Skip to main content
Top
Published in: Hormones 2/2018

01-06-2018 | Review Article

Ischemic stroke and select adipose-derived and sex hormones: a review

Author: Kristy L. Meadows

Published in: Hormones | Issue 2/2018

Login to get access

Abstract

Ischemic stroke is the fifth leading cause of death in the USA and is the leading cause of serious, long-term disability worldwide. The principle sex hormones (estrogen, progesterone, and testosterone), both endogenous and exogenous, have profound effects on various stroke outcomes and have become the focus of a number of studies evaluating risk factors and treatment options for ischemic stroke. In addition, the expression of other hormones that may influence stroke outcome, including select adipose-derived hormones (adiponectin, leptin, and ghrelin), can be regulated by sex hormones and are also the focus of several ischemic stroke studies. This review aims to summarize some of the preclinical and clinical studies investigating the principle sex hormones, as well as select adipose-derived hormones, as risk factors or potential treatments for ischemic stroke. In addition, the potential for relaxin, a lesser studied sex hormone, as a novel treatment option for ischemic stroke is explored.
Literature
1.
go back to reference Donnan GA, Fisher M, Macleod M, Davis SM (2008) Stroke. Lancet Lond. Engl 371:1612–1623CrossRef Donnan GA, Fisher M, Macleod M, Davis SM (2008) Stroke. Lancet Lond. Engl 371:1612–1623CrossRef
3.
go back to reference Broussalis E et al (2012) Current therapies in ischemic stroke. Part A. Recent developments in acute stroke treatment and in stroke prevention. Drug Discov. Today 17:296–309PubMedCrossRef Broussalis E et al (2012) Current therapies in ischemic stroke. Part A. Recent developments in acute stroke treatment and in stroke prevention. Drug Discov. Today 17:296–309PubMedCrossRef
4.
go back to reference Neuhaus AA et al (2014) Importance of preclinical research in the development of neuroprotective strategies for ischemic stroke. JAMA Neurol 71:634–639PubMedCrossRef Neuhaus AA et al (2014) Importance of preclinical research in the development of neuroprotective strategies for ischemic stroke. JAMA Neurol 71:634–639PubMedCrossRef
5.
go back to reference Fluri F, Schuhmann MK, Kleinschnitz C (2015) Animal models of ischemic stroke and their application in clinical research. Drug Des Devel Ther 9:3445–3454PubMedPubMedCentral Fluri F, Schuhmann MK, Kleinschnitz C (2015) Animal models of ischemic stroke and their application in clinical research. Drug Des Devel Ther 9:3445–3454PubMedPubMedCentral
6.
go back to reference Mozaffarian D et al (2016) Heart disease and stroke atatistics-2016 update: a report from the American Heart Association. Circulation 133:e38–e360PubMed Mozaffarian D et al (2016) Heart disease and stroke atatistics-2016 update: a report from the American Heart Association. Circulation 133:e38–e360PubMed
7.
go back to reference Durukan A, Tatlisumak T (2007) Acute ischemic stroke: overview of major experimental rodent models, pathophysiology, and therapy of focal cerebral ischemia. Pharmacol Biochem Behav 87:179–197PubMedCrossRef Durukan A, Tatlisumak T (2007) Acute ischemic stroke: overview of major experimental rodent models, pathophysiology, and therapy of focal cerebral ischemia. Pharmacol Biochem Behav 87:179–197PubMedCrossRef
8.
go back to reference The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group (1995) Tissue plasminogen activator for acute ischemic stroke. N. Engl. J. Med 333:1581–1587CrossRef The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group (1995) Tissue plasminogen activator for acute ischemic stroke. N. Engl. J. Med 333:1581–1587CrossRef
10.
go back to reference Glendenning ML, Lovekamp-Swan T, Schreihofer DA (2008) Protective effect of estrogen in endothelin-induced middle cerebral artery occlusion in female rats. Neurosci Lett 445:188–192PubMedPubMedCentralCrossRef Glendenning ML, Lovekamp-Swan T, Schreihofer DA (2008) Protective effect of estrogen in endothelin-induced middle cerebral artery occlusion in female rats. Neurosci Lett 445:188–192PubMedPubMedCentralCrossRef
11.
go back to reference Herson PS, Palmateer J, Hurn PD (2013) Biological sex and mechanisms of ischemic brain injury. Transl Stroke Res 4:413–419PubMedCrossRef Herson PS, Palmateer J, Hurn PD (2013) Biological sex and mechanisms of ischemic brain injury. Transl Stroke Res 4:413–419PubMedCrossRef
12.
go back to reference Hurn PD, Macrae IM (2000) Estrogen as a neuroprotectant in stroke. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 20:631–652CrossRef Hurn PD, Macrae IM (2000) Estrogen as a neuroprotectant in stroke. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 20:631–652CrossRef
13.
go back to reference Koellhoffer EC, McCullough LD (2013) The effects of estrogen in ischemic stroke. Transl Stroke Res 4:390–401PubMedCrossRef Koellhoffer EC, McCullough LD (2013) The effects of estrogen in ischemic stroke. Transl Stroke Res 4:390–401PubMedCrossRef
14.
go back to reference Siegel C, Turtzo C, McCullough LD (2010) Sex differences in cerebral ischemia: possible molecular mechanisms. J Neurosci Res 88:2765–2774PubMedCrossRef Siegel C, Turtzo C, McCullough LD (2010) Sex differences in cerebral ischemia: possible molecular mechanisms. J Neurosci Res 88:2765–2774PubMedCrossRef
15.
16.
go back to reference Zuo W, Zhang W, Chen N-H (2013) Sexual dimorphism in cerebral ischemia injury. Eur J Pharmacol 711:73–79PubMedCrossRef Zuo W, Zhang W, Chen N-H (2013) Sexual dimorphism in cerebral ischemia injury. Eur J Pharmacol 711:73–79PubMedCrossRef
17.
go back to reference Caminos JE et al (2003) Expression of ghrelin in the cyclic and pregnant rat ovary. Endocrinology 144:1594–1602PubMedCrossRef Caminos JE et al (2003) Expression of ghrelin in the cyclic and pregnant rat ovary. Endocrinology 144:1594–1602PubMedCrossRef
18.
go back to reference Du C et al (2009) Expression of the orexigenic peptide ghrelin in the sheep ovary. Domest Anim Endocrinol 36:89–98PubMedCrossRef Du C et al (2009) Expression of the orexigenic peptide ghrelin in the sheep ovary. Domest Anim Endocrinol 36:89–98PubMedCrossRef
19.
go back to reference Tvarijonaviciute A, Carrillo-Sanchez JD, Ceron JJ (2013) Effect of estradiol and progesterone on metabolic biomarkers in healthy bitches. Reprod Domest Anim Zuchthyg 48:520–524CrossRef Tvarijonaviciute A, Carrillo-Sanchez JD, Ceron JJ (2013) Effect of estradiol and progesterone on metabolic biomarkers in healthy bitches. Reprod Domest Anim Zuchthyg 48:520–524CrossRef
20.
go back to reference DeLahunta, A., Glass, E. & Kent, M. Veterinary neuroanatomy and clinical neurology. (Elsevier, 2015) DeLahunta, A., Glass, E. & Kent, M. Veterinary neuroanatomy and clinical neurology. (Elsevier, 2015)
21.
go back to reference Garosi L, Cerebrovascular S (2010) Disease in dogs and cats. Vet Clin North Am Small Anim Pract 40:65–79PubMedCrossRef Garosi L, Cerebrovascular S (2010) Disease in dogs and cats. Vet Clin North Am Small Anim Pract 40:65–79PubMedCrossRef
22.
go back to reference Henderson VW, Lobo RA (2012) Hormone therapy and the risk of stroke: perspectives 10 years after the Women’s Health Initiative trials. Climacteric J Int Menopause Soc 15:229–234CrossRef Henderson VW, Lobo RA (2012) Hormone therapy and the risk of stroke: perspectives 10 years after the Women’s Health Initiative trials. Climacteric J Int Menopause Soc 15:229–234CrossRef
24.
go back to reference Balaskó M, Soós S, Székely M, Pétervári E (2014) Leptin and aging: review and questions with particular emphasis on its role in the central regulation of energy balance. J Chem Neuroanat 61–62:248–255PubMedCrossRef Balaskó M, Soós S, Székely M, Pétervári E (2014) Leptin and aging: review and questions with particular emphasis on its role in the central regulation of energy balance. J Chem Neuroanat 61–62:248–255PubMedCrossRef
25.
go back to reference Pétervári E et al (2014) Age versus nutritional state in the development of central leptin resistance. Peptides 56:59–67PubMedCrossRef Pétervári E et al (2014) Age versus nutritional state in the development of central leptin resistance. Peptides 56:59–67PubMedCrossRef
26.
go back to reference Ebrahimi-Mamaeghani M, Mohammadi S, Arefhosseini SR, Fallah P, Bazi Z (2015) Adiponectin as a potential biomarker of vascular disease. Vasc Health Risk Manag 11:55–70PubMedPubMedCentral Ebrahimi-Mamaeghani M, Mohammadi S, Arefhosseini SR, Fallah P, Bazi Z (2015) Adiponectin as a potential biomarker of vascular disease. Vasc Health Risk Manag 11:55–70PubMedPubMedCentral
27.
go back to reference Kantorová E et al (2015) The intricate network of adipokines and stroke. Int J Endocrinol 2015(967698) Kantorová E et al (2015) The intricate network of adipokines and stroke. Int J Endocrinol 2015(967698)
28.
go back to reference Matsumoto M, Ishikawa S, Kajii E (2008) Association of adiponectin with cerebrovascular disease: a nested case-control study. Stroke J. Cereb. Circ. 39:323–328CrossRef Matsumoto M, Ishikawa S, Kajii E (2008) Association of adiponectin with cerebrovascular disease: a nested case-control study. Stroke J. Cereb. Circ. 39:323–328CrossRef
29.
go back to reference Meier U (2004) Endocrine regulation of energy metabolism: review of pathobiochemical and clinical chemical aspects of leptin, ghrelin, adiponectin, and resistin. Clin Chem 50:1511–1525PubMedCrossRef Meier U (2004) Endocrine regulation of energy metabolism: review of pathobiochemical and clinical chemical aspects of leptin, ghrelin, adiponectin, and resistin. Clin Chem 50:1511–1525PubMedCrossRef
30.
go back to reference Vaiopoulos AG, Marinou K, Christodoulides C, Koutsilieris M (2012) The role of adiponectin in human vascular physiology. Int J Cardiol 155:188–193PubMedCrossRef Vaiopoulos AG, Marinou K, Christodoulides C, Koutsilieris M (2012) The role of adiponectin in human vascular physiology. Int J Cardiol 155:188–193PubMedCrossRef
32.
go back to reference Schäfer K, Konstantinides S (2011) Adipokines and thrombosis. Clin Exp Pharmacol Physiol 38:864–871PubMedCrossRef Schäfer K, Konstantinides S (2011) Adipokines and thrombosis. Clin Exp Pharmacol Physiol 38:864–871PubMedCrossRef
33.
go back to reference Song J, Lee WT, Park KA, Lee JE (2014) Association between risk factors for vascular dementia and adiponectin. Biomed Res Int 2014:261672PubMedPubMedCentral Song J, Lee WT, Park KA, Lee JE (2014) Association between risk factors for vascular dementia and adiponectin. Biomed Res Int 2014:261672PubMedPubMedCentral
34.
go back to reference Chen M-P et al (2005) Hypoadiponectinemia is associated with ischemic cerebrovascular disease. Arterioscler Thromb Vasc Biol 25:821–826PubMedCrossRef Chen M-P et al (2005) Hypoadiponectinemia is associated with ischemic cerebrovascular disease. Arterioscler Thromb Vasc Biol 25:821–826PubMedCrossRef
35.
go back to reference Kuwashiro T et al (2014) Significance of plasma adiponectin for diagnosis, neurological severity and functional outcome in ischemic stroke—Research for Biomarkers in Ischemic Stroke (REBIOS). Metabolism 63:1093–1103PubMedCrossRef Kuwashiro T et al (2014) Significance of plasma adiponectin for diagnosis, neurological severity and functional outcome in ischemic stroke—Research for Biomarkers in Ischemic Stroke (REBIOS). Metabolism 63:1093–1103PubMedCrossRef
36.
go back to reference Söderberg S et al (2004) Leptin, but not adiponectin, predicts stroke in males. J Intern Med 256:128–136PubMedCrossRef Söderberg S et al (2004) Leptin, but not adiponectin, predicts stroke in males. J Intern Med 256:128–136PubMedCrossRef
37.
go back to reference Wolf I et al (2006) Adiponectin, ghrelin, and leptin in cancer cachexia in breast and colon cancer patients. Cancer 106:966–973PubMedCrossRef Wolf I et al (2006) Adiponectin, ghrelin, and leptin in cancer cachexia in breast and colon cancer patients. Cancer 106:966–973PubMedCrossRef
38.
go back to reference Arregui M et al (2014) Adiponectin and risk of stroke: prospective study and meta-analysis. Stroke J. Cereb. Circ. 45:10–17CrossRef Arregui M et al (2014) Adiponectin and risk of stroke: prospective study and meta-analysis. Stroke J. Cereb. Circ. 45:10–17CrossRef
39.
go back to reference Bienek R et al (2012) Adiponectin, leptin, resistin and insulin blood concentrations in patients with ischaemic cerebral stroke. Endokrynol Pol 63:338–345PubMed Bienek R et al (2012) Adiponectin, leptin, resistin and insulin blood concentrations in patients with ischaemic cerebral stroke. Endokrynol Pol 63:338–345PubMed
40.
go back to reference Gardener H et al (2013) Adiponectin and risk of vascular events in the Northern Manhattan study. Atherosclerosis 226:483–489PubMedCrossRef Gardener H et al (2013) Adiponectin and risk of vascular events in the Northern Manhattan study. Atherosclerosis 226:483–489PubMedCrossRef
41.
go back to reference Kos K et al (2007) Adiponectin and resistin in human cerebrospinal fluid and expression of adiponectin receptors in the human hypothalamus. J Clin Endocrinol Metab 92:1129–1136PubMedCrossRef Kos K et al (2007) Adiponectin and resistin in human cerebrospinal fluid and expression of adiponectin receptors in the human hypothalamus. J Clin Endocrinol Metab 92:1129–1136PubMedCrossRef
42.
go back to reference Bidulescu A et al (2013) Associations of adiponectin and leptin with incident coronary heart disease and ischemic stroke in African Americans: the Jackson heart study. Front Public Health 1:16PubMedPubMedCentralCrossRef Bidulescu A et al (2013) Associations of adiponectin and leptin with incident coronary heart disease and ischemic stroke in African Americans: the Jackson heart study. Front Public Health 1:16PubMedPubMedCentralCrossRef
43.
go back to reference Kizer JR et al (2013) Total and high-molecular-weight adiponectin and risk of coronary heart disease and ischemic stroke in older adults. J Clin Endocrinol Metab 98:255–263PubMedCrossRef Kizer JR et al (2013) Total and high-molecular-weight adiponectin and risk of coronary heart disease and ischemic stroke in older adults. J Clin Endocrinol Metab 98:255–263PubMedCrossRef
44.
45.
go back to reference Chen H, Montagnani M, Funahashi T, Shimomura I, Quon MJ (2003) Adiponectin stimulates production of nitric oxide in vascular endothelial cells. J Biol Chem 278:45021–45026PubMedCrossRef Chen H, Montagnani M, Funahashi T, Shimomura I, Quon MJ (2003) Adiponectin stimulates production of nitric oxide in vascular endothelial cells. J Biol Chem 278:45021–45026PubMedCrossRef
46.
go back to reference Tomicek NJ, Hunter JC, Machikas AM, Lopez V, Korzick DH (2015) Acute adiponectin delivery is cardioprotective in the aged female rat heart. Geriatr Gerontol Int 15:636–646PubMedCrossRef Tomicek NJ, Hunter JC, Machikas AM, Lopez V, Korzick DH (2015) Acute adiponectin delivery is cardioprotective in the aged female rat heart. Geriatr Gerontol Int 15:636–646PubMedCrossRef
47.
go back to reference Yatomi K et al (2009) Pathophysiological dual action of adiponectin after transient focal ischemia in mouse brain. Brain Res 1297:169–176PubMedCrossRef Yatomi K et al (2009) Pathophysiological dual action of adiponectin after transient focal ischemia in mouse brain. Brain Res 1297:169–176PubMedCrossRef
48.
go back to reference Chen B et al (2009) Adiponectin protects against cerebral ischemia-reperfusion injury through anti-inflammatory action. Brain Res 1273:129–137PubMedCrossRef Chen B et al (2009) Adiponectin protects against cerebral ischemia-reperfusion injury through anti-inflammatory action. Brain Res 1273:129–137PubMedCrossRef
50.
go back to reference Nishimura M et al (2008) Adiponectin prevents cerebral ischemic injury through endothelial nitric oxide synthase dependent mechanisms. Circulation 117:216–223PubMedCrossRef Nishimura M et al (2008) Adiponectin prevents cerebral ischemic injury through endothelial nitric oxide synthase dependent mechanisms. Circulation 117:216–223PubMedCrossRef
51.
go back to reference Song W et al (2015) Therapeutic window of globular adiponectin against cerebral ischemia in diabetic mice: the role of dynamic alteration of adiponectin/adiponectin receptor expression. Sci Rep 5:17310PubMedPubMedCentralCrossRef Song W et al (2015) Therapeutic window of globular adiponectin against cerebral ischemia in diabetic mice: the role of dynamic alteration of adiponectin/adiponectin receptor expression. Sci Rep 5:17310PubMedPubMedCentralCrossRef
52.
go back to reference Shen L-H, Miao J, Zhao Y-J, Zhao Y-J, Liang W (2014) Expression of brain adiponectin in a murine model of transient cerebral ischemia. Int J Clin Exp Med 7:4590–4596PubMedPubMedCentral Shen L-H, Miao J, Zhao Y-J, Zhao Y-J, Liang W (2014) Expression of brain adiponectin in a murine model of transient cerebral ischemia. Int J Clin Exp Med 7:4590–4596PubMedPubMedCentral
54.
go back to reference Kishida K, Funahashi T, Shimomura I (2014) Adiponectin as a routine clinical biomarker. Best Pract Res Clin Endocrinol Metab 28:119–130PubMedCrossRef Kishida K, Funahashi T, Shimomura I (2014) Adiponectin as a routine clinical biomarker. Best Pract Res Clin Endocrinol Metab 28:119–130PubMedCrossRef
55.
go back to reference McEntegart MB et al (2007) Increase in serum adiponectin concentration in patients with heart failure and cachexia: relationship with leptin, other cytokines, and B-type natriuretic peptide. Eur Heart J 28:829–835PubMedCrossRef McEntegart MB et al (2007) Increase in serum adiponectin concentration in patients with heart failure and cachexia: relationship with leptin, other cytokines, and B-type natriuretic peptide. Eur Heart J 28:829–835PubMedCrossRef
56.
go back to reference Kantorova E et al (2011) Leptin, adiponectin and ghrelin, new potential mediators of ischemic stroke. Neuro Endocrinol Lett 32:716–721PubMed Kantorova E et al (2011) Leptin, adiponectin and ghrelin, new potential mediators of ischemic stroke. Neuro Endocrinol Lett 32:716–721PubMed
57.
go back to reference Kim BJ, Lee S-H, Ryu W-S, Kim CK, Yoon B-W (2012) Adipocytokines and ischemic stroke: differential associations between stroke subtypes. J Neurol Sci 312:117–122PubMedCrossRef Kim BJ, Lee S-H, Ryu W-S, Kim CK, Yoon B-W (2012) Adipocytokines and ischemic stroke: differential associations between stroke subtypes. J Neurol Sci 312:117–122PubMedCrossRef
58.
go back to reference Efstathiou SP et al (2005) Plasma adiponectin levels and five-year survival after first-ever ischemic stroke. Stroke J. Cereb. Circ. 36:1915–1919CrossRef Efstathiou SP et al (2005) Plasma adiponectin levels and five-year survival after first-ever ischemic stroke. Stroke J. Cereb. Circ. 36:1915–1919CrossRef
59.
go back to reference Nagasawa H, Yokota C, Toyoda K, Ito A, Minematsu K (2011) High level of plasma adiponectin in acute stroke patients is associated with stroke mortality. J Neurol Sci 304:102–106PubMedCrossRef Nagasawa H, Yokota C, Toyoda K, Ito A, Minematsu K (2011) High level of plasma adiponectin in acute stroke patients is associated with stroke mortality. J Neurol Sci 304:102–106PubMedCrossRef
60.
go back to reference Stott DJ et al (2009) Adipocytokines and risk of stroke in older people: a nested case-control study. Int J Epidemiol 38:253–261PubMedCrossRef Stott DJ et al (2009) Adipocytokines and risk of stroke in older people: a nested case-control study. Int J Epidemiol 38:253–261PubMedCrossRef
61.
go back to reference Gardener H et al (2012) Adiponectin and carotid intima-media thickness in the northern Manhattan study. Stroke J. Cereb. Circ. 43:1123–1125CrossRef Gardener H et al (2012) Adiponectin and carotid intima-media thickness in the northern Manhattan study. Stroke J. Cereb. Circ. 43:1123–1125CrossRef
62.
go back to reference Prugger C et al (2012) Adipocytokines and the risk of ischemic stroke: the PRIME study. Ann Neurol 71:478–486PubMedCrossRef Prugger C et al (2012) Adipocytokines and the risk of ischemic stroke: the PRIME study. Ann Neurol 71:478–486PubMedCrossRef
63.
go back to reference Rajpathak SN et al (2011) Resistin, but not adiponectin and leptin, is associated with the risk of ischemic stroke among postmenopausal women: results from the Women’s Health Initiative. Stroke J. Cereb. Circ. 42:1813–1820CrossRef Rajpathak SN et al (2011) Resistin, but not adiponectin and leptin, is associated with the risk of ischemic stroke among postmenopausal women: results from the Women’s Health Initiative. Stroke J. Cereb. Circ. 42:1813–1820CrossRef
64.
go back to reference Ogorodnikova AD et al (2010) High-molecular-weight adiponectin and incident ischemic stroke in postmenopausal women: a Women’s Health Initiative Study. Stroke J. Cereb. Circ. 41:1376–1381CrossRef Ogorodnikova AD et al (2010) High-molecular-weight adiponectin and incident ischemic stroke in postmenopausal women: a Women’s Health Initiative Study. Stroke J. Cereb. Circ. 41:1376–1381CrossRef
65.
go back to reference Kanhai DA et al (2013) Adiponectin and incident coronary heart disease and stroke. A systematic review and meta-analysis of prospective studies. Obes Rev Off J Int Assoc Study Obes 14:555–567CrossRef Kanhai DA et al (2013) Adiponectin and incident coronary heart disease and stroke. A systematic review and meta-analysis of prospective studies. Obes Rev Off J Int Assoc Study Obes 14:555–567CrossRef
66.
go back to reference Park H-K, Ahima RS (2015) Physiology of leptin: energy homeostasis, neuroendocrine function and metabolism. Metabolism 64:24–34PubMedCrossRef Park H-K, Ahima RS (2015) Physiology of leptin: energy homeostasis, neuroendocrine function and metabolism. Metabolism 64:24–34PubMedCrossRef
68.
go back to reference Wilkinson M, Brown R, Imran SA, Ur E (2007) Adipokine gene expression in brain and pituitary gland. Neuroendocrinology 86:191–209PubMedCrossRef Wilkinson M, Brown R, Imran SA, Ur E (2007) Adipokine gene expression in brain and pituitary gland. Neuroendocrinology 86:191–209PubMedCrossRef
69.
go back to reference Paz-Filho GJ (2016) The effects of leptin replacement on neural plasticity. Neural Plast 2016(8528934) Paz-Filho GJ (2016) The effects of leptin replacement on neural plasticity. Neural Plast 2016(8528934)
70.
71.
go back to reference Denis RGP, Bing C, Naderali EK, Vernon RG, Williams G (2003) Lactation modulates diurnal expression profiles of specific leptin receptor isoforms in the rat hypothalamus. J Endocrinol 178:225–232PubMedCrossRef Denis RGP, Bing C, Naderali EK, Vernon RG, Williams G (2003) Lactation modulates diurnal expression profiles of specific leptin receptor isoforms in the rat hypothalamus. J Endocrinol 178:225–232PubMedCrossRef
72.
go back to reference Khafaji HARH, Bener AB, Rizk NM, Al Suwaidi J (2012) Elevated serum leptin levels in patients with acute myocardial infarction; correlation with coronary angiographic and echocardiographic findings. BMC Res Notes 5:262PubMedPubMedCentralCrossRef Khafaji HARH, Bener AB, Rizk NM, Al Suwaidi J (2012) Elevated serum leptin levels in patients with acute myocardial infarction; correlation with coronary angiographic and echocardiographic findings. BMC Res Notes 5:262PubMedPubMedCentralCrossRef
73.
go back to reference Mu J, Ostrowski RP, Krafft PR, Tang J, Zhang JH (2013) Serum leptin levels decrease after permanent MCAo in the rat and remain unaffected by delayed hyperbaric oxygen therapy. Med Gas Res 3:8PubMedPubMedCentralCrossRef Mu J, Ostrowski RP, Krafft PR, Tang J, Zhang JH (2013) Serum leptin levels decrease after permanent MCAo in the rat and remain unaffected by delayed hyperbaric oxygen therapy. Med Gas Res 3:8PubMedPubMedCentralCrossRef
75.
go back to reference Liu J et al (2010) Leptinemia and its association with stroke and coronary heart disease in the Jackson Heart Study. Clin Endocrinol 72(32–37) Liu J et al (2010) Leptinemia and its association with stroke and coronary heart disease in the Jackson Heart Study. Clin Endocrinol 72(32–37)
76.
go back to reference Lukasik M et al (2012) Reactive leptin resistance and the profile of platelet activation in acute ischaemic stroke patients. Thromb Haemost 108:107–118PubMedCrossRef Lukasik M et al (2012) Reactive leptin resistance and the profile of platelet activation in acute ischaemic stroke patients. Thromb Haemost 108:107–118PubMedCrossRef
77.
go back to reference Sierra-Johnson J et al (2007) Relation of increased leptin concentrations to history of myocardial infarction and stroke in the United States population. Am J Cardiol 100:234–239PubMedPubMedCentralCrossRef Sierra-Johnson J et al (2007) Relation of increased leptin concentrations to history of myocardial infarction and stroke in the United States population. Am J Cardiol 100:234–239PubMedPubMedCentralCrossRef
78.
go back to reference Söderberg S et al (2003) High leptin levels are associated with stroke. Cerebrovasc Dis Basel Switz 15:63–69CrossRef Söderberg S et al (2003) High leptin levels are associated with stroke. Cerebrovasc Dis Basel Switz 15:63–69CrossRef
79.
go back to reference Signore AP, Zhang F, Weng Z, Gao Y, Chen J (2008) Leptin neuroprotection in the CNS: mechanisms and therapeutic potentials. J Neurochem 106:1977–1990PubMedPubMedCentralCrossRef Signore AP, Zhang F, Weng Z, Gao Y, Chen J (2008) Leptin neuroprotection in the CNS: mechanisms and therapeutic potentials. J Neurochem 106:1977–1990PubMedPubMedCentralCrossRef
80.
go back to reference Zhang F, Wang S, Signore AP, Chen J (2007) Neuroprotective effects of leptin against ischemic injury induced by oxygen-glucose deprivation and transient cerebral ischemia. Stroke J Cereb Circ 38:2329–2336CrossRef Zhang F, Wang S, Signore AP, Chen J (2007) Neuroprotective effects of leptin against ischemic injury induced by oxygen-glucose deprivation and transient cerebral ischemia. Stroke J Cereb Circ 38:2329–2336CrossRef
81.
go back to reference Valerio A et al (2009) Leptin is induced in the ischemic cerebral cortex and exerts neuroprotection through NF-kappaB/c-Rel-dependent transcription. Stroke J. Cereb. Circ. 40:610–617CrossRef Valerio A et al (2009) Leptin is induced in the ischemic cerebral cortex and exerts neuroprotection through NF-kappaB/c-Rel-dependent transcription. Stroke J. Cereb. Circ. 40:610–617CrossRef
82.
go back to reference Zhang J et al (2013) Leptin attenuates cerebral ischemia injury through the promotion of energy metabolism via the PI3K/Akt pathway. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 33:567–574CrossRef Zhang J et al (2013) Leptin attenuates cerebral ischemia injury through the promotion of energy metabolism via the PI3K/Akt pathway. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 33:567–574CrossRef
83.
go back to reference Zhang J et al (2011) Leptin attenuates cerebral ischemia/reperfusion injury partially by CGRP expression. Eur J Pharmacol 671:61–69PubMedCrossRef Zhang J et al (2011) Leptin attenuates cerebral ischemia/reperfusion injury partially by CGRP expression. Eur J Pharmacol 671:61–69PubMedCrossRef
84.
go back to reference Avraham Y et al (2013) Delayed leptin administration after stroke induces neurogenesis and angiogenesis. J Neurosci Res 91:187–195PubMedCrossRef Avraham Y et al (2013) Delayed leptin administration after stroke induces neurogenesis and angiogenesis. J Neurosci Res 91:187–195PubMedCrossRef
85.
go back to reference Deng Z-H et al (2014) Inhibition of the connexin 43 elevation may be involved in the neuroprotective activity of leptin against brain ischemic injury. Cell Mol Neurobiol 34:871–879PubMedCrossRef Deng Z-H et al (2014) Inhibition of the connexin 43 elevation may be involved in the neuroprotective activity of leptin against brain ischemic injury. Cell Mol Neurobiol 34:871–879PubMedCrossRef
86.
go back to reference Calleja AI et al (2013) Blood biomarkers of insulin resistance in acute stroke patients treated with intravenous thrombolysis: temporal profile and prognostic value. J Diabetes Res Clin Metab 2:2CrossRef Calleja AI et al (2013) Blood biomarkers of insulin resistance in acute stroke patients treated with intravenous thrombolysis: temporal profile and prognostic value. J Diabetes Res Clin Metab 2:2CrossRef
87.
go back to reference Jalil A, Amir S, Majeed R, Jalil F (2010) Circulating leptin levels in elderly subjects with and without cerebrovasular disease. J Coll Physicians Surg—Pak JCPSP 20:350–351PubMed Jalil A, Amir S, Majeed R, Jalil F (2010) Circulating leptin levels in elderly subjects with and without cerebrovasular disease. J Coll Physicians Surg—Pak JCPSP 20:350–351PubMed
88.
89.
go back to reference Jiménez I et al (2009) High serum levels of leptin are associated with post-stroke depression. Psychol Med 39:1201–1209PubMedCrossRef Jiménez I et al (2009) High serum levels of leptin are associated with post-stroke depression. Psychol Med 39:1201–1209PubMedCrossRef
90.
go back to reference Wannamethee SG, Shaper AG, Whincup PH, Lennon L, Sattar N (2013) Adiposity, adipokines, and risk of incident stroke in older men. Stroke J. Cereb. Circ. 44:3–8CrossRef Wannamethee SG, Shaper AG, Whincup PH, Lennon L, Sattar N (2013) Adiposity, adipokines, and risk of incident stroke in older men. Stroke J. Cereb. Circ. 44:3–8CrossRef
91.
go back to reference Zeng R, Xu C-H, Xu Y-N, Wang Y-L, Wang M (2014) Association of leptin levels with pathogenetic risk of coronary heart disease and stroke: a meta-analysis. Arq Bras Endocrinol Metabol 58:817–823PubMedCrossRef Zeng R, Xu C-H, Xu Y-N, Wang Y-L, Wang M (2014) Association of leptin levels with pathogenetic risk of coronary heart disease and stroke: a meta-analysis. Arq Bras Endocrinol Metabol 58:817–823PubMedCrossRef
92.
go back to reference Ciccone M et al (2001) Plasma leptin is independently associated with the intima-media thickness of the common carotid artery. Int J Obes Relat Metab Disord J Int Assoc Study Obes 25:805–810CrossRef Ciccone M et al (2001) Plasma leptin is independently associated with the intima-media thickness of the common carotid artery. Int J Obes Relat Metab Disord J Int Assoc Study Obes 25:805–810CrossRef
93.
go back to reference Norata GD et al (2007) Leptin: adiponectin ratio is an independent predictor of intima media thickness of the common carotid artery. Stroke J. Cereb. Circ. 38:2844–2846CrossRef Norata GD et al (2007) Leptin: adiponectin ratio is an independent predictor of intima media thickness of the common carotid artery. Stroke J. Cereb. Circ. 38:2844–2846CrossRef
95.
go back to reference Saber H et al (2015) Serum leptin levels and the risk of stroke: the Framingham study. Stroke J. Cereb. Circ. 46:2881–2885CrossRef Saber H et al (2015) Serum leptin levels and the risk of stroke: the Framingham study. Stroke J. Cereb. Circ. 46:2881–2885CrossRef
96.
go back to reference Andrews ZB (2011) Central mechanisms involved in the orexigenic actions of ghrelin. Peptides 32:2248–2255PubMedCrossRef Andrews ZB (2011) Central mechanisms involved in the orexigenic actions of ghrelin. Peptides 32:2248–2255PubMedCrossRef
97.
go back to reference Andrews ZB (2011) The extra-hypothalamic actions of ghrelin on neuronal function. Trends Neurosci 34:31–40PubMedCrossRef Andrews ZB (2011) The extra-hypothalamic actions of ghrelin on neuronal function. Trends Neurosci 34:31–40PubMedCrossRef
98.
go back to reference Solomou S, Korbonits M (2014) The role of ghrelin in weight-regulation disorders: implications in clinical practice. Horm Athens Greece 13:458–475 Solomou S, Korbonits M (2014) The role of ghrelin in weight-regulation disorders: implications in clinical practice. Horm Athens Greece 13:458–475
99.
go back to reference Jeusette IC et al (2005) Effects of chronic obesity and weight loss on plasma ghrelin and leptin concentrations in dogs. Res Vet Sci 79:169–175PubMedCrossRef Jeusette IC et al (2005) Effects of chronic obesity and weight loss on plasma ghrelin and leptin concentrations in dogs. Res Vet Sci 79:169–175PubMedCrossRef
100.
go back to reference Stoyanova II (2014) Ghrelin: a link between ageing, metabolism and neurodegenerative disorders. Neurobiol. Dis 72 Pt A:72–83PubMedCrossRef Stoyanova II (2014) Ghrelin: a link between ageing, metabolism and neurodegenerative disorders. Neurobiol. Dis 72 Pt A:72–83PubMedCrossRef
102.
go back to reference Garcia JM et al (2015) Anamorelin for patients with cancer cachexia: an integrated analysis of two phase 2, randomised, placebo-controlled, double-blind trials. Lancet Oncol 16:108–116PubMedCrossRef Garcia JM et al (2015) Anamorelin for patients with cancer cachexia: an integrated analysis of two phase 2, randomised, placebo-controlled, double-blind trials. Lancet Oncol 16:108–116PubMedCrossRef
103.
go back to reference Seyhanli ES et al (2015) Assessment of serum and urine ghrelin levels in patients with acute stroke. Int J Clin Exp Med 8:722–729PubMedPubMedCentral Seyhanli ES et al (2015) Assessment of serum and urine ghrelin levels in patients with acute stroke. Int J Clin Exp Med 8:722–729PubMedPubMedCentral
104.
go back to reference Dos Santos VV et al (2013) Ghrelin as a neuroprotective and palliative agent in Alzheimer’s and Parkinson’s disease. Curr Pharm Des 19:6773–6790PubMedCrossRef Dos Santos VV et al (2013) Ghrelin as a neuroprotective and palliative agent in Alzheimer’s and Parkinson’s disease. Curr Pharm Des 19:6773–6790PubMedCrossRef
105.
go back to reference Wu R et al (2005) Ghrelin improves tissue perfusion in severe sepsis via downregulation of endothelin-1. Cardiovasc Res 68:318–326PubMedCrossRef Wu R et al (2005) Ghrelin improves tissue perfusion in severe sepsis via downregulation of endothelin-1. Cardiovasc Res 68:318–326PubMedCrossRef
106.
go back to reference Moon M et al (2011) Ghrelin ameliorates cognitive dysfunction and neurodegeneration in intrahippocampal amyloid-β1-42 oligomer-injected mice. J Alzheimers Dis JAD 23:147–159PubMedCrossRef Moon M et al (2011) Ghrelin ameliorates cognitive dysfunction and neurodegeneration in intrahippocampal amyloid-β1-42 oligomer-injected mice. J Alzheimers Dis JAD 23:147–159PubMedCrossRef
107.
go back to reference Xu X, Zhu Y, Chuai J (2012) Changes in serum ghrelin and small intestinal motility in rats with ischemic stroke. Anat Rec Hoboken NJ 2007(295):307–312CrossRef Xu X, Zhu Y, Chuai J (2012) Changes in serum ghrelin and small intestinal motility in rats with ischemic stroke. Anat Rec Hoboken NJ 2007(295):307–312CrossRef
108.
go back to reference Cheyuo C et al (2011) Ghrelin suppresses inflammation and neuronal nitric oxide synthase in focal cerebral ischemia via the vagus nerve. Shock Augusta Ga 35:258–265CrossRef Cheyuo C et al (2011) Ghrelin suppresses inflammation and neuronal nitric oxide synthase in focal cerebral ischemia via the vagus nerve. Shock Augusta Ga 35:258–265CrossRef
109.
go back to reference Kenny R et al (2013) Endogenous ghrelin’s role in hippocampal neuroprotection after global cerebral ischemia: does endogenous ghrelin protect against global stroke? Am J Physiol Regul Integr Comp Physiol 304:R980–R990PubMedCrossRef Kenny R et al (2013) Endogenous ghrelin’s role in hippocampal neuroprotection after global cerebral ischemia: does endogenous ghrelin protect against global stroke? Am J Physiol Regul Integr Comp Physiol 304:R980–R990PubMedCrossRef
110.
go back to reference Matsumoto M et al (2013) Decreased serum ghrelin levels in patients with acute myocardial infarction. Tohoku J Exp Med 231:235–242PubMedCrossRef Matsumoto M et al (2013) Decreased serum ghrelin levels in patients with acute myocardial infarction. Tohoku J Exp Med 231:235–242PubMedCrossRef
111.
go back to reference Sax B et al (2013) Characterization of pericardial and plasma ghrelin levels in patients with ischemic and non-ischemic heart disease. Regul Pept 186:131–136PubMedCrossRef Sax B et al (2013) Characterization of pericardial and plasma ghrelin levels in patients with ischemic and non-ischemic heart disease. Regul Pept 186:131–136PubMedCrossRef
112.
go back to reference Kadoglou NPE et al (2014) Serum levels of novel adipokines in patients with acute ischemic stroke: potential contribution to diagnosis and prognosis. Peptides 57:12–16PubMedCrossRef Kadoglou NPE et al (2014) Serum levels of novel adipokines in patients with acute ischemic stroke: potential contribution to diagnosis and prognosis. Peptides 57:12–16PubMedCrossRef
113.
go back to reference Xu X-D, Shao F (2015) Enteral ecoimmunonutrition reduced enteral permeability and serum ghrelin activity in severe cerebral stroke patients with lung infection. Cell Biochem Biophys 71:195–198PubMedCrossRef Xu X-D, Shao F (2015) Enteral ecoimmunonutrition reduced enteral permeability and serum ghrelin activity in severe cerebral stroke patients with lung infection. Cell Biochem Biophys 71:195–198PubMedCrossRef
114.
go back to reference Ritzel RM, Capozzi LA, McCullough LD (2013) Sex, stroke, and inflammation: the potential for estrogen-mediated immunoprotection in stroke. Horm Behav 63:238–253PubMedCrossRef Ritzel RM, Capozzi LA, McCullough LD (2013) Sex, stroke, and inflammation: the potential for estrogen-mediated immunoprotection in stroke. Horm Behav 63:238–253PubMedCrossRef
115.
go back to reference Duckles SP, Krause DN (2007) Cerebrovascular effects of oestrogen: multiplicity of action. Clin Exp Pharmacol Physiol 34:801–808PubMedCrossRef Duckles SP, Krause DN (2007) Cerebrovascular effects of oestrogen: multiplicity of action. Clin Exp Pharmacol Physiol 34:801–808PubMedCrossRef
117.
go back to reference Schreihofer DA, Ma Y (2013) Estrogen receptors and ischemic neuroprotection: who, what, where, and when? Brain Res 1514:107–122PubMedCrossRef Schreihofer DA, Ma Y (2013) Estrogen receptors and ischemic neuroprotection: who, what, where, and when? Brain Res 1514:107–122PubMedCrossRef
119.
go back to reference Scott E, Zhang Q, Wang R, Vadlamudi R, Brann D (2012) Estrogen neuroprotection and the critical period hypothesis. Front Neuroendocrinol 33:85–104PubMedCrossRef Scott E, Zhang Q, Wang R, Vadlamudi R, Brann D (2012) Estrogen neuroprotection and the critical period hypothesis. Front Neuroendocrinol 33:85–104PubMedCrossRef
120.
go back to reference Bramlett HM (2005) Sex differences and the effect of hormonal therapy on ischemic brain injury. Pathophysiology 12:17–27PubMedCrossRef Bramlett HM (2005) Sex differences and the effect of hormonal therapy on ischemic brain injury. Pathophysiology 12:17–27PubMedCrossRef
122.
go back to reference Geary GG, Krause DN, Duckles SP (1998) Estrogen reduces myogenic tone through a nitric oxide-dependent mechanism in rat cerebral arteries. Am J Phys 275:H292–H300 Geary GG, Krause DN, Duckles SP (1998) Estrogen reduces myogenic tone through a nitric oxide-dependent mechanism in rat cerebral arteries. Am J Phys 275:H292–H300
123.
125.
go back to reference Manwani B et al (2013) Differential effects of aging and sex on stroke induced inflammation across the lifespan. Exp Neurol 249:120–131PubMedCrossRef Manwani B et al (2013) Differential effects of aging and sex on stroke induced inflammation across the lifespan. Exp Neurol 249:120–131PubMedCrossRef
126.
go back to reference Gibson CL, Gray LJ, Murphy SP, Bath PMW (2006) Estrogens and experimental ischemic stroke: a systematic review. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 26:1103–1113CrossRef Gibson CL, Gray LJ, Murphy SP, Bath PMW (2006) Estrogens and experimental ischemic stroke: a systematic review. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 26:1103–1113CrossRef
127.
go back to reference Liu F, Yuan R, Benashski SE, McCullough LD (2009) Changes in experimental stroke outcome across the life span. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 29:792–802CrossRef Liu F, Yuan R, Benashski SE, McCullough LD (2009) Changes in experimental stroke outcome across the life span. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 29:792–802CrossRef
128.
go back to reference Liu F, Benashski SE, Xu Y, Siegel M, McCullough LD (2012) Effects of chronic and acute oestrogen replacement therapy in aged animals after experimental stroke. J Neuroendocrinol 24:319–330PubMedPubMedCentralCrossRef Liu F, Benashski SE, Xu Y, Siegel M, McCullough LD (2012) Effects of chronic and acute oestrogen replacement therapy in aged animals after experimental stroke. J Neuroendocrinol 24:319–330PubMedPubMedCentralCrossRef
129.
go back to reference Viscoli CM et al (2001) A clinical trial of estrogen-replacement therapy after ischemic stroke. N Engl J Med 345:1243–1249PubMedCrossRef Viscoli CM et al (2001) A clinical trial of estrogen-replacement therapy after ischemic stroke. N Engl J Med 345:1243–1249PubMedCrossRef
130.
go back to reference Wassertheil-Smoller S et al (2003) Effect of estrogen plus progestin on stroke in postmenopausal women: the Women’s Health Initiative: a randomized trial. JAMA 289:2673–2684PubMedCrossRef Wassertheil-Smoller S et al (2003) Effect of estrogen plus progestin on stroke in postmenopausal women: the Women’s Health Initiative: a randomized trial. JAMA 289:2673–2684PubMedCrossRef
132.
go back to reference Maggio M et al (2009) Relationship between higher estradiol levels and 9-year mortality in older women: the Invecchiare in Chianti study. J Am Geriatr Soc 57:1810–1815PubMedPubMedCentralCrossRef Maggio M et al (2009) Relationship between higher estradiol levels and 9-year mortality in older women: the Invecchiare in Chianti study. J Am Geriatr Soc 57:1810–1815PubMedPubMedCentralCrossRef
134.
go back to reference Pappa T et al (2012) Estradiol levels predict short-term adverse health outcomes in postmenopausal acute stroke women. Eur J Neurol 19:1300–1304PubMedCrossRef Pappa T et al (2012) Estradiol levels predict short-term adverse health outcomes in postmenopausal acute stroke women. Eur J Neurol 19:1300–1304PubMedCrossRef
135.
go back to reference Schumacher M et al (2014) Revisiting the roles of progesterone and allopregnanolone in the nervous system: resurgence of the progesterone receptors. Prog Neurobiol 113:6–39PubMedCrossRef Schumacher M et al (2014) Revisiting the roles of progesterone and allopregnanolone in the nervous system: resurgence of the progesterone receptors. Prog Neurobiol 113:6–39PubMedCrossRef
136.
go back to reference Gibson CL, Coomber B, Rathbone J (2009) Is progesterone a candidate neuroprotective factor for treatment following ischemic stroke? Neurosci Rev J Bringing Neurobiol Neurol Psychiatry 15:324–332 Gibson CL, Coomber B, Rathbone J (2009) Is progesterone a candidate neuroprotective factor for treatment following ischemic stroke? Neurosci Rev J Bringing Neurobiol Neurol Psychiatry 15:324–332
137.
go back to reference Gibson CL, Gray LJ, Bath PMW, Murphy SP (2008) Progesterone for the treatment of experimental brain injury; a systematic review. Brain J Neurol 131:318–328CrossRef Gibson CL, Gray LJ, Bath PMW, Murphy SP (2008) Progesterone for the treatment of experimental brain injury; a systematic review. Brain J Neurol 131:318–328CrossRef
138.
go back to reference Wong R et al (2013) Progesterone treatment for experimental stroke: an individual animal meta-analysis. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 33:1362–1372CrossRef Wong R et al (2013) Progesterone treatment for experimental stroke: an individual animal meta-analysis. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 33:1362–1372CrossRef
139.
go back to reference Peterson BL, Won S, Geddes RI, Sayeed I, Stein DG (2015) Sex-related differences in effects of progesterone following neonatal hypoxic brain injury. Behav Brain Res 286:152–165PubMedCrossRef Peterson BL, Won S, Geddes RI, Sayeed I, Stein DG (2015) Sex-related differences in effects of progesterone following neonatal hypoxic brain injury. Behav Brain Res 286:152–165PubMedCrossRef
141.
142.
go back to reference Quillinan N, Deng G, Grewal H, Herson PS (2014) Androgens and stroke: good, bad or indifferent? Exp Neurol 259:10–15PubMedCrossRef Quillinan N, Deng G, Grewal H, Herson PS (2014) Androgens and stroke: good, bad or indifferent? Exp Neurol 259:10–15PubMedCrossRef
143.
go back to reference Geary GG, Krause DN, Duckles SP (2000) Gonadal hormones affect diameter of male rat cerebral arteries through endothelium-dependent mechanisms. Am J Physiol Heart Circ Physiol 279:H610–H618PubMedCrossRef Geary GG, Krause DN, Duckles SP (2000) Gonadal hormones affect diameter of male rat cerebral arteries through endothelium-dependent mechanisms. Am J Physiol Heart Circ Physiol 279:H610–H618PubMedCrossRef
144.
go back to reference Gonzales RJ, Krause DN, Duckles SP (2004) Testosterone suppresses endothelium-dependent dilation of rat middle cerebral arteries. Am J Physiol Heart Circ Physiol 286:H552–H560PubMedCrossRef Gonzales RJ, Krause DN, Duckles SP (2004) Testosterone suppresses endothelium-dependent dilation of rat middle cerebral arteries. Am J Physiol Heart Circ Physiol 286:H552–H560PubMedCrossRef
146.
go back to reference Gibson CL (2013) Cerebral ischemic stroke: is sex important? J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 33:1355–1361CrossRef Gibson CL (2013) Cerebral ischemic stroke: is sex important? J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 33:1355–1361CrossRef
148.
go back to reference Olsen TS, Andersen KK (2010) Female survival advantage relates to male inferiority rather than female superiority: a hypothesis based on the impact of age and stroke severity on 1-week to 1-year case fatality in 40,155 men and women. Gend Med 7:284–295PubMedCrossRef Olsen TS, Andersen KK (2010) Female survival advantage relates to male inferiority rather than female superiority: a hypothesis based on the impact of age and stroke severity on 1-week to 1-year case fatality in 40,155 men and women. Gend Med 7:284–295PubMedCrossRef
150.
151.
go back to reference Halls ML, Bathgate RAD, Sutton SW, Dschietzig TB, Summers RJ (2015) International Union of Basic and Clinical Pharmacology. XCV. Recent advances in the understanding of the pharmacology and biological roles of relaxin family peptide receptors 1–4, the receptors for relaxin family peptides. Pharmacol. Rev 67:389–440PubMedPubMedCentralCrossRef Halls ML, Bathgate RAD, Sutton SW, Dschietzig TB, Summers RJ (2015) International Union of Basic and Clinical Pharmacology. XCV. Recent advances in the understanding of the pharmacology and biological roles of relaxin family peptide receptors 1–4, the receptors for relaxin family peptides. Pharmacol. Rev 67:389–440PubMedPubMedCentralCrossRef
153.
go back to reference Dschietzig T, Bartsch C, Baumann G, Stangl K (2006) Relaxin—a pleiotropic hormone and its emerging role for experimental and clinical therapeutics. Pharmacol Ther 112:38–56PubMedCrossRef Dschietzig T, Bartsch C, Baumann G, Stangl K (2006) Relaxin—a pleiotropic hormone and its emerging role for experimental and clinical therapeutics. Pharmacol Ther 112:38–56PubMedCrossRef
154.
go back to reference Jelinic M et al (2014) Localization of relaxin receptors in arteries and veins, and region-specific increases in compliance and bradykinin-mediated relaxation after in vivo serelaxin treatment. FASEB J Off Publ Fed Am Soc Exp Biol 28:275–287 Jelinic M et al (2014) Localization of relaxin receptors in arteries and veins, and region-specific increases in compliance and bradykinin-mediated relaxation after in vivo serelaxin treatment. FASEB J Off Publ Fed Am Soc Exp Biol 28:275–287
155.
go back to reference Xu Q, Chakravorty A, Bathgate RAD, Dart AM, Du XJ (2010) Relaxin therapy reverses large artery remodeling and improves arterial compliance in senescent spontaneously hypertensive rats. Hypertension 55:1260–1266PubMedCrossRef Xu Q, Chakravorty A, Bathgate RAD, Dart AM, Du XJ (2010) Relaxin therapy reverses large artery remodeling and improves arterial compliance in senescent spontaneously hypertensive rats. Hypertension 55:1260–1266PubMedCrossRef
156.
go back to reference Dschietzig T, Bartsch C, Baumann G, Stangl K (2009) RXFP1-inactive relaxin activates human glucocorticoid receptor: further investigations into the relaxin–GR pathway. Regul Pept 154:77–84PubMedCrossRef Dschietzig T, Bartsch C, Baumann G, Stangl K (2009) RXFP1-inactive relaxin activates human glucocorticoid receptor: further investigations into the relaxin–GR pathway. Regul Pept 154:77–84PubMedCrossRef
157.
158.
go back to reference Yoshida T, Kumagai H, Kohsaka T, Ikegaya N (2013) Relaxin protects against renal ischemia-reperfusion injury. Am J Physiol Renal Physiol 305:F1169–F1176PubMedCrossRef Yoshida T, Kumagai H, Kohsaka T, Ikegaya N (2013) Relaxin protects against renal ischemia-reperfusion injury. Am J Physiol Renal Physiol 305:F1169–F1176PubMedCrossRef
159.
go back to reference Alexiou K, Wilbring M, Matschke K, Dschietzig T (2013) Relaxin protects rat lungs from ischemia-reperfusion injury via inducible NO synthase: role of ERK-1/2, PI3K, and forkhead transcription factor FKHRL1. PLoS One 8:e75592PubMedPubMedCentralCrossRef Alexiou K, Wilbring M, Matschke K, Dschietzig T (2013) Relaxin protects rat lungs from ischemia-reperfusion injury via inducible NO synthase: role of ERK-1/2, PI3K, and forkhead transcription factor FKHRL1. PLoS One 8:e75592PubMedPubMedCentralCrossRef
160.
go back to reference Brecht A, Bartsch C, Baumann G, Stangl K, Dschietzig T (2011) Relaxin inhibits early steps in vascular inflammation. Regul Pept 166:76–82PubMedCrossRef Brecht A, Bartsch C, Baumann G, Stangl K, Dschietzig T (2011) Relaxin inhibits early steps in vascular inflammation. Regul Pept 166:76–82PubMedCrossRef
161.
go back to reference Teerlink JR et al (2013) Serelaxin, recombinant human relaxin-2, for treatment of acute heart failure (RELAX-AHF): a randomised, placebo-controlled trial. Lancet 381:29–39PubMedCrossRef Teerlink JR et al (2013) Serelaxin, recombinant human relaxin-2, for treatment of acute heart failure (RELAX-AHF): a randomised, placebo-controlled trial. Lancet 381:29–39PubMedCrossRef
162.
go back to reference Schöndorf T et al (2007) Relaxin expression correlates significantly with serum changes in VEGF in response to antidiabetic treatment in male patients with type 2 diabetes mellitus. Clin Lab 53:193–198PubMed Schöndorf T et al (2007) Relaxin expression correlates significantly with serum changes in VEGF in response to antidiabetic treatment in male patients with type 2 diabetes mellitus. Clin Lab 53:193–198PubMed
163.
go back to reference Schöndorf T et al (2007) Relaxin expression correlates significantly with serum fibrinogen variation in response to antidiabetic treatment in women with type 2 diabetes mellitus. Gynecol Endocrinol Off J Int Soc Gynecol Endocrinol 23:356–360CrossRef Schöndorf T et al (2007) Relaxin expression correlates significantly with serum fibrinogen variation in response to antidiabetic treatment in women with type 2 diabetes mellitus. Gynecol Endocrinol Off J Int Soc Gynecol Endocrinol 23:356–360CrossRef
164.
go back to reference Leo CH, Jelinic M, Parkington HC, Tare M, Parry LJ (2014) Acute intravenous injection of serelaxin (recombinant human relaxin-2) causes rapid and sustained bradykinin-mediated vasorelaxation. J Am Heart Assoc 3:e000493PubMedPubMedCentralCrossRef Leo CH, Jelinic M, Parkington HC, Tare M, Parry LJ (2014) Acute intravenous injection of serelaxin (recombinant human relaxin-2) causes rapid and sustained bradykinin-mediated vasorelaxation. J Am Heart Assoc 3:e000493PubMedPubMedCentralCrossRef
165.
go back to reference Dschietzig T et al (2003) Relaxin, a pregnancy hormone, is a functional endothelin-1 antagonist: attenuation of endothelin-1-mediated vasoconstriction by stimulation of endothelin type-B receptor expression via ERK-1/2 and nuclear factor-kappaB. Circ Res 92:32–40PubMedCrossRef Dschietzig T et al (2003) Relaxin, a pregnancy hormone, is a functional endothelin-1 antagonist: attenuation of endothelin-1-mediated vasoconstriction by stimulation of endothelin type-B receptor expression via ERK-1/2 and nuclear factor-kappaB. Circ Res 92:32–40PubMedCrossRef
166.
go back to reference Chan S-L, Cipolla MJ (2011) Relaxin causes selective outward remodeling of brain parenchymal arterioles via activation of peroxisome proliferator-activated receptor. FASEB J 25:3229–3239PubMedPubMedCentralCrossRef Chan S-L, Cipolla MJ (2011) Relaxin causes selective outward remodeling of brain parenchymal arterioles via activation of peroxisome proliferator-activated receptor. FASEB J 25:3229–3239PubMedPubMedCentralCrossRef
167.
go back to reference Chan S-L, Sweet JG, Cipolla MJ (2013) Treatment for cerebral small vessel disease: effect of relaxin on the function and structure of cerebral parenchymal arterioles during hypertension. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 27:3917–3927 Chan S-L, Sweet JG, Cipolla MJ (2013) Treatment for cerebral small vessel disease: effect of relaxin on the function and structure of cerebral parenchymal arterioles during hypertension. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 27:3917–3927
169.
go back to reference Wilson BC, Rappaport R (2009) An in vitro study of the protective effect of relaxin on brain tissue under ischemic stress. Ann N Y Acad Sci 1160:265–268PubMedCrossRef Wilson BC, Rappaport R (2009) An in vitro study of the protective effect of relaxin on brain tissue under ischemic stress. Ann N Y Acad Sci 1160:265–268PubMedCrossRef
170.
go back to reference Wilson BC (2005) Relaxin pretreatment decreases infarct size in male rats after middle cerebral artery occlusion. Ann N Y Acad Sci 1041:223–228PubMedCrossRef Wilson BC (2005) Relaxin pretreatment decreases infarct size in male rats after middle cerebral artery occlusion. Ann N Y Acad Sci 1041:223–228PubMedCrossRef
171.
go back to reference Wilson BC, Connell B, Saleh TM (2006) Relaxin-induced reduction of infarct size in male rats receiving MCAO is dependent on nitric oxide synthesis and not estrogenic mechanisms. Neurosci Lett 393:160–164PubMedCrossRef Wilson BC, Connell B, Saleh TM (2006) Relaxin-induced reduction of infarct size in male rats receiving MCAO is dependent on nitric oxide synthesis and not estrogenic mechanisms. Neurosci Lett 393:160–164PubMedCrossRef
172.
go back to reference Bergeron LH et al (2015) Relaxin peptide hormones are protective during the early stages of ischemic stroke in male rats. Endocrinology 156:638–646PubMedCrossRef Bergeron LH et al (2015) Relaxin peptide hormones are protective during the early stages of ischemic stroke in male rats. Endocrinology 156:638–646PubMedCrossRef
173.
go back to reference Milia P et al (2013) Efficacy of relaxin on functional recovery of post stroke patients. Ital J Anat Embryol Arch Ital Anat Ed Embriologia 118:92–97 Milia P et al (2013) Efficacy of relaxin on functional recovery of post stroke patients. Ital J Anat Embryol Arch Ital Anat Ed Embriologia 118:92–97
174.
go back to reference Savopoulos C, Michalakis K, Apostolopoulou M, Miras A, Hatzitolios A (2011) Adipokines and stroke: a review of the literature. Maturitas 70:322–327PubMedCrossRef Savopoulos C, Michalakis K, Apostolopoulou M, Miras A, Hatzitolios A (2011) Adipokines and stroke: a review of the literature. Maturitas 70:322–327PubMedCrossRef
175.
go back to reference Binder C et al (2014) Relaxins enhance growth of spontaneous murine breast cancers as well as metastatic colonization of the brain. Clin Exp Metastasis 31:57–65PubMedCrossRef Binder C et al (2014) Relaxins enhance growth of spontaneous murine breast cancers as well as metastatic colonization of the brain. Clin Exp Metastasis 31:57–65PubMedCrossRef
176.
go back to reference Lamp O et al (2013) The metastatic potential of canine mammary tumours can be assessed by mRNA expression analysis of connective tissue modulators. Vet Comp Oncol 11:70–85PubMedCrossRef Lamp O et al (2013) The metastatic potential of canine mammary tumours can be assessed by mRNA expression analysis of connective tissue modulators. Vet Comp Oncol 11:70–85PubMedCrossRef
Metadata
Title
Ischemic stroke and select adipose-derived and sex hormones: a review
Author
Kristy L. Meadows
Publication date
01-06-2018
Publisher
Springer International Publishing
Published in
Hormones / Issue 2/2018
Print ISSN: 1109-3099
Electronic ISSN: 2520-8721
DOI
https://doi.org/10.1007/s42000-018-0034-4

Other articles of this Issue 2/2018

Hormones 2/2018 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.