubMedPubMedCentralCrossRef
259.
go back to reference Meza CA, La Favor JD, Kim DH, Hickner RC. Endothelial dysfunction: is there a hyperglycemia-induced imbalance of NOX and NOS? Int J Mol Sci. 2019;20(15). Meza CA, La Favor JD, Kim DH, Hickner RC. Endothelial dysfunction: is there a hyperglycemia-induced imbalance of NOX and NOS? Int J Mol Sci. 2019;20(15).
260.
go back to reference Gielis JF, Quirynen L, Briedé JJ, Roelant E, Cos P, Van Schil PEY. Pathogenetic role of endothelial nitric oxide synthase uncoupling during lung ischaemia–reperfusion injury†. Eur J Cardiothorac Surg. 2017;52(2):256–63.PubMedCrossRef Gielis JF, Quirynen L, Briedé JJ, Roelant E, Cos P, Van Schil PEY. Pathogenetic role of endothelial nitric oxide synthase uncoupling during lung ischaemia–reperfusion injury†. Eur J Cardiothorac Surg. 2017;52(2):256–63.PubMedCrossRef
261.
go back to reference Forstermann U, Munzel T. Endothelial nitric oxide synthase in vascular disease: from marvel to menace. Circulation. 2006;113(13):1708–14.PubMedCrossRef Forstermann U, Munzel T. Endothelial nitric oxide synthase in vascular disease: from marvel to menace. Circulation. 2006;113(13):1708–14.PubMedCrossRef
262.
go back to reference Oberhuber R, Riede G, Cardini B, Bernhard D, Messner B, Watschinger K, Steger C, Brandacher G, Pratschke J, Golderer G, et al. Impaired endothelial nitric oxide synthase homodimer formation triggers development of transplant vasculopathy - insights from a murine aortic transplantation model. Sci Rep. 2016;6(1):37917.PubMedPubMedCentralCrossRef Oberhuber R, Riede G, Cardini B, Bernhard D, Messner B, Watschinger K, Steger C, Brandacher G, Pratschke J, Golderer G, et al. Impaired endothelial nitric oxide synthase homodimer formation triggers development of transplant vasculopathy - insights from a murine aortic transplantation model. Sci Rep. 2016;6(1):37917.PubMedPubMedCentralCrossRef
263.
go back to reference Daiber A, Xia N, Steven S, Oelze M, Hanf A, Kröller-Schön S, Münzel T, Li H. New therapeutic implications of endothelial nitric oxide synthase (eNOS) function/dysfunction in cardiovascular disease. Int J Mol Sci. 2019;20(1):187.PubMedCentralCrossRef Daiber A, Xia N, Steven S, Oelze M, Hanf A, Kröller-Schön S, Münzel T, Li H. New therapeutic implications of endothelial nitric oxide synthase (eNOS) function/dysfunction in cardiovascular disease. Int J Mol Sci. 2019;20(1):187.PubMedCentralCrossRef
264.
go back to reference Li H, Forstermann U. Uncoupling of endothelial NO synthase in atherosclerosis and vascular disease. Curr Opin Pharmacol. 2013;13(2):161–7.PubMedCrossRef Li H, Forstermann U. Uncoupling of endothelial NO synthase in atherosclerosis and vascular disease. Curr Opin Pharmacol. 2013;13(2):161–7.PubMedCrossRef
265.
go back to reference Vasileiou PVS, Evangelou K, Vlasis K, Fildisis G, Panayiotidis MI, Chronopoulos E, Passias P-G, Kouloukoussa M, Gorgoulis VG, Havaki S. Mitochondrial homeostasis and cellular senescence. Cells. 2019;8(7):686.PubMedCentralCrossRef Vasileiou PVS, Evangelou K, Vlasis K, Fildisis G, Panayiotidis MI, Chronopoulos E, Passias P-G, Kouloukoussa M, Gorgoulis VG, Havaki S. Mitochondrial homeostasis and cellular senescence. Cells. 2019;8(7):686.PubMedCentralCrossRef
266.
go back to reference Correia-Melo C, Passos JF. Mitochondria: are they causal players in cellular senescence? Biochim Biophys Acta. 2015;1847(11):1373–9.PubMedCrossRef Correia-Melo C, Passos JF. Mitochondria: are they causal players in cellular senescence? Biochim Biophys Acta. 2015;1847(11):1373–9.PubMedCrossRef
267.
go back to reference Minamino T, Miyauchi H, Yoshida T, Ishida Y, Yoshida H, Komuro I. Endothelial cell senescence in human atherosclerosis: role of telomere in endothelial dysfunction. Circulation. 2002;105(13):1541–4.PubMedCrossRef Minamino T, Miyauchi H, Yoshida T, Ishida Y, Yoshida H, Komuro I. Endothelial cell senescence in human atherosclerosis: role of telomere in endothelial dysfunction. Circulation. 2002;105(13):1541–4.PubMedCrossRef
268.
go back to reference Childs BG, Baker DJ, Wijshake T, Conover CA, Campisi J, van Deursen JM. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science. 2016;354(6311):472–7.PubMedPubMedCentralCrossRef Childs BG, Baker DJ, Wijshake T, Conover CA, Campisi J, van Deursen JM. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science. 2016;354(6311):472–7.PubMedPubMedCentralCrossRef
269.
go back to reference Siasos G, Tsigkou V, Kosmopoulos M, Theodosiadis D, Simantiris S, Tagkou NM, Tsimpiktsioglou A, Stampouloglou PK, Oikonomou E, Mourouzis K, et al. Mitochondria and cardiovascular diseases-from pathophysiology to treatment. Ann Transl Med. 2018;6(12):256.PubMedPubMedCentralCrossRef Siasos G, Tsigkou V, Kosmopoulos M, Theodosiadis D, Simantiris S, Tagkou NM, Tsimpiktsioglou A, Stampouloglou PK, Oikonomou E, Mourouzis K, et al. Mitochondria and cardiovascular diseases-from pathophysiology to treatment. Ann Transl Med. 2018;6(12):256.PubMedPubMedCentralCrossRef
270.
go back to reference Wang Y, Tabas I. Emerging roles of mitochondria ROS in atherosclerotic lesions: causation or association? J Atheroscler Thromb. 2014;21(5):381–90.PubMedCrossRef Wang Y, Tabas I. Emerging roles of mitochondria ROS in atherosclerotic lesions: causation or association? J Atheroscler Thromb. 2014;21(5):381–90.PubMedCrossRef
271.
go back to reference Dorighello GG, Paim BA, Kiihl SF, Ferreira M, Catharino RR, Vercesi AE, Oliveira HCF. Correlation between mitochondrial reactive oxygen and severity of atherosclerosis. Oxidative Med Cell Longev. 2016;2016:10.CrossRef Dorighello GG, Paim BA, Kiihl SF, Ferreira M, Catharino RR, Vercesi AE, Oliveira HCF. Correlation between mitochondrial reactive oxygen and severity of atherosclerosis. Oxidative Med Cell Longev. 2016;2016:10.CrossRef
272.
go back to reference Liu R, Liu H, Ha Y, Tilton RG, Zhang W. Oxidative stress induces endothelial cell senescence via downregulation of Sirt6. Biomed Res Int. 2014;2014:13. Liu R, Liu H, Ha Y, Tilton RG, Zhang W. Oxidative stress induces endothelial cell senescence via downregulation of Sirt6. Biomed Res Int. 2014;2014:13.
273.
go back to reference Cid-Castro C, Hernández-Espinosa DR, Morán J. ROS as regulators of mitochondrial dynamics in neurons. Cell Mol Neurobiol. 2018;38(5):995–1007.PubMedCrossRef Cid-Castro C, Hernández-Espinosa DR, Morán J. ROS as regulators of mitochondrial dynamics in neurons. Cell Mol Neurobiol. 2018;38(5):995–1007.PubMedCrossRef
274.
go back to reference Ježek J, Cooper KF, Strich R. Reactive oxygen species and mitochondrial dynamics: the yin and yang of mitochondrial dysfunction and cancer progression. Antioxidants (Basel). 2018;7(1):13.CrossRef Ježek J, Cooper KF, Strich R. Reactive oxygen species and mitochondrial dynamics: the yin and yang of mitochondrial dysfunction and cancer progression. Antioxidants (Basel). 2018;7(1):13.CrossRef
275.
go back to reference Kim B, Song YS. Mitochondrial dynamics altered by oxidative stress in cancer. Free Radic Res. 2016;50(10):1065–70.PubMedCrossRef Kim B, Song YS. Mitochondrial dynamics altered by oxidative stress in cancer. Free Radic Res. 2016;50(10):1065–70.PubMedCrossRef
276.
go back to reference Hung CH-L, Cheng SS-Y, Cheung Y-T, Wuwongse S, Zhang NQ, Ho Y-S, Lee SM-Y, Chang RC-C. A reciprocal relationship between reactive oxygen species and mitochondrial dynamics in neurodegeneration. Redox Biol. 2018;14:7–19.PubMedCrossRef Hung CH-L, Cheng SS-Y, Cheung Y-T, Wuwongse S, Zhang NQ, Ho Y-S, Lee SM-Y, Chang RC-C. A reciprocal relationship between reactive oxygen species and mitochondrial dynamics in neurodegeneration. Redox Biol. 2018;14:7–19.PubMedCrossRef
278.
go back to reference Quintero M, Colombo SL, Godfrey A, Moncada S. Mitochondria as signaling organelles in the vascular endothelium. Proc Natl Acad Sci U S A. 2006;103(14):5379–84.PubMedPubMedCentralCrossRef Quintero M, Colombo SL, Godfrey A, Moncada S. Mitochondria as signaling organelles in the vascular endothelium. Proc Natl Acad Sci U S A. 2006;103(14):5379–84.PubMedPubMedCentralCrossRef
279.
go back to reference Shenouda SM, Widlansky ME, Chen K, Xu G, Holbrook M, Tabit CE, Hamburg NM, Frame AA, Caiano TL, Kluge MA, et al. Altered mitochondrial dynamics contributes to endothelial dysfunction in diabetes mellitus. Circulation. 2011;124(4):444–53.PubMedPubMedCentralCrossRef Shenouda SM, Widlansky ME, Chen K, Xu G, Holbrook M, Tabit CE, Hamburg NM, Frame AA, Caiano TL, Kluge MA, et al. Altered mitochondrial dynamics contributes to endothelial dysfunction in diabetes mellitus. Circulation. 2011;124(4):444–53.PubMedPubMedCentralCrossRef
280.
go back to reference Ong SB, Subrayan S, Lim SY, Yellon DM, Davidson SM, Hausenloy DJ. Inhibiting mitochondrial fission protects the heart against ischemia/reperfusion injury. Circulation. 2010;121(18):2012–22.PubMedCrossRef Ong SB, Subrayan S, Lim SY, Yellon DM, Davidson SM, Hausenloy DJ. Inhibiting mitochondrial fission protects the heart against ischemia/reperfusion injury. Circulation. 2010;121(18):2012–22.PubMedCrossRef
281.
go back to reference Borodkina AV, Shatrova AN, Deryabin PI, Griukova AA, Abushik PA, Antonov SM, Nikolsky NN, Burova EB. Calcium alterations signal either to senescence or to autophagy induction in stem cells upon oxidative stress. Aging. 2016;8(12):3400–18.PubMedPubMedCentralCrossRef Borodkina AV, Shatrova AN, Deryabin PI, Griukova AA, Abushik PA, Antonov SM, Nikolsky NN, Burova EB. Calcium alterations signal either to senescence or to autophagy induction in stem cells upon oxidative stress. Aging. 2016;8(12):3400–18.PubMedPubMedCentralCrossRef
282.
go back to reference Ziegler DV, Wiley CD, Velarde MC. Mitochondrial effectors of cellular senescence: beyond the free radical theory of aging. Aging Cell. 2015;14(1):1–7.PubMedCrossRef Ziegler DV, Wiley CD, Velarde MC. Mitochondrial effectors of cellular senescence: beyond the free radical theory of aging. Aging Cell. 2015;14(1):1–7.PubMedCrossRef
283.
go back to reference Hom JR, Gewandter JS, Michael L, Sheu SS, Yoon Y. Thapsigargin induces biphasic fragmentation of mitochondria through calcium-mediated mitochondrial fission and apoptosis. J Cell Physiol. 2007;212(2):498–508.PubMedCrossRef Hom JR, Gewandter JS, Michael L, Sheu SS, Yoon Y. Thapsigargin induces biphasic fragmentation of mitochondria through calcium-mediated mitochondrial fission and apoptosis. J Cell Physiol. 2007;212(2):498–508.PubMedCrossRef
284.
285.
go back to reference Li L, Pan R, Li R, Niemann B, Aurich A-C, Chen Y, Rohrbach S. Mitochondrial biogenesis and peroxisome proliferator–activated receptor-γ coactivator-1α (PGC-1α) deacetylation by physical activity. Intact Adipocytokine Signal Required. 2011;60(1):157–67. Li L, Pan R, Li R, Niemann B, Aurich A-C, Chen Y, Rohrbach S. Mitochondrial biogenesis and peroxisome proliferator–activated receptor-γ coactivator-1α (PGC-1α) deacetylation by physical activity. Intact Adipocytokine Signal Required. 2011;60(1):157–67.
286.
go back to reference Kauppinen A, Suuronen T, Ojala J, Kaarniranta K, Salminen A. Antagonistic crosstalk between NF-kappaB and SIRT1 in the regulation of inflammation and metabolic disorders. Cell Signal. 2013;25(10):1939–48.PubMedCrossRef Kauppinen A, Suuronen T, Ojala J, Kaarniranta K, Salminen A. Antagonistic crosstalk between NF-kappaB and SIRT1 in the regulation of inflammation and metabolic disorders. Cell Signal. 2013;25(10):1939–48.PubMedCrossRef
287.
go back to reference Salminen A, Kaarniranta K, Kauppinen A. Crosstalk between oxidative stress and SIRT1: impact on the aging process. Int J Mol Sci. 2013;14(2):3834–59.PubMedPubMedCentralCrossRef Salminen A, Kaarniranta K, Kauppinen A. Crosstalk between oxidative stress and SIRT1: impact on the aging process. Int J Mol Sci. 2013;14(2):3834–59.PubMedPubMedCentralCrossRef
288.
go back to reference Kida Y, Goligorsky MS. Sirtuins, cell senescence, and vascular aging. Can J Cardiol. 2016;32(5):634–41.PubMedCrossRef Kida Y, Goligorsky MS. Sirtuins, cell senescence, and vascular aging. Can J Cardiol. 2016;32(5):634–41.PubMedCrossRef
289.
go back to reference Morris G, Puri BK, Walker AJ, Berk M, Walder K, Bortolasci CC, Marx W, Carvalho AF, Maes M: The compensatory antioxidant response system with a focus on neuroprogressive disorders. Prog Neuro-Psychopharmacol Biol Psychiatry 2019;95:109708. Morris G, Puri BK, Walker AJ, Berk M, Walder K, Bortolasci CC, Marx W, Carvalho AF, Maes M: The compensatory antioxidant response system with a focus on neuroprogressive disorders. Prog Neuro-Psychopharmacol Biol Psychiatry 2019;95:109708.
290.
go back to reference Morris G, Maes M, Berk M, Puri BK. Myalgic encephalomyelitis or chronic fatigue syndrome: how could the illness develop? Metab Brain Dis. 2019;34(2):385–415.PubMedPubMedCentralCrossRef Morris G, Maes M, Berk M, Puri BK. Myalgic encephalomyelitis or chronic fatigue syndrome: how could the illness develop? Metab Brain Dis. 2019;34(2):385–415.PubMedPubMedCentralCrossRef
291.
go back to reference Wang M, Li G, Yang Z, Wang L, Zhang L, Wang T, Zhang Y, Zhang S, Han Y, Jia L. Uncoupling protein 2 downregulation by hypoxia through repression of peroxisome proliferator-activated receptor γ promotes chemoresistance of non-small cell lung cancer. Oncotarget. 2017;8(5):8083–94.PubMedCrossRef Wang M, Li G, Yang Z, Wang L, Zhang L, Wang T, Zhang Y, Zhang S, Han Y, Jia L. Uncoupling protein 2 downregulation by hypoxia through repression of peroxisome proliferator-activated receptor γ promotes chemoresistance of non-small cell lung cancer. Oncotarget. 2017;8(5):8083–94.PubMedCrossRef
292.
go back to reference Corona JC, Duchen MR. PPARγ and PGC-1α as therapeutic targets in Parkinson’s. Neurochem Res. 2015;40(2):308–16.PubMedCrossRef Corona JC, Duchen MR. PPARγ and PGC-1α as therapeutic targets in Parkinson’s. Neurochem Res. 2015;40(2):308–16.PubMedCrossRef
293.
go back to reference Shimasaki Y, Pan N, Messina LM, Li C, Chen K, Liu L, Cooper MP, Vita JA, Keaney JF Jr. Uncoupling protein 2 impacts endothelial phenotype via p53-mediated control of mitochondrial dynamics. Circ Res. 2013;113(7):891–901.PubMedCrossRef Shimasaki Y, Pan N, Messina LM, Li C, Chen K, Liu L, Cooper MP, Vita JA, Keaney JF Jr. Uncoupling protein 2 impacts endothelial phenotype via p53-mediated control of mitochondrial dynamics. Circ Res. 2013;113(7):891–901.PubMedCrossRef
294.
go back to reference Koziel A, Sobieraj I, Jarmuszkiewicz W. Increased activity of mitochondrial uncoupling protein 2 improves stress resistance in cultured endothelial cells exposed in vitro to high glucose levels. Am J Phys Heart Circ Phys. 2015;309(1):H147–56. Koziel A, Sobieraj I, Jarmuszkiewicz W. Increased activity of mitochondrial uncoupling protein 2 improves stress resistance in cultured endothelial cells exposed in vitro to high glucose levels. Am J Phys Heart Circ Phys. 2015;309(1):H147–56.
295.
go back to reference Sun J, Pu Y, Wang P, Chen S, Zhao Y, Liu C, Shang Q, Zhu Z, Liu D. TRPV1-mediated UCP2 upregulation ameliorates hyperglycemia-induced endothelial dysfunction. Cardiovasc Diabetol. 2013;12:69.PubMedPubMedCentralCrossRef Sun J, Pu Y, Wang P, Chen S, Zhao Y, Liu C, Shang Q, Zhu Z, Liu D. TRPV1-mediated UCP2 upregulation ameliorates hyperglycemia-induced endothelial dysfunction. Cardiovasc Diabetol. 2013;12:69.PubMedPubMedCentralCrossRef
296.
go back to reference Ganss R. Keeping the balance right: regulator of G protein signaling 5 in vascular physiology and pathology. Prog Mol Biol Transl Sci. 2015;133:93–121.PubMedCrossRef Ganss R. Keeping the balance right: regulator of G protein signaling 5 in vascular physiology and pathology. Prog Mol Biol Transl Sci. 2015;133:93–121.PubMedCrossRef
298.
go back to reference Cai H. Hydrogen peroxide regulation of endothelial function: origins, mechanisms, and consequences. Cardiovasc Res. 2005;68(1):26–36.PubMedCrossRef Cai H. Hydrogen peroxide regulation of endothelial function: origins, mechanisms, and consequences. Cardiovasc Res. 2005;68(1):26–36.PubMedCrossRef
299.
go back to reference Enesa K, Ito K, Luong LA, Thorbjornsen I, Phua C, To Y, Dean J, Haskard DO, Boyle J, Adcock I, et al. Hydrogen peroxide prolongs nuclear localization of NF-κB in activated cells by suppressing negative regulatory mechanisms. J Biol Chem. 2008;283(27):18582–90.PubMedCrossRef Enesa K, Ito K, Luong LA, Thorbjornsen I, Phua C, To Y, Dean J, Haskard DO, Boyle J, Adcock I, et al. Hydrogen peroxide prolongs nuclear localization of NF-κB in activated cells by suppressing negative regulatory mechanisms. J Biol Chem. 2008;283(27):18582–90.PubMedCrossRef
301.
go back to reference van Bergen LA, Roos G, De Proft F. From thiol to sulfonic acid: modeling the oxidation pathway of protein thiols by hydrogen peroxide. J Phys Chem A. 2014;118(31):6078–84.PubMedCrossRef van Bergen LA, Roos G, De Proft F. From thiol to sulfonic acid: modeling the oxidation pathway of protein thiols by hydrogen peroxide. J Phys Chem A. 2014;118(31):6078–84.PubMedCrossRef
302.
go back to reference Poole LB. The basics of thiols and cysteines in redox biology and chemistry. Free Radic Biol Med. 2015;80:148–57.PubMedCrossRef Poole LB. The basics of thiols and cysteines in redox biology and chemistry. Free Radic Biol Med. 2015;80:148–57.PubMedCrossRef
303.
go back to reference Fang J, Holmgren A. Inhibition of thioredoxin and thioredoxin reductase by 4-hydroxy-2-nonenal in vitro and in vivo. J Am Chem Soc. 2006;128(6):1879–85.PubMedCrossRef Fang J, Holmgren A. Inhibition of thioredoxin and thioredoxin reductase by 4-hydroxy-2-nonenal in vitro and in vivo. J Am Chem Soc. 2006;128(6):1879–85.PubMedCrossRef
304.
go back to reference García-Nogales P, Almeida A, Bolaños JP. Peroxynitrite protects neurons against nitric oxide-mediated apoptosis: a key role for glucose-6-phosphate dehydrogenase activity in neuroprotection. J Biol Chem. 2003;278(2):864–74.PubMedCrossRef García-Nogales P, Almeida A, Bolaños JP. Peroxynitrite protects neurons against nitric oxide-mediated apoptosis: a key role for glucose-6-phosphate dehydrogenase activity in neuroprotection. J Biol Chem. 2003;278(2):864–74.PubMedCrossRef
305.
go back to reference Chauvin JR, Pratt DA. On the reactions of thiols, sulfenic acids, and sulfinic acids with hydrogen peroxide. Angew Chem Inte Ed Engl. 2017;56(22):6255–9.CrossRef Chauvin JR, Pratt DA. On the reactions of thiols, sulfenic acids, and sulfinic acids with hydrogen peroxide. Angew Chem Inte Ed Engl. 2017;56(22):6255–9.CrossRef
306.
go back to reference Cyrne L, Oliveira-Marques V, Marinho HS, Antunes F. H2O2 in the induction of NF-kappaB-dependent selective gene expression. Methods Enzymol. 2013;528:173–88.PubMedCrossRef Cyrne L, Oliveira-Marques V, Marinho HS, Antunes F. H2O2 in the induction of NF-kappaB-dependent selective gene expression. Methods Enzymol. 2013;528:173–88.PubMedCrossRef
307.
go back to reference Tilstra JS, Robinson AR, Wang J, Gregg SQ, Clauson CL, Reay DP, Nasto LA, St Croix CM, Usas A, Vo N, et al. NF-κB inhibition delays DNA damage-induced senescence and aging in mice. J Clin Invest. 2012;122(7):2601–12.PubMedPubMedCentralCrossRef Tilstra JS, Robinson AR, Wang J, Gregg SQ, Clauson CL, Reay DP, Nasto LA, St Croix CM, Usas A, Vo N, et al. NF-κB inhibition delays DNA damage-induced senescence and aging in mice. J Clin Invest. 2012;122(7):2601–12.PubMedPubMedCentralCrossRef
309.
go back to reference Salminen A, Kauppinen A, Kaarniranta K. Emerging role of NF-κB signaling in the induction of senescence-associated secretory phenotype (SASP). Cell Signal. 2012;24(4):835–45.PubMedCrossRef Salminen A, Kauppinen A, Kaarniranta K. Emerging role of NF-κB signaling in the induction of senescence-associated secretory phenotype (SASP). Cell Signal. 2012;24(4):835–45.PubMedCrossRef
310.
311.
312.
go back to reference Johnson RF, Perkins ND. Nuclear factor-kappaB, p53, and mitochondria: regulation of cellular metabolism and the Warburg effect. Trends Biochem Sci. 2012;37(8):317–24.PubMedCrossRef Johnson RF, Perkins ND. Nuclear factor-kappaB, p53, and mitochondria: regulation of cellular metabolism and the Warburg effect. Trends Biochem Sci. 2012;37(8):317–24.PubMedCrossRef
313.
go back to reference Londhe P, Yu PY, Ijiri Y, Ladner KJ, Fenger JM, London C, Houghton PJ, Guttridge DC: Classical NF-κB metabolically reprograms sarcoma cells through regulation of hexokinase 2. Front Oncol 2018, 8(104). Londhe P, Yu PY, Ijiri Y, Ladner KJ, Fenger JM, London C, Houghton PJ, Guttridge DC: Classical NF-κB metabolically reprograms sarcoma cells through regulation of hexokinase 2. Front Oncol 2018, 8(104).
314.
go back to reference Tornatore L, Thotakura AK, Bennett J, Moretti M, Franzoso G. The nuclear factor kappa B signaling pathway: integrating metabolism with inflammation. Trends Cell Biol. 2012;22(11):557–66.PubMedCrossRef Tornatore L, Thotakura AK, Bennett J, Moretti M, Franzoso G. The nuclear factor kappa B signaling pathway: integrating metabolism with inflammation. Trends Cell Biol. 2012;22(11):557–66.PubMedCrossRef
315.
go back to reference Mussbacher M, Salzmann M, Brostjan C, Hoesel B, Schoergenhofer C, Datler H, Hohensinner P, Basílio J, Petzelbauer P, Assinger A et al: Cell type-specific roles of NF-κB linking inflammation and thrombosis. Front Immunol 2019, 10(85). Mussbacher M, Salzmann M, Brostjan C, Hoesel B, Schoergenhofer C, Datler H, Hohensinner P, Basílio J, Petzelbauer P, Assinger A et al: Cell type-specific roles of NF-κB linking inflammation and thrombosis. Front Immunol 2019, 10(85).
316.
go back to reference Dey P, Panga V, Raghunathan S. A cytokine signalling network for the regulation of inducible nitric oxide synthase expression in rheumatoid arthritis. PLoS One. 2016;11(9):e0161306.PubMedPubMedCentralCrossRef Dey P, Panga V, Raghunathan S. A cytokine signalling network for the regulation of inducible nitric oxide synthase expression in rheumatoid arthritis. PLoS One. 2016;11(9):e0161306.PubMedPubMedCentralCrossRef
318.
go back to reference Arias-Salvatierra D, Silbergeld EK, Acosta-Saavedra LC, Calderon-Aranda ES. Role of nitric oxide produced by iNOS through NF-kappaB pathway in migration of cerebellar granule neurons induced by lipopolysaccharide. Cell Signal. 2011;23(2):425–35.PubMedCrossRef Arias-Salvatierra D, Silbergeld EK, Acosta-Saavedra LC, Calderon-Aranda ES. Role of nitric oxide produced by iNOS through NF-kappaB pathway in migration of cerebellar granule neurons induced by lipopolysaccharide. Cell Signal. 2011;23(2):425–35.PubMedCrossRef
320.
go back to reference Nakamura T, Tu S, Akhtar Mohd W, Sunico Carmen R, S-i O, Lipton Stuart A. Aberrant protein S-nitrosylation in neurodegenerative diseases. Neuron. 2013;78(4):596–614.PubMedPubMedCentralCrossRef Nakamura T, Tu S, Akhtar Mohd W, Sunico Carmen R, S-i O, Lipton Stuart A. Aberrant protein S-nitrosylation in neurodegenerative diseases. Neuron. 2013;78(4):596–614.PubMedPubMedCentralCrossRef
321.
go back to reference S-i O, Lipton SA. S-Nitrosylation in neurogenesis and neuronal development. Biochim Biophys Acta. 2015;1850(8):1588–93.CrossRef S-i O, Lipton SA. S-Nitrosylation in neurogenesis and neuronal development. Biochim Biophys Acta. 2015;1850(8):1588–93.CrossRef
322.
go back to reference Fan W, Fang R, Wu X, Liu J, Feng M, Dai G, Chen G, Wu G. Shear-sensitive microRNA-34a modulates flow-dependent regulation of endothelial inflammation. J Cell Sci. 2015;128(1):70–80.PubMedCrossRef Fan W, Fang R, Wu X, Liu J, Feng M, Dai G, Chen G, Wu G. Shear-sensitive microRNA-34a modulates flow-dependent regulation of endothelial inflammation. J Cell Sci. 2015;128(1):70–80.PubMedCrossRef
323.
go back to reference Sweet DR, Fan L, Hsieh PN, Jain MK: Krüppel-like factors in vascular inflammation: mechanistic insights and therapeutic potential. Fron Cardiovasc Med 2018, 5(6). Sweet DR, Fan L, Hsieh PN, Jain MK: Krüppel-like factors in vascular inflammation: mechanistic insights and therapeutic potential. Fron Cardiovasc Med 2018, 5(6).
325.
go back to reference Nakamura T, Lipton SA. Protein S-nitrosylation as a therapeutic target for neurodegenerative diseases. Trends Pharmacol Sci. 2016;37(1):73–84.PubMedCrossRef Nakamura T, Lipton SA. Protein S-nitrosylation as a therapeutic target for neurodegenerative diseases. Trends Pharmacol Sci. 2016;37(1):73–84.PubMedCrossRef
326.
go back to reference Morris G, Berk M, Klein H, Walder K, Galecki P, Maes M. Nitrosative stress, hypernitrosylation, and autoimmune responses to nitrosylated proteins: new pathways in neuroprogressive disorders including depression and chronic fatigue syndrome. Mol Neurobiol. 2017;54(6):4271–91.PubMedCrossRef Morris G, Berk M, Klein H, Walder K, Galecki P, Maes M. Nitrosative stress, hypernitrosylation, and autoimmune responses to nitrosylated proteins: new pathways in neuroprogressive disorders including depression and chronic fatigue syndrome. Mol Neurobiol. 2017;54(6):4271–91.PubMedCrossRef
327.
go back to reference Morris G, Walder K, Carvalho AF, Tye SJ, Lucas K, Berk M, Maes M. The role of hypernitrosylation in the pathogenesis and pathophysiology of neuroprogressive diseases. Neurosci Biobehav Rev. 2018;84:453–69.PubMedCrossRef Morris G, Walder K, Carvalho AF, Tye SJ, Lucas K, Berk M, Maes M. The role of hypernitrosylation in the pathogenesis and pathophysiology of neuroprogressive diseases. Neurosci Biobehav Rev. 2018;84:453–69.PubMedCrossRef
328.
329.
go back to reference Doulias P-T, Tenopoulou M, Greene JL, Raju K, Ischiropoulos H. Nitric oxide regulates mitochondrial fatty acid metabolism through reversible protein S-nitrosylation. Sci Signal. 2013;6(256):rs1.PubMedPubMedCentralCrossRef Doulias P-T, Tenopoulou M, Greene JL, Raju K, Ischiropoulos H. Nitric oxide regulates mitochondrial fatty acid metabolism through reversible protein S-nitrosylation. Sci Signal. 2013;6(256):rs1.PubMedPubMedCentralCrossRef
330.
go back to reference Gould N, Doulias P-T, Tenopoulou M, Raju K, Ischiropoulos H. Regulation of protein function and signaling by reversible cysteine S-nitrosylation. J Biol Chem. 2013;288(37):26473–9.PubMedPubMedCentralCrossRef Gould N, Doulias P-T, Tenopoulou M, Raju K, Ischiropoulos H. Regulation of protein function and signaling by reversible cysteine S-nitrosylation. J Biol Chem. 2013;288(37):26473–9.PubMedPubMedCentralCrossRef
331.
go back to reference Xu X, Qiu H, Shi F, Wang Z, Wang X, Jin L, Chi L, Zhang Q. The protein S-nitrosylation of splicing and translational machinery in vascular endothelial cells is susceptible to oxidative stress induced by oxidized low-density lipoprotein. J Proteome. 2019;195:11–22.CrossRef Xu X, Qiu H, Shi F, Wang Z, Wang X, Jin L, Chi L, Zhang Q. The protein S-nitrosylation of splicing and translational machinery in vascular endothelial cells is susceptible to oxidative stress induced by oxidized low-density lipoprotein. J Proteome. 2019;195:11–22.CrossRef
332.
go back to reference Guequen A, Carrasco R, Zamorano P, Rebolledo L, Burboa P, Sarmiento J, Boric MP, Korayem A, Duran WN, Sanchez FA. S-nitrosylation regulates VE-cadherin phosphorylation and internalization in microvascular permeability. Am J Phys Heart Circ Phys. 2016;310(8):H1039–44. Guequen A, Carrasco R, Zamorano P, Rebolledo L, Burboa P, Sarmiento J, Boric MP, Korayem A, Duran WN, Sanchez FA. S-nitrosylation regulates VE-cadherin phosphorylation and internalization in microvascular permeability. Am J Phys Heart Circ Phys. 2016;310(8):H1039–44.
333.
go back to reference Marin N, Zamorano P, Carrasco R, Mujica P, Gonzalez FG, Quezada C, Meininger CJ, Boric MP, Duran WN, Sanchez FA. S-Nitrosation of beta-catenin and p120 catenin: a novel regulatory mechanism in endothelial hyperpermeability. Circ Res. 2012;111(5):553–63.PubMedPubMedCentralCrossRef Marin N, Zamorano P, Carrasco R, Mujica P, Gonzalez FG, Quezada C, Meininger CJ, Boric MP, Duran WN, Sanchez FA. S-Nitrosation of beta-catenin and p120 catenin: a novel regulatory mechanism in endothelial hyperpermeability. Circ Res. 2012;111(5):553–63.PubMedPubMedCentralCrossRef
334.
go back to reference Shang F, Wang SC, Hsu CY, Miao Y, Martin M, Yin Y, Wu CC, Wang YT, Wu G, Chien S, et al. MicroRNA-92a mediates endothelial dysfunction in CKD. J Am Soc Nephrol. 2017;28(11):3251–61.PubMedPubMedCentralCrossRef Shang F, Wang SC, Hsu CY, Miao Y, Martin M, Yin Y, Wu CC, Wang YT, Wu G, Chien S, et al. MicroRNA-92a mediates endothelial dysfunction in CKD. J Am Soc Nephrol. 2017;28(11):3251–61.PubMedPubMedCentralCrossRef
335.
go back to reference Chen Z, Wen L, Martin M, Hsu CY, Fang L, Lin FM, Lin TY, Geary MJ, Geary GG, Zhao Y, et al. Oxidative stress activates endothelial innate immunity via sterol regulatory element binding protein 2 (SREBP2) transactivation of microRNA-92a. Circulation. 2015;131(9):805–14.PubMedCrossRef Chen Z, Wen L, Martin M, Hsu CY, Fang L, Lin FM, Lin TY, Geary MJ, Geary GG, Zhao Y, et al. Oxidative stress activates endothelial innate immunity via sterol regulatory element binding protein 2 (SREBP2) transactivation of microRNA-92a. Circulation. 2015;131(9):805–14.PubMedCrossRef
336.
go back to reference Huang T, Wang-Johanning F, Zhou F, Kallon H, Wei Y. MicroRNAs serve as a bridge between oxidative stress and gastric cancer (review). Int J Oncol. 2016;49(5):1791–800.PubMedCrossRef Huang T, Wang-Johanning F, Zhou F, Kallon H, Wei Y. MicroRNAs serve as a bridge between oxidative stress and gastric cancer (review). Int J Oncol. 2016;49(5):1791–800.PubMedCrossRef
337.
go back to reference Engedal N, Žerovnik E, Rudov A, Galli F, Olivieri F, Procopio AD, Rippo MR, Monsurrò V, Betti M, Albertini MC. From oxidative stress damage to pathways, networks, and autophagy via microRNAs. Oxidative Med Cell Longev. 2018;2018:4968321.CrossRef Engedal N, Žerovnik E, Rudov A, Galli F, Olivieri F, Procopio AD, Rippo MR, Monsurrò V, Betti M, Albertini MC. From oxidative stress damage to pathways, networks, and autophagy via microRNAs. Oxidative Med Cell Longev. 2018;2018:4968321.CrossRef
338.
go back to reference Heid J, Cencioni C, Ripa R, Baumgart M, Atlante S, Milano G, Scopece A, Kuenne C, Guenther S, Azzimato V, et al. Age-dependent increase of oxidative stress regulates microRNA-29 family preserving cardiac health. Sci Rep. 2017;7(1):16839.PubMedPubMedCentralCrossRef Heid J, Cencioni C, Ripa R, Baumgart M, Atlante S, Milano G, Scopece A, Kuenne C, Guenther S, Azzimato V, et al. Age-dependent increase of oxidative stress regulates microRNA-29 family preserving cardiac health. Sci Rep. 2017;7(1):16839.PubMedPubMedCentralCrossRef
339.
go back to reference Wang S, Guo C, Yu M, Ning X, Yan B, Zhao J, Yang A, Yan H. Identification of H2O2 induced oxidative stress associated microRNAs in HLE-B3 cells and their clinical relevance to the progression of age-related nuclear cataract. BMC Ophthalmol. 2018;18(1):93.PubMedPubMedCentralCrossRef Wang S, Guo C, Yu M, Ning X, Yan B, Zhao J, Yang A, Yan H. Identification of H2O2 induced oxidative stress associated microRNAs in HLE-B3 cells and their clinical relevance to the progression of age-related nuclear cataract. BMC Ophthalmol. 2018;18(1):93.PubMedPubMedCentralCrossRef
340.
go back to reference Wan Y, Cui R, Gu J, Zhang X, Xiang X, Liu C, Qu K, Lin T. Identification of four oxidative stress-responsive microRNAs, miR-34a-5p, miR-1915-3p, miR-638, and miR-150-3p, in hepatocellular carcinoma. Oxidative Med Cell Longev. 2017;2017:5189138.CrossRef Wan Y, Cui R, Gu J, Zhang X, Xiang X, Liu C, Qu K, Lin T. Identification of four oxidative stress-responsive microRNAs, miR-34a-5p, miR-1915-3p, miR-638, and miR-150-3p, in hepatocellular carcinoma. Oxidative Med Cell Longev. 2017;2017:5189138.CrossRef
341.
go back to reference Magenta A, Cencioni C, Fasanaro P, Zaccagnini G, Greco S, Sarra-Ferraris G, Antonini A, Martelli F, Capogrossi MC. miR-200c is upregulated by oxidative stress and induces endothelial cell apoptosis and senescence via ZEB1 inhibition. Cell Death Differ. 2011;18(10):1628–39.PubMedPubMedCentralCrossRef Magenta A, Cencioni C, Fasanaro P, Zaccagnini G, Greco S, Sarra-Ferraris G, Antonini A, Martelli F, Capogrossi MC. miR-200c is upregulated by oxidative stress and induces endothelial cell apoptosis and senescence via ZEB1 inhibition. Cell Death Differ. 2011;18(10):1628–39.PubMedPubMedCentralCrossRef
342.
go back to reference Carlomosti F, D'Agostino M, Beji S, Torcinaro A, Rizzi R, Zaccagnini G, Maimone B, Di Stefano V, De Santa F, Cordisco S, et al. Oxidative stress-induced miR-200c disrupts the regulatory loop among SIRT1, FOXO1, and eNOS. Antioxid Redox Signal. 2017;27(6):328–44.PubMedCrossRef Carlomosti F, D'Agostino M, Beji S, Torcinaro A, Rizzi R, Zaccagnini G, Maimone B, Di Stefano V, De Santa F, Cordisco S, et al. Oxidative stress-induced miR-200c disrupts the regulatory loop among SIRT1, FOXO1, and eNOS. Antioxid Redox Signal. 2017;27(6):328–44.PubMedCrossRef
343.
go back to reference Zhang W, Huang Q, Zeng Z, Wu J, Zhang Y, Chen Z. Sirt1 inhibits oxidative stress in vascular endothelial cells. Oxidative Med Cell Longev. 2017;2017:7543973. Zhang W, Huang Q, Zeng Z, Wu J, Zhang Y, Chen Z. Sirt1 inhibits oxidative stress in vascular endothelial cells. Oxidative Med Cell Longev. 2017;2017:7543973.
344.
go back to reference Zarzuelo MJ, Lopez-Sepulveda R, Sanchez M, Romero M, Gomez-Guzman M, Ungvary Z, Perez-Vizcaino F, Jimenez R, Duarte J. SIRT1 inhibits NADPH oxidase activation and protects endothelial function in the rat aorta: implications for vascular aging. Biochem Pharmacol. 2013;85(9):1288–96.PubMedCrossRef Zarzuelo MJ, Lopez-Sepulveda R, Sanchez M, Romero M, Gomez-Guzman M, Ungvary Z, Perez-Vizcaino F, Jimenez R, Duarte J. SIRT1 inhibits NADPH oxidase activation and protects endothelial function in the rat aorta: implications for vascular aging. Biochem Pharmacol. 2013;85(9):1288–96.PubMedCrossRef
345.
go back to reference Kong X, Guan J, Li J, Wei J, Wang R. P66(Shc)-SIRT1 regulation of oxidative stress protects against cardio-cerebral vascular disease. Mol Neurobiol. 2017;54(7):5277–85.PubMedCrossRef Kong X, Guan J, Li J, Wei J, Wang R. P66(Shc)-SIRT1 regulation of oxidative stress protects against cardio-cerebral vascular disease. Mol Neurobiol. 2017;54(7):5277–85.PubMedCrossRef
346.
go back to reference Zhang J, Cao Q, Li S, Lu X, Zhao Y, Guan JS, Chen JC, Wu Q, Chen GQ. 3-Hydroxybutyrate methyl ester as a potential drug against Alzheimer’s disease via mitochondria protection mechanism. Biomaterials. 2013;34(30):7552–62.PubMedCrossRef Zhang J, Cao Q, Li S, Lu X, Zhao Y, Guan JS, Chen JC, Wu Q, Chen GQ. 3-Hydroxybutyrate methyl ester as a potential drug against Alzheimer’s disease via mitochondria protection mechanism. Biomaterials. 2013;34(30):7552–62.PubMedCrossRef
347.
go back to reference Devier DJ, Lovera JF, Lukiw WJ: Increase in NF-κB-sensitive miRNA-146a and miRNA-155 in multiple sclerosis (MS) and pro-inflammatory neurodegeneration. Front Mol Neurosci 2015, 8(5). Devier DJ, Lovera JF, Lukiw WJ: Increase in NF-κB-sensitive miRNA-146a and miRNA-155 in multiple sclerosis (MS) and pro-inflammatory neurodegeneration. Front Mol Neurosci 2015, 8(5).
348.
go back to reference Huang Y, Crawford M, Higuita-Castro N, Nana-Sinkam P, Ghadiali SN. miR-146a regulates mechanotransduction and pressure-induced inflammation in small airway epithelium. FASEB J. 2012;26(8):3351–64.PubMedPubMedCentralCrossRef Huang Y, Crawford M, Higuita-Castro N, Nana-Sinkam P, Ghadiali SN. miR-146a regulates mechanotransduction and pressure-induced inflammation in small airway epithelium. FASEB J. 2012;26(8):3351–64.PubMedPubMedCentralCrossRef
349.
go back to reference Cheleschi S, De Palma A, Pascarelli NA, Giordano N, Galeazzi M, Tenti S, Fioravanti A. Could oxidative stress regulate the expression of microRNA-146a and microRNA-34a in human osteoarthritic chondrocyte cultures? Int J Mol Sci. 2017;18(12). Cheleschi S, De Palma A, Pascarelli NA, Giordano N, Galeazzi M, Tenti S, Fioravanti A. Could oxidative stress regulate the expression of microRNA-146a and microRNA-34a in human osteoarthritic chondrocyte cultures? Int J Mol Sci. 2017;18(12).
350.
go back to reference Rippo MR, Olivieri F, Monsurro V, Prattichizzo F, Albertini MC, Procopio AD. MitomiRs in human inflamm-aging: a hypothesis involving miR-181a, miR-34a and miR-146a. Exp Gerontol. 2014;56:154–63.PubMedCrossRef Rippo MR, Olivieri F, Monsurro V, Prattichizzo F, Albertini MC, Procopio AD. MitomiRs in human inflamm-aging: a hypothesis involving miR-181a, miR-34a and miR-146a. Exp Gerontol. 2014;56:154–63.PubMedCrossRef
351.
go back to reference Giuliani A, Prattichizzo F, Micolucci L, Ceriello A, Procopio AD, Rippo MR. Mitochondrial (dys) function in inflammaging: do mitomiRs influence the energetic, oxidative, and inflammatory status of senescent cells? Mediat Inflamm. 2017;2017:2309034.CrossRef Giuliani A, Prattichizzo F, Micolucci L, Ceriello A, Procopio AD, Rippo MR. Mitochondrial (dys) function in inflammaging: do mitomiRs influence the energetic, oxidative, and inflammatory status of senescent cells? Mediat Inflamm. 2017;2017:2309034.CrossRef
352.
go back to reference Leirer DJ, Iyegbe CO, Di Forti M, Patel H, Carra E, Fraietta S, Colizzi M, Mondelli V, Quattrone D, Lally J, et al. Differential gene expression analysis in blood of first episode psychosis patients. Schizophr Res. 2019;209:88–97.PubMedPubMedCentralCrossRef Leirer DJ, Iyegbe CO, Di Forti M, Patel H, Carra E, Fraietta S, Colizzi M, Mondelli V, Quattrone D, Lally J, et al. Differential gene expression analysis in blood of first episode psychosis patients. Schizophr Res. 2019;209:88–97.PubMedPubMedCentralCrossRef
353.
go back to reference Pesce M, Ferrone A, Rizzuto A, Tatangelo R, Iezzi I, Ladu S, Franceschelli S, Speranza L, Patruno A, Felaco M, et al. The SHP-1 expression is associated with cytokines and psychopathological status in unmedicated first episode schizophrenia patients. Brain Behav Immun. 2014;41. Pesce M, Ferrone A, Rizzuto A, Tatangelo R, Iezzi I, Ladu S, Franceschelli S, Speranza L, Patruno A, Felaco M, et al. The SHP-1 expression is associated with cytokines and psychopathological status in unmedicated first episode schizophrenia patients. Brain Behav Immun. 2014;41.
354.
go back to reference Miklowitz DJ, Portnoff LC, Armstrong CC, Keenan-Miller D, Breen EC, Muscatell KA, Eisenberger NI, Irwin MR. Inflammatory cytokines and nuclear factor-kappa B activation in adolescents with bipolar and major depressive disorders. Psychiatry Res. 2016;241:315–22.PubMedPubMedCentralCrossRef Miklowitz DJ, Portnoff LC, Armstrong CC, Keenan-Miller D, Breen EC, Muscatell KA, Eisenberger NI, Irwin MR. Inflammatory cytokines and nuclear factor-kappa B activation in adolescents with bipolar and major depressive disorders. Psychiatry Res. 2016;241:315–22.PubMedPubMedCentralCrossRef
355.
go back to reference Spiliotaki M, Salpeas V, Malitas P, Alevizos V, Moutsatsou P. Altered glucocorticoid receptor signaling cascade in lymphocytes of bipolar disorder patients. Psychoneuroendocrinology. 2006;31(6):748–60.PubMedCrossRef Spiliotaki M, Salpeas V, Malitas P, Alevizos V, Moutsatsou P. Altered glucocorticoid receptor signaling cascade in lymphocytes of bipolar disorder patients. Psychoneuroendocrinology. 2006;31(6):748–60.PubMedCrossRef
356.
go back to reference Ormonde do Carmo MB, Mendes-Ribeiro AC, Matsuura C, Pinto VL, Mury WV, Pinto NO, Moss MB, Ferraz MR, Brunini TM. Major depression induces oxidative stress and platelet hyperaggregability. J Psychiatr Res. 2015;61:19–24.PubMedCrossRef Ormonde do Carmo MB, Mendes-Ribeiro AC, Matsuura C, Pinto VL, Mury WV, Pinto NO, Moss MB, Ferraz MR, Brunini TM. Major depression induces oxidative stress and platelet hyperaggregability. J Psychiatr Res. 2015;61:19–24.PubMedCrossRef
357.
go back to reference Cai L, Xu L, Wei L, Chen W. Relationship of mean platelet volume to MDD: a retrospective study. Shanghai Arch Psychiatry. 2017;29(1):21–9.PubMed Cai L, Xu L, Wei L, Chen W. Relationship of mean platelet volume to MDD: a retrospective study. Shanghai Arch Psychiatry. 2017;29(1):21–9.PubMed
358.
go back to reference Qiu H, Liu Y, He H, Wu Y, He W, Huang G, He J. The association between mean platelet volume levels and poststroke depression. Brain Behav. 2018;8(10):e01114.PubMedPubMedCentralCrossRef Qiu H, Liu Y, He H, Wu Y, He W, Huang G, He J. The association between mean platelet volume levels and poststroke depression. Brain Behav. 2018;8(10):e01114.PubMedPubMedCentralCrossRef
359.
go back to reference Fontoura PC, Pinto VL, Matsuura C, Resende Ade C, de Bem GF, Ferraz MR, Cheniaux E, Brunini TM, Mendes-Ribeiro AC. Defective nitric oxide-cyclic guanosine monophosphate signaling in patients with bipolar disorder: a potential role for platelet dysfunction. Psychosom Med. 2012;74(8):873–7.PubMedCrossRef Fontoura PC, Pinto VL, Matsuura C, Resende Ade C, de Bem GF, Ferraz MR, Cheniaux E, Brunini TM, Mendes-Ribeiro AC. Defective nitric oxide-cyclic guanosine monophosphate signaling in patients with bipolar disorder: a potential role for platelet dysfunction. Psychosom Med. 2012;74(8):873–7.PubMedCrossRef
360.
go back to reference Wysokiński A, Szczepocka E. Platelet parameters (PLT, MPV, P-LCR) in patients with schizophrenia, unipolar depression and bipolar disorder. Psychiatry Res. 2016;237:238–45.PubMedCrossRef Wysokiński A, Szczepocka E. Platelet parameters (PLT, MPV, P-LCR) in patients with schizophrenia, unipolar depression and bipolar disorder. Psychiatry Res. 2016;237:238–45.PubMedCrossRef
361.
362.
go back to reference Inanli I, Aydin M, Caliskan AM, Eren I. Neutrophil/lymphocyte ratio, monocyte/lymphocyte ratio, and mean platelet volume as systemic inflammatory markers in different states of bipolar disorder. Nordic J Psychiatry. 2019;73(6):372–9.CrossRef Inanli I, Aydin M, Caliskan AM, Eren I. Neutrophil/lymphocyte ratio, monocyte/lymphocyte ratio, and mean platelet volume as systemic inflammatory markers in different states of bipolar disorder. Nordic J Psychiatry. 2019;73(6):372–9.CrossRef
363.
go back to reference Wu CC, Tsai FM, Chen ML, Wu S, Lee MC, Tsai TC, Wang LK, Wang CH. Antipsychotic drugs inhibit platelet aggregation via P2Y 1 and P2Y 12 receptors. Biomed Res Int. 2016;2016:2532371.PubMedPubMedCentral Wu CC, Tsai FM, Chen ML, Wu S, Lee MC, Tsai TC, Wang LK, Wang CH. Antipsychotic drugs inhibit platelet aggregation via P2Y 1 and P2Y 12 receptors. Biomed Res Int. 2016;2016:2532371.PubMedPubMedCentral
364.
go back to reference Semiz M, Yucel H, Kavakci O, Yildirim O, Zorlu A, Yilmaz MB, Kucukdurmaz Z, Canan F. Atypical antipsychotic use is an independent predictor for the increased mean platelet volume in patients with schizophrenia: a preliminary study. J Res Med Sci. 2013;18(7):561–6.PubMedPubMedCentral Semiz M, Yucel H, Kavakci O, Yildirim O, Zorlu A, Yilmaz MB, Kucukdurmaz Z, Canan F. Atypical antipsychotic use is an independent predictor for the increased mean platelet volume in patients with schizophrenia: a preliminary study. J Res Med Sci. 2013;18(7):561–6.PubMedPubMedCentral
365.
go back to reference Lee J, Powell V, Remington G. Mean platelet volume in schizophrenia unaltered after 1year of clozapine exposure. Schizophr Res. 2014;157(1–3):134–6.PubMedCrossRef Lee J, Powell V, Remington G. Mean platelet volume in schizophrenia unaltered after 1year of clozapine exposure. Schizophr Res. 2014;157(1–3):134–6.PubMedCrossRef
366.
go back to reference Camkurt MA, Findikli E, Izci F, Kurutas EB, Tuman TC. Evaluation of malondialdehyde, superoxide dismutase and catalase activity and their diagnostic value in drug naive, first episode, non-smoker major depression patients and healthy controls. Psychiatry Res. 2016;238:81–5.PubMedCrossRef Camkurt MA, Findikli E, Izci F, Kurutas EB, Tuman TC. Evaluation of malondialdehyde, superoxide dismutase and catalase activity and their diagnostic value in drug naive, first episode, non-smoker major depression patients and healthy controls. Psychiatry Res. 2016;238:81–5.PubMedCrossRef
367.
go back to reference Geaghan M, Cairns MJ. MicroRNA and posttranscriptional dysregulation in psychiatry. Biol Psychiatry. 2015;78(4):231–9.PubMedCrossRef Geaghan M, Cairns MJ. MicroRNA and posttranscriptional dysregulation in psychiatry. Biol Psychiatry. 2015;78(4):231–9.PubMedCrossRef
368.
go back to reference He K, Guo C, Guo M, Tong S, Zhang Q, Sun H, He L, Shi Y. Identification of serum microRNAs as diagnostic biomarkers for schizophrenia. Hereditas. 2019;156:23.PubMedPubMedCentralCrossRef He K, Guo C, Guo M, Tong S, Zhang Q, Sun H, He L, Shi Y. Identification of serum microRNAs as diagnostic biomarkers for schizophrenia. Hereditas. 2019;156:23.PubMedPubMedCentralCrossRef
369.
go back to reference Vaccarino V, Brennan M-L, Miller AH, Bremner JD, Ritchie JC, Lindau F, Veledar E, Su S, Murrah NV, Jones L, et al. Association of major depressive disorder with serum myeloperoxidase and other markers of inflammation: a twin study. Biol Psychiatry. 2008;64(6):476–83.PubMedPubMedCentralCrossRef Vaccarino V, Brennan M-L, Miller AH, Bremner JD, Ritchie JC, Lindau F, Veledar E, Su S, Murrah NV, Jones L, et al. Association of major depressive disorder with serum myeloperoxidase and other markers of inflammation: a twin study. Biol Psychiatry. 2008;64(6):476–83.PubMedPubMedCentralCrossRef
370.
go back to reference Talarowska M, Szemraj J, Galecki P. Myeloperoxidase gene expression and cognitive functions in depression. Adv Med Sci. 2015;60(1):1–5.PubMedCrossRef Talarowska M, Szemraj J, Galecki P. Myeloperoxidase gene expression and cognitive functions in depression. Adv Med Sci. 2015;60(1):1–5.PubMedCrossRef
371.
go back to reference Selek S, Altindag A, Saracoglu G, Aksoy N. Oxidative markers of myeloperoxidase and catalase and their diagnostic performance in bipolar disorder. J Affect Disord. 2015;181:92–5.PubMedCrossRef Selek S, Altindag A, Saracoglu G, Aksoy N. Oxidative markers of myeloperoxidase and catalase and their diagnostic performance in bipolar disorder. J Affect Disord. 2015;181:92–5.PubMedCrossRef
372.
go back to reference Reponen E, Dieset I, Tesli M, Mørch RH, Steen NE, Hope S, Vedal TSJ, Aas M, Szabo A, Gohar SM, et al. T8. Atherogenic lipid ratios related to myeloperoxidase and C-reactive protein levels in psychotic disorders. Schizophr Bull. 2019;45(Supplement_2):S206.PubMedCentralCrossRef Reponen E, Dieset I, Tesli M, Mørch RH, Steen NE, Hope S, Vedal TSJ, Aas M, Szabo A, Gohar SM, et al. T8. Atherogenic lipid ratios related to myeloperoxidase and C-reactive protein levels in psychotic disorders. Schizophr Bull. 2019;45(Supplement_2):S206.PubMedCentralCrossRef
373.
go back to reference Kesebir S, Süner O, Yaylaci ET, Bayrak A, Turan C. Increased uric acid levels in bipolar disorder: is it trait or state? J Biol Regul Homeost Agents. 2013;27(4):981–8.PubMed Kesebir S, Süner O, Yaylaci ET, Bayrak A, Turan C. Increased uric acid levels in bipolar disorder: is it trait or state? J Biol Regul Homeost Agents. 2013;27(4):981–8.PubMed
374.
go back to reference Albert U, De Cori D, Aguglia A, Barbaro F, Bogetto F, Maina G. Increased uric acid levels in bipolar disorder subjects during different phases of illness. J Affect Disord. 2015;173:170–5.PubMedCrossRef Albert U, De Cori D, Aguglia A, Barbaro F, Bogetto F, Maina G. Increased uric acid levels in bipolar disorder subjects during different phases of illness. J Affect Disord. 2015;173:170–5.PubMedCrossRef
375.
go back to reference Bartoli F, Crocamo C, Clerici M, Carra G. Allopurinol as add-on treatment for mania symptoms in bipolar disorder: systematic review and meta-analysis of randomised controlled trials. Br J Psychiatry. 2017;210(1):10–5.PubMedCrossRef Bartoli F, Crocamo C, Clerici M, Carra G. Allopurinol as add-on treatment for mania symptoms in bipolar disorder: systematic review and meta-analysis of randomised controlled trials. Br J Psychiatry. 2017;210(1):10–5.PubMedCrossRef
376.
go back to reference Chen AT, Malmstrom T, Nasrallah HA. Allopurinol augmentation in acute mania: a meta-analysis of placebo-controlled trials. J Affect Disord. 2018;226:245–50.PubMedCrossRef Chen AT, Malmstrom T, Nasrallah HA. Allopurinol augmentation in acute mania: a meta-analysis of placebo-controlled trials. J Affect Disord. 2018;226:245–50.PubMedCrossRef
377.
go back to reference Gültekin BK, Kesebir S, Kabak SG, Ergün FF, Tatlidil Yaylaci E. Are uric acid levels different from healthy subjects in bipolar affective disorder and schizophrenia?: relationship between clinical improvement and episode severity in male patients. Noro Psikiyatr Ars. 2014;51(3):229–32.PubMedPubMedCentralCrossRef Gültekin BK, Kesebir S, Kabak SG, Ergün FF, Tatlidil Yaylaci E. Are uric acid levels different from healthy subjects in bipolar affective disorder and schizophrenia?: relationship between clinical improvement and episode severity in male patients. Noro Psikiyatr Ars. 2014;51(3):229–32.PubMedPubMedCentralCrossRef
378.
go back to reference Michel TM, Sheldrick AJ, Camara S, Grunblatt E, Schneider F, Riederer P. Alteration of the pro-oxidant xanthine oxidase (XO) in the thalamus and occipital cortex of patients with schizophrenia. World J Biol Psychiatry. 2011;12(8):588–97.PubMedCrossRef Michel TM, Sheldrick AJ, Camara S, Grunblatt E, Schneider F, Riederer P. Alteration of the pro-oxidant xanthine oxidase (XO) in the thalamus and occipital cortex of patients with schizophrenia. World J Biol Psychiatry. 2011;12(8):588–97.PubMedCrossRef
379.
go back to reference Godin O, Llorca PM, Girerd N, Leboyer M, Fond G. Metabolic syndrome, abdominal obesity and hyperuricemia in schizophrenia: results from the FACE-SZ dataset. Eur Psychiatry. 2015;30(8):S130.CrossRef Godin O, Llorca PM, Girerd N, Leboyer M, Fond G. Metabolic syndrome, abdominal obesity and hyperuricemia in schizophrenia: results from the FACE-SZ dataset. Eur Psychiatry. 2015;30(8):S130.CrossRef
380.
go back to reference Rajan S, Zalpuri I, Harrington A, Cimpeanu C, Song X, Fan X. Relationship between serum uric acid level and cardiometabolic risks in nondiabetic patients with schizophrenia. Int Clin Psychopharmacol. 2016;31(1):51–6.PubMedCrossRef Rajan S, Zalpuri I, Harrington A, Cimpeanu C, Song X, Fan X. Relationship between serum uric acid level and cardiometabolic risks in nondiabetic patients with schizophrenia. Int Clin Psychopharmacol. 2016;31(1):51–6.PubMedCrossRef
381.
go back to reference Nagamine T. Abnormal laboratory values during the acute and recovery phases in schizophrenic patients: a retrospective study. Neuropsychiatr Dis Treat. 2010;6:281–8.PubMedPubMedCentral Nagamine T. Abnormal laboratory values during the acute and recovery phases in schizophrenic patients: a retrospective study. Neuropsychiatr Dis Treat. 2010;6:281–8.PubMedPubMedCentral
382.
go back to reference Yao JK, Dougherty GG Jr, Reddy RD, Keshavan MS, Montrose DM, Matson WR, McEvoy J, Kaddurah-Daouk R. Homeostatic imbalance of purine catabolism in first-episode neuroleptic-naïve patients with schizophrenia. PLoS One. 2010;5(3):e9508.PubMedPubMedCentralCrossRef Yao JK, Dougherty GG Jr, Reddy RD, Keshavan MS, Montrose DM, Matson WR, McEvoy J, Kaddurah-Daouk R. Homeostatic imbalance of purine catabolism in first-episode neuroleptic-naïve patients with schizophrenia. PLoS One. 2010;5(3):e9508.PubMedPubMedCentralCrossRef
383.
go back to reference Yao JK, Dougherty GG, Reddy RD, Matson WR, Kaddurah-Daouk R, Keshavan MS. Associations between purine metabolites and monoamine neurotransmitters in first-episode psychosis. Front Cell Neurosci. 2013;7:90.PubMedPubMedCentral Yao JK, Dougherty GG, Reddy RD, Matson WR, Kaddurah-Daouk R, Keshavan MS. Associations between purine metabolites and monoamine neurotransmitters in first-episode psychosis. Front Cell Neurosci. 2013;7:90.PubMedPubMedCentral
384.
go back to reference Herken H, Gurel A, Selek S, Armutcu F, Ozen ME, Bulut M, Kap O, Yumru M, Savas HA, Akyol O. Adenosine deaminase, nitric oxide, superoxide dismutase, and xanthine oxidase in patients with major depression: impact of antidepressant treatment. Arch Med Res. 2007;38(2):247–52.PubMedCrossRef Herken H, Gurel A, Selek S, Armutcu F, Ozen ME, Bulut M, Kap O, Yumru M, Savas HA, Akyol O. Adenosine deaminase, nitric oxide, superoxide dismutase, and xanthine oxidase in patients with major depression: impact of antidepressant treatment. Arch Med Res. 2007;38(2):247–52.PubMedCrossRef
385.
go back to reference Michel TM, Camara S, Tatschner T, Frangou S, Sheldrick AJ, Riederer P, Grunblatt E. Increased xanthine oxidase in the thalamus and putamen in depression. World J Biol Psychiatry. 2010;11(2 Pt 2):314–20.PubMedCrossRef Michel TM, Camara S, Tatschner T, Frangou S, Sheldrick AJ, Riederer P, Grunblatt E. Increased xanthine oxidase in the thalamus and putamen in depression. World J Biol Psychiatry. 2010;11(2 Pt 2):314–20.PubMedCrossRef
386.
go back to reference Dos Santos Oliveira PM, Santos V, Coroa M, Ribeiro J, Madeira N. Serum uric acid as a predictor of bipolarity in individuals with a major depressive episode. Bipolar Disord. 2019;21(3):235–43.PubMedCrossRef Dos Santos Oliveira PM, Santos V, Coroa M, Ribeiro J, Madeira N. Serum uric acid as a predictor of bipolarity in individuals with a major depressive episode. Bipolar Disord. 2019;21(3):235–43.PubMedCrossRef
387.
go back to reference Black CN, Bot M, Scheffer PG, Snieder H, Penninx BWJH. Uric acid in major depressive and anxiety disorders. J Affect Disord. 2018;225:684–90.PubMedCrossRef Black CN, Bot M, Scheffer PG, Snieder H, Penninx BWJH. Uric acid in major depressive and anxiety disorders. J Affect Disord. 2018;225:684–90.PubMedCrossRef
388.
go back to reference Bartoli F, Trotta G, Crocamo C, Malerba MR, Clerici M, Carrà G. Antioxidant uric acid in treated and untreated subjects with major depressive disorder: a meta-analysis and meta-regression. Eur Arch Psychiatry Clin Neurosci. 2018;268(2):119–27.PubMedCrossRef Bartoli F, Trotta G, Crocamo C, Malerba MR, Clerici M, Carrà G. Antioxidant uric acid in treated and untreated subjects with major depressive disorder: a meta-analysis and meta-regression. Eur Arch Psychiatry Clin Neurosci. 2018;268(2):119–27.PubMedCrossRef
389.
go back to reference Komaki Y, Sugiura H, Koarai A, Tomaki M, Ogawa H, Akita T, Hattori T, Ichinose M. Cytokine-mediated xanthine oxidase upregulation in chronic obstructive pulmonary disease's airways. Pulm Pharmacol Ther. 2005;18(4):297–302.PubMedCrossRef Komaki Y, Sugiura H, Koarai A, Tomaki M, Ogawa H, Akita T, Hattori T, Ichinose M. Cytokine-mediated xanthine oxidase upregulation in chronic obstructive pulmonary disease's airways. Pulm Pharmacol Ther. 2005;18(4):297–302.PubMedCrossRef
390.
go back to reference Xuan J, Pan G, Qiu Y, Yang L, Su M, Liu Y, Chen J, Feng G, Fang Y, Jia W, et al. Metabolomic profiling to identify potential serum biomarkers for schizophrenia and risperidone action. J Proteome Res. 2011;10(12):5433–43.PubMedCrossRef Xuan J, Pan G, Qiu Y, Yang L, Su M, Liu Y, Chen J, Feng G, Fang Y, Jia W, et al. Metabolomic profiling to identify potential serum biomarkers for schizophrenia and risperidone action. J Proteome Res. 2011;10(12):5433–43.PubMedCrossRef
391.
go back to reference Gersch C, Palii SP, Imaram W, Kim KM, Karumanchi SA, Angerhofer A, Johnson RJ, Henderson GN. Reactions of peroxynitrite with uric acid: formation of reactive intermediates, alkylated products and triuret, and in vivo production of triuret under conditions of oxidative stress. Nucleosides Nucleotides Nucleic Acids. 2009;28(2):118–49.PubMedPubMedCentralCrossRef Gersch C, Palii SP, Imaram W, Kim KM, Karumanchi SA, Angerhofer A, Johnson RJ, Henderson GN. Reactions of peroxynitrite with uric acid: formation of reactive intermediates, alkylated products and triuret, and in vivo production of triuret under conditions of oxidative stress. Nucleosides Nucleotides Nucleic Acids. 2009;28(2):118–49.PubMedPubMedCentralCrossRef
393.
go back to reference Dietrich-Muszalska A, Malinowska J, Olas B, Głowacki R, Bald E, Wachowicz B, Rabe-Jabłońska J. The oxidative stress may be induced by the elevated homocysteine in schizophrenic patients. Neurochem Res. 2012;37(5):1057–62.PubMedPubMedCentralCrossRef Dietrich-Muszalska A, Malinowska J, Olas B, Głowacki R, Bald E, Wachowicz B, Rabe-Jabłońska J. The oxidative stress may be induced by the elevated homocysteine in schizophrenic patients. Neurochem Res. 2012;37(5):1057–62.PubMedPubMedCentralCrossRef
394.
go back to reference Lin KM, Lu CL, Hung KC, Wu PC, Pan CF, Wu CJ, Syu RS, Chen JS, Hsiao PJ, Lu KC. The paradoxical role of uric acid in osteoporosis. Nutrients. 2019;11(9). Lin KM, Lu CL, Hung KC, Wu PC, Pan CF, Wu CJ, Syu RS, Chen JS, Hsiao PJ, Lu KC. The paradoxical role of uric acid in osteoporosis. Nutrients. 2019;11(9).
395.
go back to reference Tchalla AE, Wellenius GA, Sorond FA, Travison TG, Dantoine T, Lipsitz LA. Elevated circulating vascular cell adhesion molecule-1 (sVCAM-1) is associated with concurrent depressive symptoms and cerebral white matter hyperintensities in older adults. BMC Geriatr. 2015;15(1). Tchalla AE, Wellenius GA, Sorond FA, Travison TG, Dantoine T, Lipsitz LA. Elevated circulating vascular cell adhesion molecule-1 (sVCAM-1) is associated with concurrent depressive symptoms and cerebral white matter hyperintensities in older adults. BMC Geriatr. 2015;15(1).
396.
go back to reference Müller N. The role of intercellular adhesion molecule-1 in the pathogenesis of psychiatric disorders. Front Pharmacol. 2019;10. Müller N. The role of intercellular adhesion molecule-1 in the pathogenesis of psychiatric disorders. Front Pharmacol. 2019;10.
397.
go back to reference Lopez-Vilchez I, Diaz-Ricart M, Navarro V, Torramade S, Zamorano-Leon J, Lopez-Farre A, Galan AM, Gasto C, Escolar G. Endothelial damage in major depression patients is modulated by SSRI treatment, as demonstrated by circulating biomarkers and an in vitro cell model. Transl Psychiatry. 2016;6(9):e886.PubMedPubMedCentralCrossRef Lopez-Vilchez I, Diaz-Ricart M, Navarro V, Torramade S, Zamorano-Leon J, Lopez-Farre A, Galan AM, Gasto C, Escolar G. Endothelial damage in major depression patients is modulated by SSRI treatment, as demonstrated by circulating biomarkers and an in vitro cell model. Transl Psychiatry. 2016;6(9):e886.PubMedPubMedCentralCrossRef
398.
go back to reference Poljask B, Milisav I. NAD+ as the link between oxidative stress, inflammation, caloric restriction, exercise, DNA repair, longevity, and health span. Rejuvenation Res 2016;19(5):406–413. Poljask B, Milisav I. NAD+ as the link between oxidative stress, inflammation, caloric restriction, exercise, DNA repair, longevity, and health span. Rejuvenation Res 2016;19(5):406–413.
399.
go back to reference Bot M, Chan MK, Jansen R, Lamers F, Vogelzangs N, Steiner J, Leweke FM, Rothermundt M, Cooper J, Bahn S, et al. Serum proteomic profiling of major depressive disorder. Transl Psychiatry. 2015;5(7):e599.PubMedPubMedCentralCrossRef Bot M, Chan MK, Jansen R, Lamers F, Vogelzangs N, Steiner J, Leweke FM, Rothermundt M, Cooper J, Bahn S, et al. Serum proteomic profiling of major depressive disorder. Transl Psychiatry. 2015;5(7):e599.PubMedPubMedCentralCrossRef
400.
go back to reference Kalkman HO. The association between vascular inflammation and depressive disorder. Causality, biomarkers and targeted treatment. Pharmaceuticals (Basel). 2020;13(5):92.CrossRef Kalkman HO. The association between vascular inflammation and depressive disorder. Causality, biomarkers and targeted treatment. Pharmaceuticals (Basel). 2020;13(5):92.CrossRef
401.
go back to reference van Sloten TT, Schram MT, Adriaanse MC, Dekker JM, Nijpels G, Teerlink T, Scheffer PG, Pouwer F, Schalkwijk CG, Stehouwer CD, et al. Endothelial dysfunction is associated with a greater depressive symptom score in a general elderly population: the Hoorn Study. Psychol Med. 2014;44(7):1403–16.PubMedCrossRef van Sloten TT, Schram MT, Adriaanse MC, Dekker JM, Nijpels G, Teerlink T, Scheffer PG, Pouwer F, Schalkwijk CG, Stehouwer CD, et al. Endothelial dysfunction is associated with a greater depressive symptom score in a general elderly population: the Hoorn Study. Psychol Med. 2014;44(7):1403–16.PubMedCrossRef
402.
go back to reference Peng L, Bi S, Liu X, Long T, Zhao Y, Li F, Yang T, Zhang C. Association between depressive symptoms and arterial stiffness: a cross-sectional study in the general Chinese population. BMJ Open. 2020;10(2):e033408.PubMedPubMedCentralCrossRef Peng L, Bi S, Liu X, Long T, Zhao Y, Li F, Yang T, Zhang C. Association between depressive symptoms and arterial stiffness: a cross-sectional study in the general Chinese population. BMJ Open. 2020;10(2):e033408.PubMedPubMedCentralCrossRef
403.
go back to reference Palta P, Samuel LJ, Miller ERI, Szanton SL. Depression and oxidative stress: results from a meta-analysis of observational studies. Psychosom Med. 2014;76(1):12–9.PubMedCrossRef Palta P, Samuel LJ, Miller ERI, Szanton SL. Depression and oxidative stress: results from a meta-analysis of observational studies. Psychosom Med. 2014;76(1):12–9.PubMedCrossRef
404.
go back to reference Greaney JL, Saunders EFH, Santhanam L, Alexander LM. Oxidative stress contributes to microvascular endothelial dysfunction in men and women with major depressive disorder. Circ Res. 2019;124(4):564–74.PubMedPubMedCentralCrossRef Greaney JL, Saunders EFH, Santhanam L, Alexander LM. Oxidative stress contributes to microvascular endothelial dysfunction in men and women with major depressive disorder. Circ Res. 2019;124(4):564–74.PubMedPubMedCentralCrossRef
405.
go back to reference Tonhajzerova I, Sekaninova N, Bona Olexova L, Visnovcova Z. Novel insight into neuroimmune regulatory mechanisms and biomarkers linking major depression and vascular diseases: the dilemma continues. Int J Mol Sci. 2020;21(7):2317.PubMedCentralCrossRef Tonhajzerova I, Sekaninova N, Bona Olexova L, Visnovcova Z. Novel insight into neuroimmune regulatory mechanisms and biomarkers linking major depression and vascular diseases: the dilemma continues. Int J Mol Sci. 2020;21(7):2317.PubMedCentralCrossRef
406.
go back to reference Brietzke E, Teixeira AL. Similar immune profile in bipolar disorder and schizophrenia: selective increase in soluble tumor necrosis factor receptor I and von Willebrand factor. Bipolar Disord. 2010;12(4):453–4.PubMedCrossRef Brietzke E, Teixeira AL. Similar immune profile in bipolar disorder and schizophrenia: selective increase in soluble tumor necrosis factor receptor I and von Willebrand factor. Bipolar Disord. 2010;12(4):453–4.PubMedCrossRef
407.
go back to reference Reininghaus EZ, Lackner N, Birner A, Bengesser S, Fellendorf FT, Platzer M, Rieger A, Queissner R, Kainzbauer N, Reininghaus B, et al. Extracellular matrix proteins matrix metallopeptidase 9 (MMP9) and soluble intercellular adhesion molecule 1 (sICAM-1) and correlations with clinical staging in euthymic bipolar disorder. Bipolar Disord. 2016;18(2):155–63.PubMedCrossRef Reininghaus EZ, Lackner N, Birner A, Bengesser S, Fellendorf FT, Platzer M, Rieger A, Queissner R, Kainzbauer N, Reininghaus B, et al. Extracellular matrix proteins matrix metallopeptidase 9 (MMP9) and soluble intercellular adhesion molecule 1 (sICAM-1) and correlations with clinical staging in euthymic bipolar disorder. Bipolar Disord. 2016;18(2):155–63.PubMedCrossRef
408.
go back to reference Schaefer M, Sarkar S, Schwarz M, Friebe A. Soluble intracellular adhesion molecule-1 in patients with unipolar or bipolar affective disorders: results from a pilot trial. Neuropsychobiology. 2016;74(1):8–14.PubMedCrossRef Schaefer M, Sarkar S, Schwarz M, Friebe A. Soluble intracellular adhesion molecule-1 in patients with unipolar or bipolar affective disorders: results from a pilot trial. Neuropsychobiology. 2016;74(1):8–14.PubMedCrossRef
411.
go back to reference Cai HQ, Catts VS, Webster MJ, Galletly C, Liu D, O’Donnell M, Weickert TW, Weickert CS. Increased macrophages and changed brain endothelial cell gene expression in the frontal cortex of people with schizophrenia displaying inflammation. Mol Psychiatry. 2020;25(4):761–75.PubMedCrossRef Cai HQ, Catts VS, Webster MJ, Galletly C, Liu D, O’Donnell M, Weickert TW, Weickert CS. Increased macrophages and changed brain endothelial cell gene expression in the frontal cortex of people with schizophrenia displaying inflammation. Mol Psychiatry. 2020;25(4):761–75.PubMedCrossRef
412.
go back to reference Dieset I, Haukvik UK, Melle I, Røssberg JI, Ueland T, Hope S, Dale AM, Djurovic S, Aukrust P, Agartz I, et al. Association between altered brain morphology and elevated peripheral endothelial markers--implications for psychotic disorders. Schizophr Res. 2015;161(2–3):222–8.PubMedCrossRef Dieset I, Haukvik UK, Melle I, Røssberg JI, Ueland T, Hope S, Dale AM, Djurovic S, Aukrust P, Agartz I, et al. Association between altered brain morphology and elevated peripheral endothelial markers--implications for psychotic disorders. Schizophr Res. 2015;161(2–3):222–8.PubMedCrossRef
413.
go back to reference Hope S, Ueland T, Steen NE, Dieset I, Lorentzen S, Berg AO, Agartz I, Aukrust P, Andreassen OA. Interleukin 1 receptor antagonist and soluble tumor necrosis factor receptor 1 are associated with general severity and psychotic symptoms in schizophrenia and bipolar disorder. Schizophr Res. 2013;145(1):36–42.PubMedCrossRef Hope S, Ueland T, Steen NE, Dieset I, Lorentzen S, Berg AO, Agartz I, Aukrust P, Andreassen OA. Interleukin 1 receptor antagonist and soluble tumor necrosis factor receptor 1 are associated with general severity and psychotic symptoms in schizophrenia and bipolar disorder. Schizophr Res. 2013;145(1):36–42.PubMedCrossRef
414.
go back to reference Cristiano VB, Vieira Szortyka MF, Lobato MI, Ceresér KM, Belmonte-de-Abreu P. Postural changes in different stages of schizophrenia is associated with inflammation and pain: a cross-sectional observational study. Int J Psychiatry Clin Pract. 2017;21(2):104–11.PubMedCrossRef Cristiano VB, Vieira Szortyka MF, Lobato MI, Ceresér KM, Belmonte-de-Abreu P. Postural changes in different stages of schizophrenia is associated with inflammation and pain: a cross-sectional observational study. Int J Psychiatry Clin Pract. 2017;21(2):104–11.PubMedCrossRef
415.
go back to reference Schwarz MJ, Riedel M, Ackenheil M, Müller N. Decreased levels of soluble intercellular adhesion molecule-1 (sICAM-1) in unmedicated and medicated schizophrenic patients. Biol Psychiatry. 2000;47(1):29–33.PubMedCrossRef Schwarz MJ, Riedel M, Ackenheil M, Müller N. Decreased levels of soluble intercellular adhesion molecule-1 (sICAM-1) in unmedicated and medicated schizophrenic patients. Biol Psychiatry. 2000;47(1):29–33.PubMedCrossRef
416.
go back to reference Shcherbakova IV, Kaleda VG, Barkhatova AN, Kliushnik TP. Markers of endothelial dysfunction in attack-like schizophrenia. Zhurnal nevrologii i psikhiatrii imeni SS Korsakova. 2005;105(3):43–6. Shcherbakova IV, Kaleda VG, Barkhatova AN, Kliushnik TP. Markers of endothelial dysfunction in attack-like schizophrenia. Zhurnal nevrologii i psikhiatrii imeni SS Korsakova. 2005;105(3):43–6.
417.
go back to reference Kimmel SE, Schelleman H, Berlin JA, Oslin DW, Weinstein RB, Kinman JL, Sauer WH, Lewis JD. The effect of selective serotonin re-uptake inhibitors on the risk of myocardial infarction in a cohort of patients with depression. Br J Clin Pharmacol. 2011;72(3):514–7.PubMedPubMedCentralCrossRef Kimmel SE, Schelleman H, Berlin JA, Oslin DW, Weinstein RB, Kinman JL, Sauer WH, Lewis JD. The effect of selective serotonin re-uptake inhibitors on the risk of myocardial infarction in a cohort of patients with depression. Br J Clin Pharmacol. 2011;72(3):514–7.PubMedPubMedCentralCrossRef
418.
go back to reference Santangelo A, Testaì M, Barbagallo P, Crisafulli C, Grasso S, Manuele S, Muscarà G, Rizzotto M, Tomarchio M, Maugeri D. Use of specific serotonin reuptake inhibitors (SSRIs) (sertraline or citalopram) in the treatment of depression reduces the cardiovascular risk in the elderly: evidence from a Sicilian population >80 years recovered in the assisted sanitary residences (RSA). Arch Gerontol Geriatr. 2009;48(3):350–2.PubMedCrossRef Santangelo A, Testaì M, Barbagallo P, Crisafulli C, Grasso S, Manuele S, Muscarà G, Rizzotto M, Tomarchio M, Maugeri D. Use of specific serotonin reuptake inhibitors (SSRIs) (sertraline or citalopram) in the treatment of depression reduces the cardiovascular risk in the elderly: evidence from a Sicilian population >80 years recovered in the assisted sanitary residences (RSA). Arch Gerontol Geriatr. 2009;48(3):350–2.PubMedCrossRef
419.
go back to reference Tseng YL, Chiang ML, Huang TF, Su KP, Lane HY, Lai YC. A selective serotonin reuptake inhibitor, citalopram, inhibits collagen-induced platelet aggregation and activation. Thromb Res. 2010;126(6):517–23.PubMedCrossRef Tseng YL, Chiang ML, Huang TF, Su KP, Lane HY, Lai YC. A selective serotonin reuptake inhibitor, citalopram, inhibits collagen-induced platelet aggregation and activation. Thromb Res. 2010;126(6):517–23.PubMedCrossRef
420.
go back to reference McCloskey DJ, Postolache TT, Vittone BJ, Nghiem KL, Monsale JL, Wesley RA, Rick ME. Selective serotonin reuptake inhibitors: measurement of effect on platelet function. Transl Res. 2008;151(3):168–72.PubMedCrossRef McCloskey DJ, Postolache TT, Vittone BJ, Nghiem KL, Monsale JL, Wesley RA, Rick ME. Selective serotonin reuptake inhibitors: measurement of effect on platelet function. Transl Res. 2008;151(3):168–72.PubMedCrossRef
421.
go back to reference van Zyl LT, Lespérance F, Frasure-Smith N, Malinin AI, Atar D, Laliberté MA, Serebruany VL. Platelet and endothelial activity in comorbid major depression and coronary artery disease patients treated with citalopram: the Canadian Cardiac Randomized Evaluation of Antidepressant and Psychotherapy Efficacy Trial (CREATE) biomarker sub-study. J Thromb Thrombolysis. 2009;27(1):48–56.PubMedCrossRef van Zyl LT, Lespérance F, Frasure-Smith N, Malinin AI, Atar D, Laliberté MA, Serebruany VL. Platelet and endothelial activity in comorbid major depression and coronary artery disease patients treated with citalopram: the Canadian Cardiac Randomized Evaluation of Antidepressant and Psychotherapy Efficacy Trial (CREATE) biomarker sub-study. J Thromb Thrombolysis. 2009;27(1):48–56.PubMedCrossRef
422.
go back to reference Pizzi C, Mancini S, Angeloni L, Fontana F, Manzoli L, Costa GM. Effects of selective serotonin reuptake inhibitor therapy on endothelial function and inflammatory markers in patients with coronary heart disease. Clin Pharmacol Ther. 2009;86(5):527–32.PubMedCrossRef Pizzi C, Mancini S, Angeloni L, Fontana F, Manzoli L, Costa GM. Effects of selective serotonin reuptake inhibitor therapy on endothelial function and inflammatory markers in patients with coronary heart disease. Clin Pharmacol Ther. 2009;86(5):527–32.PubMedCrossRef
423.
go back to reference Kokras N, Papadopoulou E, Georgiopoulos G, Dalla C, Petropoulos I, Kontogiannis C, Laina A, Bampatsias D, Stellos K, Kouzoupis AV, et al. The effect of treatment response on endothelial function and arterial stiffness in depression. A prospective study. J Affect Disord. 2019;252:190–200.PubMedCrossRef Kokras N, Papadopoulou E, Georgiopoulos G, Dalla C, Petropoulos I, Kontogiannis C, Laina A, Bampatsias D, Stellos K, Kouzoupis AV, et al. The effect of treatment response on endothelial function and arterial stiffness in depression. A prospective study. J Affect Disord. 2019;252:190–200.PubMedCrossRef
424.
go back to reference Hantsoo L, Czarkowski KA, Child J, Howes C, Epperson CN. Selective serotonin reuptake inhibitors and endothelial function in women. J Women's Health (Larchmt). 2014;23(7):613–8.CrossRef Hantsoo L, Czarkowski KA, Child J, Howes C, Epperson CN. Selective serotonin reuptake inhibitors and endothelial function in women. J Women's Health (Larchmt). 2014;23(7):613–8.CrossRef
425.
go back to reference Mohammadianinejad SE, Majdinasab N, Sajedi SA, Abdollahi F, Moqaddam MM, Sadr F. The effect of lithium in post-stroke motor recovery: a double-blind, placebo-controlled, randomized clinical trial. Clin Neuropharmacol. 2014;37(3):73–8.PubMedCrossRef Mohammadianinejad SE, Majdinasab N, Sajedi SA, Abdollahi F, Moqaddam MM, Sadr F. The effect of lithium in post-stroke motor recovery: a double-blind, placebo-controlled, randomized clinical trial. Clin Neuropharmacol. 2014;37(3):73–8.PubMedCrossRef
426.
go back to reference Lyoo IK, Dager SR, Kim JE, Yoon SJ, Friedman SD, Dunner DL, Renshaw PF. Lithium-induced gray matter volume increase as a neural correlate of treatment response in bipolar disorder: a longitudinal brain imaging study. Neuropsychopharmacology. 2010;35(8):1743–50.PubMedPubMedCentralCrossRef Lyoo IK, Dager SR, Kim JE, Yoon SJ, Friedman SD, Dunner DL, Renshaw PF. Lithium-induced gray matter volume increase as a neural correlate of treatment response in bipolar disorder: a longitudinal brain imaging study. Neuropsychopharmacology. 2010;35(8):1743–50.PubMedPubMedCentralCrossRef
427.
go back to reference Li W, Li R, Zhao S, Jiang C, Liu Z, Tang X. Lithium posttreatment alleviates blood–brain barrier injury after intracerebral hemorrhage in rats. Neuroscience. 2018;383:129–37.PubMedCrossRef Li W, Li R, Zhao S, Jiang C, Liu Z, Tang X. Lithium posttreatment alleviates blood–brain barrier injury after intracerebral hemorrhage in rats. Neuroscience. 2018;383:129–37.PubMedCrossRef
428.
go back to reference Wang Z-f, Fessler EB, Chuang D-M. Beneficial effects of mood stabilizers lithium, valproate and lamotrigine in experimental stroke models. Acta Pharmacol Sin. 2011;32(12):1433–45.PubMedPubMedCentralCrossRef Wang Z-f, Fessler EB, Chuang D-M. Beneficial effects of mood stabilizers lithium, valproate and lamotrigine in experimental stroke models. Acta Pharmacol Sin. 2011;32(12):1433–45.PubMedPubMedCentralCrossRef
429.
go back to reference Murugavel S, Bugyei-Twum A, Matkar PN, Al-Mubarak H, Chen HH, Adam M, Jain S, Narang T, Abdin RM, Qadura M et al: Valproic acid induces endothelial-to-mesenchymal transition-like phenotypic switching. Front Pharmacol 2018, 9(737). Murugavel S, Bugyei-Twum A, Matkar PN, Al-Mubarak H, Chen HH, Adam M, Jain S, Narang T, Abdin RM, Qadura M et al: Valproic acid induces endothelial-to-mesenchymal transition-like phenotypic switching. Front Pharmacol 2018, 9(737).
430.
go back to reference Ellingrod VL, Taylor SF, Brook RD, Evans SJ, Zöllner SK, Grove TB, Gardner KM, Bly MJ, Pop-Busui R, Dalack G. Dietary, lifestyle and pharmacogenetic factors associated with arteriole endothelial-dependent vasodilatation in schizophrenia patients treated with atypical antipsychotics (AAPs). Schizophr Res. 2011;130(1–3):20–6.PubMedPubMedCentralCrossRef Ellingrod VL, Taylor SF, Brook RD, Evans SJ, Zöllner SK, Grove TB, Gardner KM, Bly MJ, Pop-Busui R, Dalack G. Dietary, lifestyle and pharmacogenetic factors associated with arteriole endothelial-dependent vasodilatation in schizophrenia patients treated with atypical antipsychotics (AAPs). Schizophr Res. 2011;130(1–3):20–6.PubMedPubMedCentralCrossRef
431.
go back to reference Burghardt K, Grove T, Ellingrod V. Endothelial nitric oxide synthetase genetic variants, metabolic syndrome and endothelial function in schizophrenia. J Psychopharmacol. 2014;28(4):349–56.PubMedCrossRef Burghardt K, Grove T, Ellingrod V. Endothelial nitric oxide synthetase genetic variants, metabolic syndrome and endothelial function in schizophrenia. J Psychopharmacol. 2014;28(4):349–56.PubMedCrossRef
432.
go back to reference Xu H, Zhuang X. Atypical antipsychotics-induced metabolic syndrome and nonalcoholic fatty liver disease: a critical review. Neuropsychiatr Dis Treat. 2019;15:2087–99.PubMedPubMedCentralCrossRef Xu H, Zhuang X. Atypical antipsychotics-induced metabolic syndrome and nonalcoholic fatty liver disease: a critical review. Neuropsychiatr Dis Treat. 2019;15:2087–99.PubMedPubMedCentralCrossRef
433.
go back to reference Kavzoglu S, Hariri A. Intracellular adhesion molecule (ICAM-1), vascular cell adhesion molecule (VCAM-1) and E-selectin levels in first episode schizophrenic patients. Bull Clin Psychopharmacol. 2013;23:1.CrossRef Kavzoglu S, Hariri A. Intracellular adhesion molecule (ICAM-1), vascular cell adhesion molecule (VCAM-1) and E-selectin levels in first episode schizophrenic patients. Bull Clin Psychopharmacol. 2013;23:1.CrossRef
434.
go back to reference Clark AM, DesMeules M, Luo W, Duncan AS, Wielgosz A. Socioeconomic status and cardiovascular disease: risks and implications for care. Nat Rev Cardiol. 2009;6(11):712–22.PubMedCrossRef Clark AM, DesMeules M, Luo W, Duncan AS, Wielgosz A. Socioeconomic status and cardiovascular disease: risks and implications for care. Nat Rev Cardiol. 2009;6(11):712–22.PubMedCrossRef
435.
go back to reference Cohen S, Janicki-Deverts D, Chen E, Matthews KA. Childhood socioeconomic status and adult health. Ann N Y Acad Sci. 2010;1186:37–55.PubMedCrossRef Cohen S, Janicki-Deverts D, Chen E, Matthews KA. Childhood socioeconomic status and adult health. Ann N Y Acad Sci. 2010;1186:37–55.PubMedCrossRef
436.
go back to reference Schultz WM, Kelli HM, Lisko JC, Varghese T, Shen J, Sandesara P, Quyyumi AA, Taylor HA, Gulati M, Harold JG, et al. Socioeconomic status and cardiovascular outcomes. Circulation. 2018;137(20):2166–78.PubMedPubMedCentralCrossRef Schultz WM, Kelli HM, Lisko JC, Varghese T, Shen J, Sandesara P, Quyyumi AA, Taylor HA, Gulati M, Harold JG, et al. Socioeconomic status and cardiovascular outcomes. Circulation. 2018;137(20):2166–78.PubMedPubMedCentralCrossRef
437.
go back to reference East P, Doom J, Delker E, Blanco E, Burrows R, Correa-Burrows P, Lozoff B, Gahagan S. Childhood socioeconomic hardship, family conflict, and young adult hypertension: the Santiago Longitudinal Study. Soc Sci Med. 2020;253:112962.PubMedCrossRefPubMedCentral East P, Doom J, Delker E, Blanco E, Burrows R, Correa-Burrows P, Lozoff B, Gahagan S. Childhood socioeconomic hardship, family conflict, and young adult hypertension: the Santiago Longitudinal Study. Soc Sci Med. 2020;253:112962.PubMedCrossRefPubMedCentral
438.
go back to reference Reid BM, Doom JR, Argote RB, Correa-Burrows P, Lozoff B, Blanco E, Gahagan S. Pathways to inflammation in adolescence through early adversity, childhood depressive symptoms, and body mass index: a prospective longitudinal study of Chilean infants. Brain Behav Immun. 2020;86:4–13.PubMedCrossRef Reid BM, Doom JR, Argote RB, Correa-Burrows P, Lozoff B, Blanco E, Gahagan S. Pathways to inflammation in adolescence through early adversity, childhood depressive symptoms, and body mass index: a prospective longitudinal study of Chilean infants. Brain Behav Immun. 2020;86:4–13.PubMedCrossRef
439.
go back to reference Kivimäki M, Vahtera J, Tabák AG, Halonen JI, Vineis P, Pentti J, Pahkala K, Rovio S, Viikari J, Kähönen M, et al. Neighbourhood socioeconomic disadvantage, risk factors, and diabetes from childhood to middle age in the Young Finns Study: a cohort study. Lancet Public Health. 2018;3(8):e365–73.PubMedPubMedCentralCrossRef Kivimäki M, Vahtera J, Tabák AG, Halonen JI, Vineis P, Pentti J, Pahkala K, Rovio S, Viikari J, Kähönen M, et al. Neighbourhood socioeconomic disadvantage, risk factors, and diabetes from childhood to middle age in the Young Finns Study: a cohort study. Lancet Public Health. 2018;3(8):e365–73.PubMedPubMedCentralCrossRef
440.
go back to reference Lee SC, DelPozo-Banos M, Lloyd K, Jones I, Walters JTR, Owen MJ, O'Donovan M, John A. Area deprivation, urbanicity, severe mental illness and social drift — a population-based linkage study using routinely collected primary and secondary care data. Schizophr Res. 2020;220:130–40.PubMedCrossRef Lee SC, DelPozo-Banos M, Lloyd K, Jones I, Walters JTR, Owen MJ, O'Donovan M, John A. Area deprivation, urbanicity, severe mental illness and social drift — a population-based linkage study using routinely collected primary and secondary care data. Schizophr Res. 2020;220:130–40.PubMedCrossRef
441.
go back to reference Martin JL, McLean G, Park J, Martin DJ, Connolly M, Mercer SW, Smith DJ. Impact of socioeconomic deprivation on rate and cause of death in severe mental illness. BMC Psychiatry. 2014;14(1):261.PubMedPubMedCentralCrossRef Martin JL, McLean G, Park J, Martin DJ, Connolly M, Mercer SW, Smith DJ. Impact of socioeconomic deprivation on rate and cause of death in severe mental illness. BMC Psychiatry. 2014;14(1):261.PubMedPubMedCentralCrossRef
442.
go back to reference Diniz BS, Reynolds Iii CF, Sibille E, Bot M, Penninx BWJH. Major depression and enhanced molecular senescence abnormalities in young and middle-aged adults. Transl Psychiatry. 2019;9(1):198.PubMedPubMedCentralCrossRef Diniz BS, Reynolds Iii CF, Sibille E, Bot M, Penninx BWJH. Major depression and enhanced molecular senescence abnormalities in young and middle-aged adults. Transl Psychiatry. 2019;9(1):198.PubMedPubMedCentralCrossRef
443.
445.
go back to reference Kaptoge S, Di Angelantonio E, Pennells L, Wood AM, White IR, Gao P, Walker M, Thompson A, Sarwar N, Caslake M, et al. C-reactive protein, fibrinogen, and cardiovascular disease prediction. N Engl J Med. 2012;367(14):1310–20.PubMedCrossRef Kaptoge S, Di Angelantonio E, Pennells L, Wood AM, White IR, Gao P, Walker M, Thompson A, Sarwar N, Caslake M, et al. C-reactive protein, fibrinogen, and cardiovascular disease prediction. N Engl J Med. 2012;367(14):1310–20.PubMedCrossRef
446.
go back to reference Esser N, Legrand-Poels S, Piette J, Scheen AJ, Paquot N. Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res Clin Pract. 2014;105(2):141–50.PubMedCrossRef Esser N, Legrand-Poels S, Piette J, Scheen AJ, Paquot N. Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res Clin Pract. 2014;105(2):141–50.PubMedCrossRef
447.
go back to reference Fung TT, McCullough ML, Newby PK, Manson JE, Meigs JB, Rifai N, Willett WC, Hu FB. Diet-quality scores and plasma concentrations of markers of inflammation and endothelial dysfunction. Am J Clin Nutr. 2005;82(1):163–73.PubMedCrossRef Fung TT, McCullough ML, Newby PK, Manson JE, Meigs JB, Rifai N, Willett WC, Hu FB. Diet-quality scores and plasma concentrations of markers of inflammation and endothelial dysfunction. Am J Clin Nutr. 2005;82(1):163–73.PubMedCrossRef
448.
go back to reference Akbaraly TN, Shipley MJ, Ferrie JE, Virtanen M, Lowe G, Hamer M, Kivimaki M. Long-term adherence to healthy dietary guidelines and chronic inflammation in the prospective Whitehall II study. Am J Med. 2015;128(2):152–160.e154.PubMedPubMedCentralCrossRef Akbaraly TN, Shipley MJ, Ferrie JE, Virtanen M, Lowe G, Hamer M, Kivimaki M. Long-term adherence to healthy dietary guidelines and chronic inflammation in the prospective Whitehall II study. Am J Med. 2015;128(2):152–160.e154.PubMedPubMedCentralCrossRef
449.
go back to reference Hamer M, Sabia S, Batty GD, Shipley MJ, Tabák AG, Singh-Manoux A, Kivimaki M. Physical activity and inflammatory markers over 10 years: follow-up in men and women from the Whitehall II cohort study. Circulation. 2012;126(8):928–33.PubMedPubMedCentralCrossRef Hamer M, Sabia S, Batty GD, Shipley MJ, Tabák AG, Singh-Manoux A, Kivimaki M. Physical activity and inflammatory markers over 10 years: follow-up in men and women from the Whitehall II cohort study. Circulation. 2012;126(8):928–33.PubMedPubMedCentralCrossRef
450.
go back to reference Jarvie JL, Whooley MA, Regan MC, Sin NL, Cohen BE. Effect of physical activity level on biomarkers of inflammation and insulin resistance over 5 years in outpatients with coronary heart disease (from the Heart and Soul Study). Am J Cardiol. 2014;114(8):1192–7.PubMedPubMedCentralCrossRef Jarvie JL, Whooley MA, Regan MC, Sin NL, Cohen BE. Effect of physical activity level on biomarkers of inflammation and insulin resistance over 5 years in outpatients with coronary heart disease (from the Heart and Soul Study). Am J Cardiol. 2014;114(8):1192–7.PubMedPubMedCentralCrossRef
451.
go back to reference Motivala SJ. Sleep and inflammation: psychoneuroimmunology in the context of cardiovascular disease. Ann Behav Med. 2011;42(2):141–52.PubMedCrossRef Motivala SJ. Sleep and inflammation: psychoneuroimmunology in the context of cardiovascular disease. Ann Behav Med. 2011;42(2):141–52.PubMedCrossRef
452.
go back to reference Qin B, Deng Y. Overexpression of circadian clock protein cryptochrome (CRY) 1 alleviates sleep deprivation-induced vascular inflammation in a mouse model. Immunol Lett. 2015;163(1):76–83.PubMedCrossRef Qin B, Deng Y. Overexpression of circadian clock protein cryptochrome (CRY) 1 alleviates sleep deprivation-induced vascular inflammation in a mouse model. Immunol Lett. 2015;163(1):76–83.PubMedCrossRef
453.
go back to reference Shiels MS, Katki HA, Freedman ND, Purdue MP, Wentzensen N, Trabert B, Kitahara CM, Furr M, Li Y, Kemp TJ, et al. Cigarette smoking and variations in systemic immune and inflammation markers. J Natl Cancer Inst. 2014;106(11):dju294.PubMedPubMedCentralCrossRef Shiels MS, Katki HA, Freedman ND, Purdue MP, Wentzensen N, Trabert B, Kitahara CM, Furr M, Li Y, Kemp TJ, et al. Cigarette smoking and variations in systemic immune and inflammation markers. J Natl Cancer Inst. 2014;106(11):dju294.PubMedPubMedCentralCrossRef
454.
go back to reference Asthana A, Johnson HM, Piper ME, Fiore MC, Baker TB, Stein JH. Effects of smoking intensity and cessation on inflammatory markers in a large cohort of active smokers. Am Heart J. 2010;160(3):458.PubMedPubMedCentralCrossRef Asthana A, Johnson HM, Piper ME, Fiore MC, Baker TB, Stein JH. Effects of smoking intensity and cessation on inflammatory markers in a large cohort of active smokers. Am Heart J. 2010;160(3):458.PubMedPubMedCentralCrossRef
455.
go back to reference Leclercq S, de Timary P, Delzenne NM, Stärkel P. The link between inflammation, bugs, the intestine and the brain in alcohol dependence. Transl Psychiatry. 2017;7(2):e1048.PubMedPubMedCentralCrossRef Leclercq S, de Timary P, Delzenne NM, Stärkel P. The link between inflammation, bugs, the intestine and the brain in alcohol dependence. Transl Psychiatry. 2017;7(2):e1048.PubMedPubMedCentralCrossRef
456.
go back to reference Schmitz S, Abosi O, Persons J, Sinkey C, Fiedorowicz J. Impact of mood on endothelial function and arterial stiffness in bipolar disorder. Heart Mind. 2018;2(3):78–84.PubMedCrossRef Schmitz S, Abosi O, Persons J, Sinkey C, Fiedorowicz J. Impact of mood on endothelial function and arterial stiffness in bipolar disorder. Heart Mind. 2018;2(3):78–84.PubMedCrossRef
457.
go back to reference Shi H, Feng G, Wang Z, Zhou C, Zhong G, Hu Y, Wang G. Relationships between depressive symptoms and endothelial function among outpatients of a general hospital in China. Med Sci Monit. 2015;21:1812–9.PubMedPubMedCentralCrossRef Shi H, Feng G, Wang Z, Zhou C, Zhong G, Hu Y, Wang G. Relationships between depressive symptoms and endothelial function among outpatients of a general hospital in China. Med Sci Monit. 2015;21:1812–9.PubMedPubMedCentralCrossRef
458.
go back to reference Velten J, Bieda A, Scholten S, Wannemüller A, Margraf J. Lifestyle choices and mental health: a longitudinal survey with German and Chinese students. BMC Public Health. 2018;18(1):632.PubMedPubMedCentralCrossRef Velten J, Bieda A, Scholten S, Wannemüller A, Margraf J. Lifestyle choices and mental health: a longitudinal survey with German and Chinese students. BMC Public Health. 2018;18(1):632.PubMedPubMedCentralCrossRef
459.
go back to reference Mintzer J, Donovan KA, Kindy AZ, Lock SL, Chura LR, Barracca N. Lifestyle choices and brain health. Front Med (Lausanne). 2019;6:204.CrossRef Mintzer J, Donovan KA, Kindy AZ, Lock SL, Chura LR, Barracca N. Lifestyle choices and brain health. Front Med (Lausanne). 2019;6:204.CrossRef
460.
go back to reference Morris G, Berk M, Maes M, Carvalho AF, Puri BK. Socioeconomic deprivation, adverse childhood experiences and medical disorders in adulthood: mechanisms and associations. Mol Neurobiol. 2019;56(8):5866–90.PubMedPubMedCentralCrossRef Morris G, Berk M, Maes M, Carvalho AF, Puri BK. Socioeconomic deprivation, adverse childhood experiences and medical disorders in adulthood: mechanisms and associations. Mol Neurobiol. 2019;56(8):5866–90.PubMedPubMedCentralCrossRef
461.
go back to reference Tang KL, Rashid R, Godley J, Ghali WA. Association between subjective social status and cardiovascular disease and cardiovascular risk factors: a systematic review and meta-analysis. BMJ Open. 2016;6(3):e010137.PubMedPubMedCentralCrossRef Tang KL, Rashid R, Godley J, Ghali WA. Association between subjective social status and cardiovascular disease and cardiovascular risk factors: a systematic review and meta-analysis. BMJ Open. 2016;6(3):e010137.PubMedPubMedCentralCrossRef
462.
go back to reference Cooper DC, Milic MS, Mills PJ, Bardwell WA, Ziegler MG, Dimsdale JE. Endothelial function: the impact of objective and subjective socioeconomic status on flow-mediated dilation. Ann Behav Med. 2010;39(3):222–31.PubMedCrossRef Cooper DC, Milic MS, Mills PJ, Bardwell WA, Ziegler MG, Dimsdale JE. Endothelial function: the impact of objective and subjective socioeconomic status on flow-mediated dilation. Ann Behav Med. 2010;39(3):222–31.PubMedCrossRef
463.
go back to reference Tang KL, Pilote L, Behlouli H, Godley J, Ghali WA. An exploration of the subjective social status construct in patients with acute coronary syndrome. BMC Cardiovasc Disord. 2018;18(1):22.PubMedPubMedCentralCrossRef Tang KL, Pilote L, Behlouli H, Godley J, Ghali WA. An exploration of the subjective social status construct in patients with acute coronary syndrome. BMC Cardiovasc Disord. 2018;18(1):22.PubMedPubMedCentralCrossRef
464.
465.
go back to reference Kim C-H, Noh I-K, Ryu JM, Bae EJ, Cho HJ, Kim MS. Canonical correlation between behavioral-psychological variables and predictors of coronary artery disease prognosis. Int J Environ Res Public Health. 2020;17(5):1608.PubMedCentralCrossRef Kim C-H, Noh I-K, Ryu JM, Bae EJ, Cho HJ, Kim MS. Canonical correlation between behavioral-psychological variables and predictors of coronary artery disease prognosis. Int J Environ Res Public Health. 2020;17(5):1608.PubMedCentralCrossRef
466.
go back to reference Bezgin CH, Bezgin T, Kesebir S. Temperament and character profiles and psychiatric comorbidities in patients with coronary artery or valvular heart disease: relationship with cardiac disease severity. J Clin Med Res. 2016;8(3):202–9.PubMedPubMedCentralCrossRef Bezgin CH, Bezgin T, Kesebir S. Temperament and character profiles and psychiatric comorbidities in patients with coronary artery or valvular heart disease: relationship with cardiac disease severity. J Clin Med Res. 2016;8(3):202–9.PubMedPubMedCentralCrossRef
467.
go back to reference Eory A, Gonda X, Lang Z, Torzsa P, Kalman J, Kalabay L, Rihmer Z. Personality and cardiovascular risk: association between hypertension and affective temperaments—a cross-sectional observational study in primary care settings. Eur J Gen Pract. 2014;20(4):247–52.PubMedCrossRef Eory A, Gonda X, Lang Z, Torzsa P, Kalman J, Kalabay L, Rihmer Z. Personality and cardiovascular risk: association between hypertension and affective temperaments—a cross-sectional observational study in primary care settings. Eur J Gen Pract. 2014;20(4):247–52.PubMedCrossRef
468.
go back to reference Korosi BZ, Batta D, Gonda X, Rihmer Z, Nemcsik-Bencze Z, László A, Vecsey-Nagy M, Nemcsik J. Association between irritable affective temperament and nighttime peripheral and central systolic blood pressure in hypertension. Artery Res. 2019;25. Korosi BZ, Batta D, Gonda X, Rihmer Z, Nemcsik-Bencze Z, László A, Vecsey-Nagy M, Nemcsik J. Association between irritable affective temperament and nighttime peripheral and central systolic blood pressure in hypertension. Artery Res. 2019;25.
469.
go back to reference László A, Tabák Á, Kőrösi B, Eörsi D, Torzsa P, Cseprekál O, Tislér A, Reusz G, Nemcsik-Bencze Z, Gonda X, et al. Association of affective temperaments with blood pressure and arterial stiffness in hypertensive patients: a cross-sectional study. BMC Cardiovasc Disord. 2016;16(1):158.PubMedPubMedCentralCrossRef László A, Tabák Á, Kőrösi B, Eörsi D, Torzsa P, Cseprekál O, Tislér A, Reusz G, Nemcsik-Bencze Z, Gonda X, et al. Association of affective temperaments with blood pressure and arterial stiffness in hypertensive patients: a cross-sectional study. BMC Cardiovasc Disord. 2016;16(1):158.PubMedPubMedCentralCrossRef
470.
471.
go back to reference Verhoeven F, Prati C, Maguin-Gaté K, Wendling D, Demougeot C. Glucocorticoids and endothelial function in inflammatory diseases: focus on rheumatoid arthritis. Arthritis Res Ther. 2016;18(1):258.PubMedPubMedCentralCrossRef Verhoeven F, Prati C, Maguin-Gaté K, Wendling D, Demougeot C. Glucocorticoids and endothelial function in inflammatory diseases: focus on rheumatoid arthritis. Arthritis Res Ther. 2016;18(1):258.PubMedPubMedCentralCrossRef
472.
go back to reference Zielińska KA, Van Moortel L, Opdenakker G, De Bosscher K, Van den Steen PE: Endothelial response to glucocorticoids in inflammatory diseases. Front Immunol 2016, 7(592). Zielińska KA, Van Moortel L, Opdenakker G, De Bosscher K, Van den Steen PE: Endothelial response to glucocorticoids in inflammatory diseases. Front Immunol 2016, 7(592).
473.
go back to reference Iuchi T, Akaike M, Mitsui T, Ohshima Y, Shintani Y, Azuma H, Matsumoto T. Glucocorticoid excess induces superoxide production in vascular endothelial cells and elicits vascular endothelial dysfunction. Circ Res. 2003;92(1):81–7.PubMedCrossRef Iuchi T, Akaike M, Mitsui T, Ohshima Y, Shintani Y, Azuma H, Matsumoto T. Glucocorticoid excess induces superoxide production in vascular endothelial cells and elicits vascular endothelial dysfunction. Circ Res. 2003;92(1):81–7.PubMedCrossRef
474.
go back to reference Brotman DJ, Girod JP, Garcia MJ, Patel JV, Gupta M, Posch A, Saunders S, Lip GYH, Worley S, Reddy S. Effects of short-term glucocorticoids on cardiovascular biomarkers. J Clin Endocrinol Metabol. 2005;90(6):3202–8.CrossRef Brotman DJ, Girod JP, Garcia MJ, Patel JV, Gupta M, Posch A, Saunders S, Lip GYH, Worley S, Reddy S. Effects of short-term glucocorticoids on cardiovascular biomarkers. J Clin Endocrinol Metabol. 2005;90(6):3202–8.CrossRef
475.
go back to reference Haapakoski R, Mathieu J, Ebmeier KP, Alenius H, Kivimäki M. Cumulative meta-analysis of interleukins 6 and 1β, tumour necrosis factor α and C-reactive protein in patients with major depressive disorder. Brain Behav Immun. 2015;49:206–15.PubMedPubMedCentralCrossRef Haapakoski R, Mathieu J, Ebmeier KP, Alenius H, Kivimäki M. Cumulative meta-analysis of interleukins 6 and 1β, tumour necrosis factor α and C-reactive protein in patients with major depressive disorder. Brain Behav Immun. 2015;49:206–15.PubMedPubMedCentralCrossRef
476.
go back to reference Howren MB, Lamkin DM, Suls J. Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosom Med. 2009;71(2):171–86.PubMedCrossRef Howren MB, Lamkin DM, Suls J. Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosom Med. 2009;71(2):171–86.PubMedCrossRef
477.
go back to reference Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK. A meta-analysis of cytokines in major depression. Biol Psychiatry. 2010;67. Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK. A meta-analysis of cytokines in major depression. Biol Psychiatry. 2010;67.
478.
go back to reference Liu Y, Ho RC-M, Mak A. Interleukin (IL)-6, tumour necrosis factor alpha (TNF-α) and soluble interleukin-2 receptors (sIL-2R) are elevated in patients with major depressive disorder: a meta-analysis and meta-regression. J Affect Disord. 2012;139(3):230–9.PubMedCrossRef Liu Y, Ho RC-M, Mak A. Interleukin (IL)-6, tumour necrosis factor alpha (TNF-α) and soluble interleukin-2 receptors (sIL-2R) are elevated in patients with major depressive disorder: a meta-analysis and meta-regression. J Affect Disord. 2012;139(3):230–9.PubMedCrossRef
479.
480.
go back to reference Silva B, Pernomian L, Bendhack L: Contribution of oxidative stress to endothelial dysfunction in hypertension. Front Physiol 2012, 3(441). Silva B, Pernomian L, Bendhack L: Contribution of oxidative stress to endothelial dysfunction in hypertension. Front Physiol 2012, 3(441).
482.
go back to reference van der Poll T, van de Veerdonk FL, Scicluna BP, Netea MG. The immunopathology of sepsis and potential therapeutic targets. Nat Rev Immunol. 2017;17(7):407–20.PubMedCrossRef van der Poll T, van de Veerdonk FL, Scicluna BP, Netea MG. The immunopathology of sepsis and potential therapeutic targets. Nat Rev Immunol. 2017;17(7):407–20.PubMedCrossRef
483.
go back to reference Szałach ŁP, Lisowska KA, Cubała WJ. The influence of antidepressants on the immune system. Arch Immunol Ther Exp. 2019;67(3):143–51.CrossRef Szałach ŁP, Lisowska KA, Cubała WJ. The influence of antidepressants on the immune system. Arch Immunol Ther Exp. 2019;67(3):143–51.CrossRef
484.
go back to reference Gałecki P, Mossakowska-Wójcik J, Talarowska M. The anti-inflammatory mechanism of antidepressants - SSRIs, SNRIs. Prog Neuro-Psychopharmacol Biol Psychiatry. 2018;80(Pt C):291–4.CrossRef Gałecki P, Mossakowska-Wójcik J, Talarowska M. The anti-inflammatory mechanism of antidepressants - SSRIs, SNRIs. Prog Neuro-Psychopharmacol Biol Psychiatry. 2018;80(Pt C):291–4.CrossRef
485.
go back to reference Kotan VO, Sarandol E, Kirhan E, Ozkaya G, Kirli S. Effects of long-term antidepressant treatment on oxidative status in major depressive disorder: a 24-week follow-up study. Prog Neuro-Psychopharmacol Biol Psychiatry. 2011;35. Kotan VO, Sarandol E, Kirhan E, Ozkaya G, Kirli S. Effects of long-term antidepressant treatment on oxidative status in major depressive disorder: a 24-week follow-up study. Prog Neuro-Psychopharmacol Biol Psychiatry. 2011;35.
486.
go back to reference Behr GA, Moreira JCF, Frey BN. Preclinical and clinical evidence of antioxidant effects of antidepressant agents: implications for the pathophysiology of major depressive disorder. Oxidative Med Cell Longev. 2012;2012. Behr GA, Moreira JCF, Frey BN. Preclinical and clinical evidence of antioxidant effects of antidepressant agents: implications for the pathophysiology of major depressive disorder. Oxidative Med Cell Longev. 2012;2012.
487.
go back to reference Morris G, Berk M. The putative use of lithium in Alzheimer’s disease. Curr Alzheimer Res. 2016;13(8):853–61.PubMedCrossRef Morris G, Berk M. The putative use of lithium in Alzheimer’s disease. Curr Alzheimer Res. 2016;13(8):853–61.PubMedCrossRef
488.
go back to reference Beumer W, Gibney SM, Drexhage RC, Pont-Lezica L, Doorduin J, Klein HC, Steiner J, Connor TJ, Harkin A, Versnel MA, et al. The immune theory of psychiatric diseases: a key role for activated microglia and circulating monocytes. J Leukoc Biol. 2012;92(5):959–75.PubMedCrossRef Beumer W, Gibney SM, Drexhage RC, Pont-Lezica L, Doorduin J, Klein HC, Steiner J, Connor TJ, Harkin A, Versnel MA, et al. The immune theory of psychiatric diseases: a key role for activated microglia and circulating monocytes. J Leukoc Biol. 2012;92(5):959–75.PubMedCrossRef
490.
go back to reference Ince C, Mayeux PR, Nguyen T, Gomez H, Kellum JA, Ospina-Tascon GA, Hernandez G, Murray P, De Backer D. The endothelium in sepsis. Shock. 2016;45(3):259–70.PubMedPubMedCentralCrossRef Ince C, Mayeux PR, Nguyen T, Gomez H, Kellum JA, Ospina-Tascon GA, Hernandez G, Murray P, De Backer D. The endothelium in sepsis. Shock. 2016;45(3):259–70.PubMedPubMedCentralCrossRef
491.
go back to reference Iba T, Levy JH. Derangement of the endothelial glycocalyx in sepsis. J Thromb Haemostasis. 2019;17(2):283–94.CrossRef Iba T, Levy JH. Derangement of the endothelial glycocalyx in sepsis. J Thromb Haemostasis. 2019;17(2):283–94.CrossRef
493.
go back to reference Steinhagen F, Schmidt SV, Schewe J-C, Peukert K, Klinman DM, Bode C. Immunotherapy in sepsis - brake or accelerate? Pharmacol Ther. 2020;208:107476.PubMedCrossRef Steinhagen F, Schmidt SV, Schewe J-C, Peukert K, Klinman DM, Bode C. Immunotherapy in sepsis - brake or accelerate? Pharmacol Ther. 2020;208:107476.PubMedCrossRef
494.
495.
go back to reference Genkel VV, Shaposhnik II. Conceptualization of heterogeneity of chronic diseases and atherosclerosis as a pathway to precision medicine: endophenotype, endotype, and residual cardiovascular risk. Int J Chronic Dis. 2020;2020:5950813.PubMedPubMedCentral Genkel VV, Shaposhnik II. Conceptualization of heterogeneity of chronic diseases and atherosclerosis as a pathway to precision medicine: endophenotype, endotype, and residual cardiovascular risk. Int J Chronic Dis. 2020;2020:5950813.PubMedPubMedCentral
496.
go back to reference Vardon-Bounes F, Ruiz S, Gratacap M-P, Garcia C, Payrastre B, Minville V. Platelets are critical key players in sepsis. Int J Mol Sci. 2019;20(14):3494.PubMedCentralCrossRef Vardon-Bounes F, Ruiz S, Gratacap M-P, Garcia C, Payrastre B, Minville V. Platelets are critical key players in sepsis. Int J Mol Sci. 2019;20(14):3494.PubMedCentralCrossRef
497.
go back to reference Li X, Wang P, Xu X, Wang Y, Xia Y, Wang D. Simvastatin increases the activity of endothelial nitric oxide synthase via enhancing phosphorylation. J Huazhong Univ Sci Technol Med Sci. 2009;29(3):286–90.CrossRef Li X, Wang P, Xu X, Wang Y, Xia Y, Wang D. Simvastatin increases the activity of endothelial nitric oxide synthase via enhancing phosphorylation. J Huazhong Univ Sci Technol Med Sci. 2009;29(3):286–90.CrossRef
498.
go back to reference Rossoni LV, Wareing M, Wenceslau CF, Al-Abri M, Cobb C, Austin C. Acute simvastatin increases endothelial nitric oxide synthase phosphorylation via AMP-activated protein kinase and reduces contractility of isolated rat mesenteric resistance arteries. Clin Sci (London). 2011;121(10):449–58.CrossRef Rossoni LV, Wareing M, Wenceslau CF, Al-Abri M, Cobb C, Austin C. Acute simvastatin increases endothelial nitric oxide synthase phosphorylation via AMP-activated protein kinase and reduces contractility of isolated rat mesenteric resistance arteries. Clin Sci (London). 2011;121(10):449–58.CrossRef
500.
go back to reference Agewall S, Hernberg A. Atorvastatin normalizes endothelial function in healthy smokers. Clin Sci (London). 2006;111(1):87–91.CrossRef Agewall S, Hernberg A. Atorvastatin normalizes endothelial function in healthy smokers. Clin Sci (London). 2006;111(1):87–91.CrossRef
501.
go back to reference Martínez Aguilar E, De Haro Miralles J, Flórez González A, Varela Casariego C, Bleda Moreno S, Acín García F. In vivo confirmation of the role of statins in reducing nitric oxide and C-reactive protein levels in peripheral arterial disease. Eur J Vasc Endovasc Surg. 2009;37(4):443–7.PubMedCrossRef Martínez Aguilar E, De Haro Miralles J, Flórez González A, Varela Casariego C, Bleda Moreno S, Acín García F. In vivo confirmation of the role of statins in reducing nitric oxide and C-reactive protein levels in peripheral arterial disease. Eur J Vasc Endovasc Surg. 2009;37(4):443–7.PubMedCrossRef
502.
go back to reference Margaritis M, Channon KM, Antoniades C. Statins as regulators of redox state in the vascular endothelium: beyond lipid lowering. Antioxid Redox Signal. 2014;20(8):1198–215.PubMedPubMedCentralCrossRef Margaritis M, Channon KM, Antoniades C. Statins as regulators of redox state in the vascular endothelium: beyond lipid lowering. Antioxid Redox Signal. 2014;20(8):1198–215.PubMedPubMedCentralCrossRef
503.
go back to reference Beckman JA, Liao JK, Hurley S, Garrett LA, Chui D, Mitra D, Creager MA. Atorvastatin restores endothelial function in normocholesterolemic smokers independent of changes in low-density lipoprotein. Circ Res. 2004;95(2):217–23.PubMedPubMedCentralCrossRef Beckman JA, Liao JK, Hurley S, Garrett LA, Chui D, Mitra D, Creager MA. Atorvastatin restores endothelial function in normocholesterolemic smokers independent of changes in low-density lipoprotein. Circ Res. 2004;95(2):217–23.PubMedPubMedCentralCrossRef
504.
go back to reference John S, Schneider MP, Delles C, Jacobi J, Schmieder RE. Lipid-independent effects of statins on endothelial function and bioavailability of nitric oxide in hypercholesterolemic patients. Am Heart J. 2005;149(3):473.PubMedCrossRef John S, Schneider MP, Delles C, Jacobi J, Schmieder RE. Lipid-independent effects of statins on endothelial function and bioavailability of nitric oxide in hypercholesterolemic patients. Am Heart J. 2005;149(3):473.PubMedCrossRef
505.
go back to reference Sikora J, Kostka B, Marczyk I, Krajewska U, Chałubiński M, Broncel M. Effect of statins on platelet function in patients with hyperlipidemia. Arch Med Sci. 2013;9(4):622–8.PubMedPubMedCentralCrossRef Sikora J, Kostka B, Marczyk I, Krajewska U, Chałubiński M, Broncel M. Effect of statins on platelet function in patients with hyperlipidemia. Arch Med Sci. 2013;9(4):622–8.PubMedPubMedCentralCrossRef
506.
go back to reference Barale C, Frascaroli C, Senkeev R, Cavalot F, Russo I. Simvastatin effects on inflammation and platelet activation markers in hypercholesterolemia. Biomed Res Int. 2018;2018:6508709.PubMedPubMedCentralCrossRef Barale C, Frascaroli C, Senkeev R, Cavalot F, Russo I. Simvastatin effects on inflammation and platelet activation markers in hypercholesterolemia. Biomed Res Int. 2018;2018:6508709.PubMedPubMedCentralCrossRef
507.
go back to reference Zhou T, Zhou SH, Qi SS, Shen XQ, Zeng GF, Zhou HN. The effect of atorvastatin on serum myeloperoxidase and CRP levels in patients with acute coronary syndrome. Clin Chim Acta. 2006;368(1–2):168–72.PubMedCrossRef Zhou T, Zhou SH, Qi SS, Shen XQ, Zeng GF, Zhou HN. The effect of atorvastatin on serum myeloperoxidase and CRP levels in patients with acute coronary syndrome. Clin Chim Acta. 2006;368(1–2):168–72.PubMedCrossRef
508.
go back to reference Stenvinkel P, Rodríguez-Ayala E, Massy ZA, Qureshi AR, Barany P, Fellström B, Heimburger O, Lindholm B, Alvestrand A. Statin treatment and diabetes affect myeloperoxidase activity in maintenance hemodialysis patients. Clin J Am Soc Nephrol. 2006;1(2):281–7.PubMedCrossRef Stenvinkel P, Rodríguez-Ayala E, Massy ZA, Qureshi AR, Barany P, Fellström B, Heimburger O, Lindholm B, Alvestrand A. Statin treatment and diabetes affect myeloperoxidase activity in maintenance hemodialysis patients. Clin J Am Soc Nephrol. 2006;1(2):281–7.PubMedCrossRef
509.
go back to reference Yoshida T, Yamashita M, Iwai M, Hayashi M. Endothelial Kruppel-like factor 4 mediates the protective effect of statins against ischemic AKI. J Am Soc Nephrol. 2016;27(5):1379–88.PubMedCrossRef Yoshida T, Yamashita M, Iwai M, Hayashi M. Endothelial Kruppel-like factor 4 mediates the protective effect of statins against ischemic AKI. J Am Soc Nephrol. 2016;27(5):1379–88.PubMedCrossRef
510.
go back to reference Gao Y, Liu XF, Lu XC, Ma C, Cao J, Fan L. Protective effects of atorvastatin against oxidized LDL-induced downregulation of KLF expression in EA.hy926 cells. Int J Mol Med. 2012;30(2):330–6.PubMedCrossRef Gao Y, Liu XF, Lu XC, Ma C, Cao J, Fan L. Protective effects of atorvastatin against oxidized LDL-induced downregulation of KLF expression in EA.hy926 cells. Int J Mol Med. 2012;30(2):330–6.PubMedCrossRef
511.
go back to reference Li Y, Xian M, Yang B, Ying M, He Q. Inhibition of KLF4 by statins reverses adriamycin-induced metastasis and cancer stemness in osteosarcoma cells. Stem Cell Rep. 2017;8(6):1617–29.CrossRef Li Y, Xian M, Yang B, Ying M, He Q. Inhibition of KLF4 by statins reverses adriamycin-induced metastasis and cancer stemness in osteosarcoma cells. Stem Cell Rep. 2017;8(6):1617–29.CrossRef
512.
go back to reference Alis R, Sanchis-Gomar F, Risso-Ballester J, Perez-Quilis C, Cortell-Ballester J, Romagnoli M, Blesa JR, Emanuele E. Inhibition of xanthine oxidase to prevent statin-induced myalgia and rhabdomiolysis. Atherosclerosis. 2015;239(1):38–42.PubMedCrossRef Alis R, Sanchis-Gomar F, Risso-Ballester J, Perez-Quilis C, Cortell-Ballester J, Romagnoli M, Blesa JR, Emanuele E. Inhibition of xanthine oxidase to prevent statin-induced myalgia and rhabdomiolysis. Atherosclerosis. 2015;239(1):38–42.PubMedCrossRef
513.
go back to reference Stewart RA. Predicting benefit from statins by C-reactive protein, LDL-cholesterol or absolute cardiovascular risk. Futur Cardiol. 2009;5(3):231–6.CrossRef Stewart RA. Predicting benefit from statins by C-reactive protein, LDL-cholesterol or absolute cardiovascular risk. Futur Cardiol. 2009;5(3):231–6.CrossRef
514.
go back to reference Rosendo AB, Lima LO, Dal-Pizzol F, Almeida S. Lipid and C-reactive protein levels, cardiovascular disease risk factors and simvastatin treatment in Brazilian individuals. Inflammation. 2010;33(4):244–50.PubMedCrossRef Rosendo AB, Lima LO, Dal-Pizzol F, Almeida S. Lipid and C-reactive protein levels, cardiovascular disease risk factors and simvastatin treatment in Brazilian individuals. Inflammation. 2010;33(4):244–50.PubMedCrossRef
515.
go back to reference Datta S, Iqbal Z, Prasad KR. Comparison between serum hsCRP and LDL cholesterol for search of a better predictor for ischemic heart disease. Indian J Clin Biochem. 2011;26(2):210–3.PubMedPubMedCentralCrossRef Datta S, Iqbal Z, Prasad KR. Comparison between serum hsCRP and LDL cholesterol for search of a better predictor for ischemic heart disease. Indian J Clin Biochem. 2011;26(2):210–3.PubMedPubMedCentralCrossRef
516.
go back to reference Mandosi E, Fallarino M, Gatti A, Carnovale A, Rossetti M, Lococo E, Buchetti B, Filetti S, Lenti L, Morano S. Atorvastatin downregulates monocyte CD36 expression, nuclear NFkappaB and TNFalpha levels in type 2 diabetes. J Atheroscler Thromb. 2010;17(6):539–45.PubMedCrossRef Mandosi E, Fallarino M, Gatti A, Carnovale A, Rossetti M, Lococo E, Buchetti B, Filetti S, Lenti L, Morano S. Atorvastatin downregulates monocyte CD36 expression, nuclear NFkappaB and TNFalpha levels in type 2 diabetes. J Atheroscler Thromb. 2010;17(6):539–45.PubMedCrossRef
517.
go back to reference Xie W, Li P, Wang Z, Chen J, Lin Z, Liang X, Mo Y. Rosuvastatin may reduce the incidence of cardiovascular events in patients with acute coronary syndromes receiving percutaneous coronary intervention by suppressing miR-155/SHIP-1 signaling pathway. Cardiovasc Ther. 2014;32(6):276–82.PubMedCrossRef Xie W, Li P, Wang Z, Chen J, Lin Z, Liang X, Mo Y. Rosuvastatin may reduce the incidence of cardiovascular events in patients with acute coronary syndromes receiving percutaneous coronary intervention by suppressing miR-155/SHIP-1 signaling pathway. Cardiovasc Ther. 2014;32(6):276–82.PubMedCrossRef
518.
go back to reference Rezaie-Majd A, Maca T, Bucek RA, Valent P, Muller MR, Husslein P, Kashanipour A, Minar E, Baghestanian M. Simvastatin reduces expression of cytokines interleukin-6, interleukin-8, and monocyte chemoattractant protein-1 in circulating monocytes from hypercholesterolemic patients. Arterioscler Thromb Vasc Biol. 2002;22(7):1194–9.PubMedCrossRef Rezaie-Majd A, Maca T, Bucek RA, Valent P, Muller MR, Husslein P, Kashanipour A, Minar E, Baghestanian M. Simvastatin reduces expression of cytokines interleukin-6, interleukin-8, and monocyte chemoattractant protein-1 in circulating monocytes from hypercholesterolemic patients. Arterioscler Thromb Vasc Biol. 2002;22(7):1194–9.PubMedCrossRef
519.
go back to reference Zinellu A, Paliogiannis P, Usai MF, Carru C, Mangoni AA. Effect of statin treatment on circulating malondialdehyde concentrations: a systematic review and meta-analysis. Ther Adv Chronic Dis. 2019;10:2040622319862714.PubMedPubMedCentralCrossRef Zinellu A, Paliogiannis P, Usai MF, Carru C, Mangoni AA. Effect of statin treatment on circulating malondialdehyde concentrations: a systematic review and meta-analysis. Ther Adv Chronic Dis. 2019;10:2040622319862714.PubMedPubMedCentralCrossRef
520.
go back to reference Rasmussen ST, Andersen JT, Nielsen TK, Cejvanovic V, Petersen KM, Henriksen T, Weimann A, Lykkesfeldt J, Poulsen HE. Simvastatin and oxidative stress in humans: a randomized, double-blinded, placebo-controlled clinical trial. Redox Biol. 2016;9:32–8.PubMedPubMedCentralCrossRef Rasmussen ST, Andersen JT, Nielsen TK, Cejvanovic V, Petersen KM, Henriksen T, Weimann A, Lykkesfeldt J, Poulsen HE. Simvastatin and oxidative stress in humans: a randomized, double-blinded, placebo-controlled clinical trial. Redox Biol. 2016;9:32–8.PubMedPubMedCentralCrossRef
521.
go back to reference Bouitbir J, Sanvee GM, Panajatovic MV, Singh F, Krahenbuhl S: Mechanisms of statin-associated skeletal muscle-associated symptoms. Pharmacol Res 2019:104201. Bouitbir J, Sanvee GM, Panajatovic MV, Singh F, Krahenbuhl S: Mechanisms of statin-associated skeletal muscle-associated symptoms. Pharmacol Res 2019:104201.
522.
go back to reference Yilmaz MI, Baykal Y, Kilic M, Sonmez A, Bulucu F, Aydin A, Sayal A, Kocar IH. Effects of statins on oxidative stress. Biol Trace Elem Res. 2004;98(2):119–27.PubMedCrossRef Yilmaz MI, Baykal Y, Kilic M, Sonmez A, Bulucu F, Aydin A, Sayal A, Kocar IH. Effects of statins on oxidative stress. Biol Trace Elem Res. 2004;98(2):119–27.PubMedCrossRef
523.
go back to reference Liu A, Wu Q, Guo J, Ares I, Rodriguez JL, Martinez-Larranaga MR, Yuan Z, Anadon A, Wang X, Martinez MA. Statins: adverse reactions, oxidative stress and metabolic interactions. Pharmacol Ther. 2019;195:54–84.PubMedCrossRef Liu A, Wu Q, Guo J, Ares I, Rodriguez JL, Martinez-Larranaga MR, Yuan Z, Anadon A, Wang X, Martinez MA. Statins: adverse reactions, oxidative stress and metabolic interactions. Pharmacol Ther. 2019;195:54–84.PubMedCrossRef
525.
go back to reference Abid H, Cartier D, Hamieh A, Francois-Bellan AM, Bucharles C, Pothion H, Manecka DL, Leprince J, Adriouch S, Boyer O, et al. AMPK activation of PGC-1alpha/NRF-1-dependent SELENOT gene transcription promotes PACAP-induced neuroendocrine cell differentiation through tolerance to oxidative stress. Mol Neurobiol. 2019;56(6):4086–101.PubMedCrossRef Abid H, Cartier D, Hamieh A, Francois-Bellan AM, Bucharles C, Pothion H, Manecka DL, Leprince J, Adriouch S, Boyer O, et al. AMPK activation of PGC-1alpha/NRF-1-dependent SELENOT gene transcription promotes PACAP-induced neuroendocrine cell differentiation through tolerance to oxidative stress. Mol Neurobiol. 2019;56(6):4086–101.PubMedCrossRef
526.
go back to reference Lin ZF, Wang CY, Shen LJ, Hsiao FY, Lin Wu FL. Statin use and the risk for incident diabetes mellitus in patients with acute coronary syndrome after percutaneous coronary intervention: a population-based retrospective cohort study in Taiwan. Can J Diabetes. 2016;40(3):264–9.PubMedCrossRef Lin ZF, Wang CY, Shen LJ, Hsiao FY, Lin Wu FL. Statin use and the risk for incident diabetes mellitus in patients with acute coronary syndrome after percutaneous coronary intervention: a population-based retrospective cohort study in Taiwan. Can J Diabetes. 2016;40(3):264–9.PubMedCrossRef
527.
go back to reference Urbano F, Bugliani M, Filippello A, Scamporrino A, Di Mauro S, Di Pino A, Scicali R, Noto D, Rabuazzo AM, Averna M, et al. Atorvastatin but not pravastatin impairs mitochondrial function in human pancreatic islets and rat β-cells. Direct effect of oxidative stress. Sci Rep. 2017;7(1):11863.PubMedPubMedCentralCrossRef Urbano F, Bugliani M, Filippello A, Scamporrino A, Di Mauro S, Di Pino A, Scicali R, Noto D, Rabuazzo AM, Averna M, et al. Atorvastatin but not pravastatin impairs mitochondrial function in human pancreatic islets and rat β-cells. Direct effect of oxidative stress. Sci Rep. 2017;7(1):11863.PubMedPubMedCentralCrossRef
528.
go back to reference Qu H, Y-y M, Chai H, Liang F, J-y Z, Z-y G, D-z S. The effect of statin treatment on circulating coenzyme Q10 concentrations: an updated meta-analysis of randomized controlled trials. Eur J Med Res. 2018;23(1):57.PubMedPubMedCentralCrossRef Qu H, Y-y M, Chai H, Liang F, J-y Z, Z-y G, D-z S. The effect of statin treatment on circulating coenzyme Q10 concentrations: an updated meta-analysis of randomized controlled trials. Eur J Med Res. 2018;23(1):57.PubMedPubMedCentralCrossRef
529.
go back to reference Banach M, Serban C, Ursoniu S, Rysz J, Muntner P, Toth PP, Jones SR, Rizzo M, Glasser SP, Watts GF, et al. Statin therapy and plasma coenzyme Q10 concentrations--a systematic review and meta-analysis of placebo-controlled trials. Pharmacol Res. 2015;99:329–36.PubMedCrossRef Banach M, Serban C, Ursoniu S, Rysz J, Muntner P, Toth PP, Jones SR, Rizzo M, Glasser SP, Watts GF, et al. Statin therapy and plasma coenzyme Q10 concentrations--a systematic review and meta-analysis of placebo-controlled trials. Pharmacol Res. 2015;99:329–36.PubMedCrossRef
530.
go back to reference Qu H, Guo M, Chai H, Wang W, Gao ZY, Shi DZ. Effects of coenzyme Q10 on statin-induced myopathy: an updated meta-analysis of randomized controlled trials. J Am Heart Assoc. 2018;7(19):e009835.PubMedPubMedCentralCrossRef Qu H, Guo M, Chai H, Wang W, Gao ZY, Shi DZ. Effects of coenzyme Q10 on statin-induced myopathy: an updated meta-analysis of randomized controlled trials. J Am Heart Assoc. 2018;7(19):e009835.PubMedPubMedCentralCrossRef
531.
go back to reference Banach M, Serban C, Sahebkar A, Ursoniu S, Rysz J, Muntner P, Toth PP, Jones SR, Rizzo M, Glasser SP, et al. Effects of coenzyme Q10 on statin-induced myopathy: a meta-analysis of randomized controlled trials. Mayo Clin Proc. 2015;90(1):24–34.PubMedCrossRef Banach M, Serban C, Sahebkar A, Ursoniu S, Rysz J, Muntner P, Toth PP, Jones SR, Rizzo M, Glasser SP, et al. Effects of coenzyme Q10 on statin-induced myopathy: a meta-analysis of randomized controlled trials. Mayo Clin Proc. 2015;90(1):24–34.PubMedCrossRef
532.
go back to reference Morris G, Anderson G, Berk M, Maes M. Coenzyme Q10 depletion in medical and neuropsychiatric disorders: potential repercussions and therapeutic implications. Mol Neurobiol. 2013;48(3):883–903.PubMedCrossRef Morris G, Anderson G, Berk M, Maes M. Coenzyme Q10 depletion in medical and neuropsychiatric disorders: potential repercussions and therapeutic implications. Mol Neurobiol. 2013;48(3):883–903.PubMedCrossRef
533.
534.
go back to reference Gao L, Mao Q, Cao J, Wang Y, Zhou X, Fan L. Effects of coenzyme Q10 on vascular endothelial function in humans: a meta-analysis of randomized controlled trials. Atherosclerosis. 2012;221(2):311–6.PubMedCrossRef Gao L, Mao Q, Cao J, Wang Y, Zhou X, Fan L. Effects of coenzyme Q10 on vascular endothelial function in humans: a meta-analysis of randomized controlled trials. Atherosclerosis. 2012;221(2):311–6.PubMedCrossRef
535.
go back to reference Huo J, Xu Z, Hosoe K, Kubo H, Miyahara H, Dai J, Mori M, Sawashita J, Higuchi K. Coenzyme Q10 prevents senescence and dysfunction caused by oxidative stress in vascular endothelial cells. Oxidative Med Cell Longev. 2018;2018:3181759.CrossRef Huo J, Xu Z, Hosoe K, Kubo H, Miyahara H, Dai J, Mori M, Sawashita J, Higuchi K. Coenzyme Q10 prevents senescence and dysfunction caused by oxidative stress in vascular endothelial cells. Oxidative Med Cell Longev. 2018;2018:3181759.CrossRef
536.
go back to reference Yang YK, Wang LP, Chen L, Yao XP, Yang KQ, Gao LG, Zhou XL. Coenzyme Q10 treatment of cardiovascular disorders of ageing including heart failure, hypertension and endothelial dysfunction. Clin Chim Acta. 2015;450:83–9.PubMedCrossRef Yang YK, Wang LP, Chen L, Yao XP, Yang KQ, Gao LG, Zhou XL. Coenzyme Q10 treatment of cardiovascular disorders of ageing including heart failure, hypertension and endothelial dysfunction. Clin Chim Acta. 2015;450:83–9.PubMedCrossRef
537.
go back to reference Rossman MJ, Santos-Parker JR, Steward CAC, Bispham NZ, Cuevas LM, Rosenberg HL, Woodward KA, Chonchol M, Gioscia-Ryan RA, Murphy MP, et al. Chronic supplementation with a mitochondrial antioxidant (mitoQ) improves vascular function in healthy older adults. Hypertension. 2018;71(6):1056–63.PubMedCrossRef Rossman MJ, Santos-Parker JR, Steward CAC, Bispham NZ, Cuevas LM, Rosenberg HL, Woodward KA, Chonchol M, Gioscia-Ryan RA, Murphy MP, et al. Chronic supplementation with a mitochondrial antioxidant (mitoQ) improves vascular function in healthy older adults. Hypertension. 2018;71(6):1056–63.PubMedCrossRef
538.
go back to reference Dai Y-L, Luk T-H, Yiu K-H, Wang M, Yip PMC, Lee SWL, Li S-W, Tam S, Fong B, Lau C-P, et al. Reversal of mitochondrial dysfunction by coenzyme Q10 supplement improves endothelial function in patients with ischaemic left ventricular systolic dysfunction: a randomized controlled trial. Atherosclerosis. 2011;216(2):395–401.PubMedCrossRef Dai Y-L, Luk T-H, Yiu K-H, Wang M, Yip PMC, Lee SWL, Li S-W, Tam S, Fong B, Lau C-P, et al. Reversal of mitochondrial dysfunction by coenzyme Q10 supplement improves endothelial function in patients with ischaemic left ventricular systolic dysfunction: a randomized controlled trial. Atherosclerosis. 2011;216(2):395–401.PubMedCrossRef
539.
go back to reference Hamilton SJ, Chew GT, Watts GF. Coenzyme Q10 improves endothelial dysfunction in statin-treated type 2 diabetic patients. Diabetes Care. 2009;32:810–2. Hamilton SJ, Chew GT, Watts GF. Coenzyme Q10 improves endothelial dysfunction in statin-treated type 2 diabetic patients. Diabetes Care. 2009;32:810–2.
540.
go back to reference Tiano L, Belardinelli R, Carnevali P, Principi F, Seddaiu G, Littarru GP. Effect of coenzyme Q10 administration on endothelial function and extracellular superoxide dismutase in patients with ischaemic heart disease: a double-blind, randomized controlled study. Eur Heart J. 2007;28(18):2249–55.PubMedCrossRef Tiano L, Belardinelli R, Carnevali P, Principi F, Seddaiu G, Littarru GP. Effect of coenzyme Q10 administration on endothelial function and extracellular superoxide dismutase in patients with ischaemic heart disease: a double-blind, randomized controlled study. Eur Heart J. 2007;28(18):2249–55.PubMedCrossRef
541.
go back to reference Suarez-Rivero JM, Pastor-Maldonado CJ, de la Mata M, Villanueva-Paz M, Povea-Cabello S, Alvarez-Cordoba M, Villalon-Garcia I, Suarez-Carrillo A, Talaveron-Rey M, Munuera M, et al. Atherosclerosis and coenzyme Q10. Int J Mol Sci. 2019;20(20). Suarez-Rivero JM, Pastor-Maldonado CJ, de la Mata M, Villanueva-Paz M, Povea-Cabello S, Alvarez-Cordoba M, Villalon-Garcia I, Suarez-Carrillo A, Talaveron-Rey M, Munuera M, et al. Atherosclerosis and coenzyme Q10. Int J Mol Sci. 2019;20(20).
542.
go back to reference Zozina VI, Covantev S, Goroshko OA, Krasnykh LM, Kukes VG. Coenzyme Q10 in cardiovascular and metabolic diseases: current state of the problem. Curr Cardiol Rev. 2018;14(3):164–74.PubMedPubMedCentralCrossRef Zozina VI, Covantev S, Goroshko OA, Krasnykh LM, Kukes VG. Coenzyme Q10 in cardiovascular and metabolic diseases: current state of the problem. Curr Cardiol Rev. 2018;14(3):164–74.PubMedPubMedCentralCrossRef
543.
go back to reference Ayers J, Cook J, Koenig RA, Sisson EM, Dixon DL. Recent developments in the role of coenzyme Q10 for coronary heart disease: a systematic review. Curr Atheroscler Rep. 2018;20(6):29.PubMedCrossRef Ayers J, Cook J, Koenig RA, Sisson EM, Dixon DL. Recent developments in the role of coenzyme Q10 for coronary heart disease: a systematic review. Curr Atheroscler Rep. 2018;20(6):29.PubMedCrossRef
544.
go back to reference Serebruany VL, Gurbel PA, Ordonez JV, Herzog WR, Rohde M, Mortensen SA, Folkers K. Could coenzyme Q10 affect hemostasis by inhibiting platelet vitronectin (CD51/CD61) receptor? Mol Asp Med. 1997;18(Suppl):S189–94.CrossRef Serebruany VL, Gurbel PA, Ordonez JV, Herzog WR, Rohde M, Mortensen SA, Folkers K. Could coenzyme Q10 affect hemostasis by inhibiting platelet vitronectin (CD51/CD61) receptor? Mol Asp Med. 1997;18(Suppl):S189–94.CrossRef
545.
go back to reference Serebruany VL, Ordonez JV, Herzog WR, Rohde M, Mortensen SA, Folkers K, Gurbel PA. Dietary coenzyme Q10 supplementation alters platelet size and inhibits human vitronectin (CD51/CD61) receptor expression. J Cardiovasc Pharmacol. 1997;29(1):16–22.PubMedCrossRef Serebruany VL, Ordonez JV, Herzog WR, Rohde M, Mortensen SA, Folkers K, Gurbel PA. Dietary coenzyme Q10 supplementation alters platelet size and inhibits human vitronectin (CD51/CD61) receptor expression. J Cardiovasc Pharmacol. 1997;29(1):16–22.PubMedCrossRef
546.
go back to reference Ya F, Xu XR, Shi Y, Gallant RC, Song F, Zuo X, Zhao Y, Tian Z, Zhang C, Xu X, et al. Coenzyme Q10 upregulates platelet cAMP/PKA pathway and attenuates integrin αIIbβ3 signaling and thrombus growth. Mol Nutr Food Res. 2019;63(23):1900662.CrossRef Ya F, Xu XR, Shi Y, Gallant RC, Song F, Zuo X, Zhao Y, Tian Z, Zhang C, Xu X, et al. Coenzyme Q10 upregulates platelet cAMP/PKA pathway and attenuates integrin αIIbβ3 signaling and thrombus growth. Mol Nutr Food Res. 2019;63(23):1900662.CrossRef
547.
go back to reference Belardinelli R, Muçaj A, Lacalaprice F, Solenghi M, Seddaiu G, Principi F, Tiano L, Littarru GP. Coenzyme Q10 and exercise training in chronic heart failure. Eur Heart J. 2006;27(22):2675–81.PubMedCrossRef Belardinelli R, Muçaj A, Lacalaprice F, Solenghi M, Seddaiu G, Principi F, Tiano L, Littarru GP. Coenzyme Q10 and exercise training in chronic heart failure. Eur Heart J. 2006;27(22):2675–81.PubMedCrossRef
548.
go back to reference Moccia M, Capacchione A, Lanzillo R, Carbone F, Micillo T, Perna F, De Rosa A, Carotenuto A, Albero R, Matarese G, et al. Coenzyme Q10 supplementation reduces peripheral oxidative stress and inflammation in interferon-beta1a-treated multiple sclerosis. Ther Adv Neurol Disord. 2019;12:1756286418819074.PubMedPubMedCentral Moccia M, Capacchione A, Lanzillo R, Carbone F, Micillo T, Perna F, De Rosa A, Carotenuto A, Albero R, Matarese G, et al. Coenzyme Q10 supplementation reduces peripheral oxidative stress and inflammation in interferon-beta1a-treated multiple sclerosis. Ther Adv Neurol Disord. 2019;12:1756286418819074.PubMedPubMedCentral
549.
go back to reference Barden AE, Shinde S, Burke V, Puddey IB, Beilin LJ, Irish AB, Watts GF, Mori TA. The effect of n-3 fatty acids and coenzyme Q10 supplementation on neutrophil leukotrienes, mediators of inflammation resolution and myeloperoxidase in chronic kidney disease. Prostaglandins Other Lipid Mediat. 2018;136:1–8.PubMedCrossRef Barden AE, Shinde S, Burke V, Puddey IB, Beilin LJ, Irish AB, Watts GF, Mori TA. The effect of n-3 fatty acids and coenzyme Q10 supplementation on neutrophil leukotrienes, mediators of inflammation resolution and myeloperoxidase in chronic kidney disease. Prostaglandins Other Lipid Mediat. 2018;136:1–8.PubMedCrossRef
550.
go back to reference Fan L, Feng Y, Chen GC, Qin LQ, Fu CL, Chen LH. Effects of coenzyme Q10 supplementation on inflammatory markers: a systematic review and meta-analysis of randomized controlled trials. Pharmacol Res. 2017;119:128–36.PubMedCrossRef Fan L, Feng Y, Chen GC, Qin LQ, Fu CL, Chen LH. Effects of coenzyme Q10 supplementation on inflammatory markers: a systematic review and meta-analysis of randomized controlled trials. Pharmacol Res. 2017;119:128–36.PubMedCrossRef
551.
go back to reference Sanoobar M, Eghtesadi S, Azimi A, Khalili M, Jazayeri S, Reza Gohari M. Coenzyme Q10 supplementation reduces oxidative stress and increases antioxidant enzyme activity in patients with relapsing-remitting multiple sclerosis. Int J Neurosci. 2013;123(11):776–82.PubMedCrossRef Sanoobar M, Eghtesadi S, Azimi A, Khalili M, Jazayeri S, Reza Gohari M. Coenzyme Q10 supplementation reduces oxidative stress and increases antioxidant enzyme activity in patients with relapsing-remitting multiple sclerosis. Int J Neurosci. 2013;123(11):776–82.PubMedCrossRef
552.
go back to reference Sanoobar M, Eghtesadi S, Azimi A, Khalili M, Khodadadi B, Jazayeri S, Gohari MR, Aryaeian N. Coenzyme Q10 supplementation ameliorates inflammatory markers in patients with multiple sclerosis: a double blind, placebo, controlled randomized clinical trial. Nutr Neurosci. 2015;18(4):169–76.PubMedCrossRef Sanoobar M, Eghtesadi S, Azimi A, Khalili M, Khodadadi B, Jazayeri S, Gohari MR, Aryaeian N. Coenzyme Q10 supplementation ameliorates inflammatory markers in patients with multiple sclerosis: a double blind, placebo, controlled randomized clinical trial. Nutr Neurosci. 2015;18(4):169–76.PubMedCrossRef
553.
go back to reference Zhai J, Bo Y, Lu Y, Liu C, Zhang L. Effects of coenzyme Q10 on markers of inflammation: a systematic review and meta-analysis. PLoS One. 2017;12(1):e0170172.PubMedPubMedCentralCrossRef Zhai J, Bo Y, Lu Y, Liu C, Zhang L. Effects of coenzyme Q10 on markers of inflammation: a systematic review and meta-analysis. PLoS One. 2017;12(1):e0170172.PubMedPubMedCentralCrossRef
554.
go back to reference Tsai KL, Huang YH, Kao CL, Yang DM, Lee HC, Chou HY, Chen YC, Chiou GY, Chen LH, Yang YP, et al. A novel mechanism of coenzyme Q10 protects against human endothelial cells from oxidative stress-induced injury by modulating NO-related pathways. J Nutr Biochem. 2012;23(5):458–68.PubMedCrossRef Tsai KL, Huang YH, Kao CL, Yang DM, Lee HC, Chou HY, Chen YC, Chiou GY, Chen LH, Yang YP, et al. A novel mechanism of coenzyme Q10 protects against human endothelial cells from oxidative stress-induced injury by modulating NO-related pathways. J Nutr Biochem. 2012;23(5):458–68.PubMedCrossRef
555.
go back to reference Forester BP, Harper DG, Georgakas J, Ravichandran C, Madurai N, Cohen BM. Antidepressant effects of open label treatment with coenzyme Q10 in geriatric bipolar depression. J Clin Psychopharmacol. 2015;35(3):338–40.PubMedPubMedCentralCrossRef Forester BP, Harper DG, Georgakas J, Ravichandran C, Madurai N, Cohen BM. Antidepressant effects of open label treatment with coenzyme Q10 in geriatric bipolar depression. J Clin Psychopharmacol. 2015;35(3):338–40.PubMedPubMedCentralCrossRef
556.
go back to reference Mehrpooya M, Yasrebifar F, Haghighi M, Mohammadi Y, Jahangard L. Evaluating the effect of coenzyme Q10 augmentation on treatment of bipolar depression: a double-blind controlled clinical trial. J Clin Psychopharmacol. 2018;38(5):460–6.PubMedCrossRef Mehrpooya M, Yasrebifar F, Haghighi M, Mohammadi Y, Jahangard L. Evaluating the effect of coenzyme Q10 augmentation on treatment of bipolar depression: a double-blind controlled clinical trial. J Clin Psychopharmacol. 2018;38(5):460–6.PubMedCrossRef
558.
go back to reference Maes M, Mihaylova I, Kubera M, Uytterhoeven M, Vrydags N, Bosmans E. Lower plasma Coenzyme Q10 in depression: a marker for treatment resistance and chronic fatigue in depression and a risk factor to cardiovascular disorder in that illness. Neuroendocrinol Lett. 2009;30(4):462–9. Maes M, Mihaylova I, Kubera M, Uytterhoeven M, Vrydags N, Bosmans E. Lower plasma Coenzyme Q10 in depression: a marker for treatment resistance and chronic fatigue in depression and a risk factor to cardiovascular disorder in that illness. Neuroendocrinol Lett. 2009;30(4):462–9.
559.
go back to reference Bello RI, Kagan VE, Tyurin V, Navarro F, Alcain FJ, Villalba JM. Regeneration of lipophilic antioxidants by NAD(P)H:quinone oxidoreductase 1. Protoplasma. 2003;221(1–2):129–35.PubMedCrossRef Bello RI, Kagan VE, Tyurin V, Navarro F, Alcain FJ, Villalba JM. Regeneration of lipophilic antioxidants by NAD(P)H:quinone oxidoreductase 1. Protoplasma. 2003;221(1–2):129–35.PubMedCrossRef
560.
go back to reference Gomez-Diaz C, Rodriguez-Aguilera JC, Barroso MP, Villalba JM, Navarro F, Crane FL, Navas P. Antioxidant ascorbate is stabilized by NADH-coenzyme Q10 reductase in the plasma membrane. J Bioenerg Biomembr. 1997;29(3):251–7.PubMedCrossRef Gomez-Diaz C, Rodriguez-Aguilera JC, Barroso MP, Villalba JM, Navarro F, Crane FL, Navas P. Antioxidant ascorbate is stabilized by NADH-coenzyme Q10 reductase in the plasma membrane. J Bioenerg Biomembr. 1997;29(3):251–7.PubMedCrossRef
561.
go back to reference Isobe C, Abe T, Terayama Y. Increase in the oxidized/total coenzyme Q-10 ratio in the cerebrospinal fluid of Alzheimer’s disease patients. Dement Geriatr Cogn Disord. 2009;28(5):434–9.CrossRef Isobe C, Abe T, Terayama Y. Increase in the oxidized/total coenzyme Q-10 ratio in the cerebrospinal fluid of Alzheimer’s disease patients. Dement Geriatr Cogn Disord. 2009;28(5):434–9.CrossRef
562.
go back to reference Morris G, Puri BK, Walker AJ, Maes M, Carvalho AF, Walder K, Mazza C, Berk M. Myalgic encephalomyelitis/chronic fatigue syndrome: from pathophysiological insights to novel therapeutic opportunities. Pharmacol Res. 2019;148:104450.PubMedCrossRef Morris G, Puri BK, Walker AJ, Maes M, Carvalho AF, Walder K, Mazza C, Berk M. Myalgic encephalomyelitis/chronic fatigue syndrome: from pathophysiological insights to novel therapeutic opportunities. Pharmacol Res. 2019;148:104450.PubMedCrossRef
563.
go back to reference Yubero D, Allen G, Artuch R, Montero R. The value of coenzyme Q(10) determination in mitochondrial patients. J Clin Med. 2017;6(4):37.PubMedCentralCrossRef Yubero D, Allen G, Artuch R, Montero R. The value of coenzyme Q(10) determination in mitochondrial patients. J Clin Med. 2017;6(4):37.PubMedCentralCrossRef
565.
go back to reference McGarry A, McDermott M, Kieburtz K, de Blieck EA, Beal F, Marder K, Ross C, Shoulson I, Gilbert P, Mallonee WM, et al. A randomized, double-blind, placebo-controlled trial of coenzyme Q10 in Huntington disease. Neurology. 2017;88(2):152–9.PubMedPubMedCentralCrossRef McGarry A, McDermott M, Kieburtz K, de Blieck EA, Beal F, Marder K, Ross C, Shoulson I, Gilbert P, Mallonee WM, et al. A randomized, double-blind, placebo-controlled trial of coenzyme Q10 in Huntington disease. Neurology. 2017;88(2):152–9.PubMedPubMedCentralCrossRef
566.
go back to reference LFd RI, Burón MI, Clarke CF, Villalba JM. Polyunsaturated fatty acids directly regulate coenzyme Q biosynthesis. FASEB J. 2018;32(1_supplement):539.515. LFd RI, Burón MI, Clarke CF, Villalba JM. Polyunsaturated fatty acids directly regulate coenzyme Q biosynthesis. FASEB J. 2018;32(1_supplement):539.515.
567.
go back to reference Beharry KD, Cai CL, Henry MM, Chowdhury S, Valencia GB, Aranda JV. Co-enzyme Q10 and n-3 polyunsaturated fatty acid supplementation reverse intermittent hypoxia-induced growth restriction and improved antioxidant profiles in neonatal rats. Antioxidants (Basel). 2017;6(4). Beharry KD, Cai CL, Henry MM, Chowdhury S, Valencia GB, Aranda JV. Co-enzyme Q10 and n-3 polyunsaturated fatty acid supplementation reverse intermittent hypoxia-induced growth restriction and improved antioxidant profiles in neonatal rats. Antioxidants (Basel). 2017;6(4).
568.
569.
go back to reference Choi HD, Chae SM. Comparison of efficacy and safety of combination therapy with statins and omega-3 fatty acids versus statin monotherapy in patients with dyslipidemia: a systematic review and meta-analysis. Medicine (Baltimore). 2018;97(50):e13593.CrossRef Choi HD, Chae SM. Comparison of efficacy and safety of combination therapy with statins and omega-3 fatty acids versus statin monotherapy in patients with dyslipidemia: a systematic review and meta-analysis. Medicine (Baltimore). 2018;97(50):e13593.CrossRef
570.
go back to reference Toth S, Sajty M, Pekarova T, Mughees A, Stefanic P, Katz M, Spisakova K, Pella J, Pella D. Addition of omega-3 fatty acid and coenzyme Q10 to statin therapy in patients with combined dyslipidemia. J Basic Clin Physiol Pharmacol. 2017;28(4):327–36.PubMedCrossRef Toth S, Sajty M, Pekarova T, Mughees A, Stefanic P, Katz M, Spisakova K, Pella J, Pella D. Addition of omega-3 fatty acid and coenzyme Q10 to statin therapy in patients with combined dyslipidemia. J Basic Clin Physiol Pharmacol. 2017;28(4):327–36.PubMedCrossRef
571.
go back to reference Zehr KR, Walker MK. Omega-3 polyunsaturated fatty acids improve endothelial function in humans at risk for atherosclerosis: a review. Prostaglandins Other Lipid Mediators. 2018;134:131–40.PubMedCrossRef Zehr KR, Walker MK. Omega-3 polyunsaturated fatty acids improve endothelial function in humans at risk for atherosclerosis: a review. Prostaglandins Other Lipid Mediators. 2018;134:131–40.PubMedCrossRef
572.
go back to reference Wiest EF, Walsh-Wilcox MT, Walker MK. Omega-3 polyunsaturated fatty acids protect against cigarette smoke-induced oxidative stress and vascular dysfunction. Toxicol Sci. 2017;156(1):300–10.PubMedPubMedCentral Wiest EF, Walsh-Wilcox MT, Walker MK. Omega-3 polyunsaturated fatty acids protect against cigarette smoke-induced oxidative stress and vascular dysfunction. Toxicol Sci. 2017;156(1):300–10.PubMedPubMedCentral
573.
go back to reference Hu Y, Hu FB, Manson JE. Marine omega-3 supplementation and cardiovascular disease: an updated meta-analysis of 13 randomized controlled trials involving 127 477 participants. J Am Heart Assoc. 2019;8(19):e013543.PubMedPubMedCentralCrossRef Hu Y, Hu FB, Manson JE. Marine omega-3 supplementation and cardiovascular disease: an updated meta-analysis of 13 randomized controlled trials involving 127 477 participants. J Am Heart Assoc. 2019;8(19):e013543.PubMedPubMedCentralCrossRef
574.
go back to reference Cohen MG, Rossi JS, Garbarino J, Bowling R, Motsinger-Reif AA, Schuler C, Dupont AG, Gabriel D. Insights into the inhibition of platelet activation by omega-3 polyunsaturated fatty acids: beyond aspirin and clopidogrel. Thromb Res. 2011;128(4):335–40.PubMedCrossRef Cohen MG, Rossi JS, Garbarino J, Bowling R, Motsinger-Reif AA, Schuler C, Dupont AG, Gabriel D. Insights into the inhibition of platelet activation by omega-3 polyunsaturated fatty acids: beyond aspirin and clopidogrel. Thromb Res. 2011;128(4):335–40.PubMedCrossRef
575.
go back to reference McEwen BJ, Morel-Kopp MC, Chen W, Tofler GH, Ward CM. Effects of omega-3 polyunsaturated fatty acids on platelet function in healthy subjects and subjects with cardiovascular disease. Semin Thromb Hemost. 2013;39(1):25–32.PubMedCrossRef McEwen BJ, Morel-Kopp MC, Chen W, Tofler GH, Ward CM. Effects of omega-3 polyunsaturated fatty acids on platelet function in healthy subjects and subjects with cardiovascular disease. Semin Thromb Hemost. 2013;39(1):25–32.PubMedCrossRef
576.
go back to reference Adili R, Hawley M, Holinstat M. Regulation of platelet function and thrombosis by omega-3 and omega-6 polyunsaturated fatty acids. Prostaglandins Other Lipid Mediators. 2018;139:10–8.PubMedPubMedCentralCrossRef Adili R, Hawley M, Holinstat M. Regulation of platelet function and thrombosis by omega-3 and omega-6 polyunsaturated fatty acids. Prostaglandins Other Lipid Mediators. 2018;139:10–8.PubMedPubMedCentralCrossRef
577.
go back to reference Prossomariti A, Scaioli E, Piazzi G, Fazio C, Bellanova M, Biagi E, Candela M, Brigidi P, Consolandi C, Balbi T, et al. Short-term treatment with eicosapentaenoic acid improves inflammation and affects colonic differentiation markers and microbiota in patients with ulcerative colitis. Sci Rep. 2017;7(1):7458.PubMedPubMedCentralCrossRef Prossomariti A, Scaioli E, Piazzi G, Fazio C, Bellanova M, Biagi E, Candela M, Brigidi P, Consolandi C, Balbi T, et al. Short-term treatment with eicosapentaenoic acid improves inflammation and affects colonic differentiation markers and microbiota in patients with ulcerative colitis. Sci Rep. 2017;7(1):7458.PubMedPubMedCentralCrossRef
578.
go back to reference Andersen VL, Vogt J, Obel T, Christensen JH, Schmidt EB. The effect of N-3 fatty acids on plasma myeloperoxidase levels in healthy adults. Cell Mol Biology (Noisy-le-Grand). 2010;56(1):3–9. Andersen VL, Vogt J, Obel T, Christensen JH, Schmidt EB. The effect of N-3 fatty acids on plasma myeloperoxidase levels in healthy adults. Cell Mol Biology (Noisy-le-Grand). 2010;56(1):3–9.
579.
go back to reference Tan A, Sullenbarger B, Prakash R, McDaniel JC. Supplementation with eicosapentaenoic acid and docosahexaenoic acid reduces high levels of circulating proinflammatory cytokines in aging adults: a randomized, controlled study. Prostaglandins Leukot Essent Fat Acids. 2018;132:23–9.CrossRef Tan A, Sullenbarger B, Prakash R, McDaniel JC. Supplementation with eicosapentaenoic acid and docosahexaenoic acid reduces high levels of circulating proinflammatory cytokines in aging adults: a randomized, controlled study. Prostaglandins Leukot Essent Fat Acids. 2018;132:23–9.CrossRef
580.
go back to reference Becic T, Studenik C. Effects of Omega-3 supplementation on adipocytokines in prediabetes and type 2 diabetes mellitus: systematic review and meta-analysis of randomized controlled trials. Diabetes Metab J. 2018;42(2):101–16.PubMedPubMedCentralCrossRef Becic T, Studenik C. Effects of Omega-3 supplementation on adipocytokines in prediabetes and type 2 diabetes mellitus: systematic review and meta-analysis of randomized controlled trials. Diabetes Metab J. 2018;42(2):101–16.PubMedPubMedCentralCrossRef
581.
go back to reference O’Mahoney LL, Matu J, Price OJ, Birch KM, Ajjan RA, Farrar D, Tapp R, West DJ, Deighton K, Campbell MD. Omega-3 polyunsaturated fatty acids favourably modulate cardiometabolic biomarkers in type 2 diabetes: a meta-analysis and meta-regression of randomized controlled trials. Cardiovasc Diabetol. 2018;17(1):98.PubMedPubMedCentralCrossRef O’Mahoney LL, Matu J, Price OJ, Birch KM, Ajjan RA, Farrar D, Tapp R, West DJ, Deighton K, Campbell MD. Omega-3 polyunsaturated fatty acids favourably modulate cardiometabolic biomarkers in type 2 diabetes: a meta-analysis and meta-regression of randomized controlled trials. Cardiovasc Diabetol. 2018;17(1):98.PubMedPubMedCentralCrossRef
582.
go back to reference Meital LT, Windsor MT, Perissiou M, Schulze K, Magee R, Kuballa A, Golledge J, Bailey TG, Askew CD, Russell FD. Omega-3 fatty acids decrease oxidative stress and inflammation in macrophages from patients with small abdominal aortic aneurysm. Sci Rep. 2019;9(1):12978.PubMedPubMedCentralCrossRef Meital LT, Windsor MT, Perissiou M, Schulze K, Magee R, Kuballa A, Golledge J, Bailey TG, Askew CD, Russell FD. Omega-3 fatty acids decrease oxidative stress and inflammation in macrophages from patients with small abdominal aortic aneurysm. Sci Rep. 2019;9(1):12978.PubMedPubMedCentralCrossRef
583.
go back to reference Mazereeuw G, Herrmann N, Andreazza AC, Scola G, Ma DWL, Oh PI, Lanctôt KL. Oxidative stress predicts depressive symptom changes with omega-3 fatty acid treatment in coronary artery disease patients. Brain Behav Immun. 2017;60:136–41.PubMedCrossRef Mazereeuw G, Herrmann N, Andreazza AC, Scola G, Ma DWL, Oh PI, Lanctôt KL. Oxidative stress predicts depressive symptom changes with omega-3 fatty acid treatment in coronary artery disease patients. Brain Behav Immun. 2017;60:136–41.PubMedCrossRef
584.
go back to reference Mas E, Woodman RJ, Burke V, Puddey IB, Beilin LJ, Durand T, Mori TA. The omega-3 fatty acids EPA and DHA decrease plasma F2-isoprostanes: results from two placebo-controlled interventions. Free Radic Res. 2010;44(9):983–90.PubMedCrossRef Mas E, Woodman RJ, Burke V, Puddey IB, Beilin LJ, Durand T, Mori TA. The omega-3 fatty acids EPA and DHA decrease plasma F2-isoprostanes: results from two placebo-controlled interventions. Free Radic Res. 2010;44(9):983–90.PubMedCrossRef
585.
go back to reference Herbst EAF, Paglialunga S, Gerling C, Whitfield J, Mukai K, Chabowski A, Heigenhauser GJF, Spriet LL, Holloway GP. Omega-3 supplementation alters mitochondrial membrane composition and respiration kinetics in human skeletal muscle. J Physiol. 2014;592(6):1341–52.PubMedPubMedCentralCrossRef Herbst EAF, Paglialunga S, Gerling C, Whitfield J, Mukai K, Chabowski A, Heigenhauser GJF, Spriet LL, Holloway GP. Omega-3 supplementation alters mitochondrial membrane composition and respiration kinetics in human skeletal muscle. J Physiol. 2014;592(6):1341–52.PubMedPubMedCentralCrossRef
586.
go back to reference Gerling CJ, Mukai K, Chabowski A, Heigenhauser GJF, Holloway GP, Spriet LL, Jannas-Vela S: Incorporation of omega-3 fatty acids into human skeletal muscle sarcolemmal and mitochondrial membranes following 12 weeks of fish oil supplementation. Front Physiol 2019, 10(348). Gerling CJ, Mukai K, Chabowski A, Heigenhauser GJF, Holloway GP, Spriet LL, Jannas-Vela S: Incorporation of omega-3 fatty acids into human skeletal muscle sarcolemmal and mitochondrial membranes following 12 weeks of fish oil supplementation. Front Physiol 2019, 10(348).
587.
go back to reference Koga N, Ogura J, Yoshida F, Hattori K, Hori H, Aizawa E, Ishida I, Kunugi H. Altered polyunsaturated fatty acid levels in relation to proinflammatory cytokines, fatty acid desaturase genotype, and diet in bipolar disorder. Transl Psychiatry. 2019;9(1):208.PubMedPubMedCentralCrossRef Koga N, Ogura J, Yoshida F, Hattori K, Hori H, Aizawa E, Ishida I, Kunugi H. Altered polyunsaturated fatty acid levels in relation to proinflammatory cytokines, fatty acid desaturase genotype, and diet in bipolar disorder. Transl Psychiatry. 2019;9(1):208.PubMedPubMedCentralCrossRef
588.
go back to reference Solberg DK, Refsum H, Andreassen OA, Bentsen H. A five-year follow-up study of antioxidants, oxidative stress and polyunsaturated fatty acids in schizophrenia. Acta Neuropsychiatrica. 2019;31(4):202–12.PubMedCrossRef Solberg DK, Refsum H, Andreassen OA, Bentsen H. A five-year follow-up study of antioxidants, oxidative stress and polyunsaturated fatty acids in schizophrenia. Acta Neuropsychiatrica. 2019;31(4):202–12.PubMedCrossRef
589.
go back to reference Messamore E, Almeida DM, Jandacek RJ, McNamara RK. Polyunsaturated fatty acids and recurrent mood disorders: phenomenology, mechanisms, and clinical application. Prog Lipid Res. 2017;66:1–13.PubMedPubMedCentralCrossRef Messamore E, Almeida DM, Jandacek RJ, McNamara RK. Polyunsaturated fatty acids and recurrent mood disorders: phenomenology, mechanisms, and clinical application. Prog Lipid Res. 2017;66:1–13.PubMedPubMedCentralCrossRef
590.
go back to reference Morris G, Walder K, Puri BK, Berk M, Maes M. The deleterious effects of oxidative and nitrosative stress on palmitoylation, membrane lipid rafts and lipid-based cellular signalling: new drug targets in neuroimmune disorders. Mol Neurobiol. 2016;53(7):4638–58.PubMedCrossRef Morris G, Walder K, Puri BK, Berk M, Maes M. The deleterious effects of oxidative and nitrosative stress on palmitoylation, membrane lipid rafts and lipid-based cellular signalling: new drug targets in neuroimmune disorders. Mol Neurobiol. 2016;53(7):4638–58.PubMedCrossRef
591.
go back to reference Reimers A, Ljung H. The emerging role of omega-3 fatty acids as a therapeutic option in neuropsychiatric disorders. Ther Adv Psychopharmacol. 2019;9:2045125319858901.PubMedPubMedCentralCrossRef Reimers A, Ljung H. The emerging role of omega-3 fatty acids as a therapeutic option in neuropsychiatric disorders. Ther Adv Psychopharmacol. 2019;9:2045125319858901.PubMedPubMedCentralCrossRef
Metadata
Title
Endothelial dysfunction in neuroprogressive disorders—causes and suggested treatments
Authors
Gerwyn Morris
Basant K. Puri
Lisa Olive
Andre Carvalho
Michael Berk
Ken Walder
Lise Tuset Gustad
Michael Maes
Publication date
01-12-2020
Publisher
BioMed Central
Published in
BMC Medicine / Issue 1/2020
Electronic ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-020-01749-w

Other articles of this Issue 1/2020

BMC Medicine 1/2020 Go to the issue