Skip to main content
Top
Published in: Journal of Translational Medicine 1/2020

Open Access 01-12-2020 | Insulins | Review

Is insulin-like growth factor-1 involved in Parkinson’s disease development?

Authors: Inma Castilla-Cortázar, Gabriel A. Aguirre, Giovana Femat-Roldán, Irene Martín-Estal, Luis Espinosa

Published in: Journal of Translational Medicine | Issue 1/2020

Login to get access

Abstract

Parkinson’s disease (PD) is a neurodegenerative disorder that results in the death of dopaminergic neurons within the substantia nigra pars compacta and the reduction in dopaminergic control over striatal output neurons, leading to a movement disorder most commonly characterized by akinesia or bradykinesia, rigidity and tremor. Also, PD is less frequently depicted by sensory symptoms (pain and tingling), hyposmia, sleep alterations, depression and anxiety, and abnormal executive and working memory related functions. On the other hand, insulin-like growth factor 1 (IGF-1) is an endocrine, paracrine and autocrine hormone with several functions including tissue growth and development, insulin-like activity, proliferation, pro-survival, anti-aging, antioxidant and neuroprotection, among others. Herein this review tries to summarize all experimental and clinical data to understand the pathophysiology and development of PD, as well as its clear association with IGF-1, supported by several lines of evidence: (1) IGF-1 decreases with age, while aging is the major risk for PD establishment and development; (2) numerous basic and translational data have appointed direct protective and homeostasis IGF-1 roles in all brain cells; (3) estrogens seem to confer women strong protection to PD via IGF-1; and (4) clinical correlations in PD cohorts have confirmed elevated IGF-1 levels at the onset of the disease, suggesting an ongoing compensatory or “fight-to-injury” mechanism.
Literature
2.
go back to reference Erkkinen MG, Kim M-O, Geschwind MD. Clinical neurology and epidemiology of the major neurodegenerative diseases. Cold Spring Harb Perspect Biol. 2018;10(4):a033118.PubMedCrossRefPubMedCentral Erkkinen MG, Kim M-O, Geschwind MD. Clinical neurology and epidemiology of the major neurodegenerative diseases. Cold Spring Harb Perspect Biol. 2018;10(4):a033118.PubMedCrossRefPubMedCentral
3.
go back to reference GBD 2016 Parkinson’s Disease Collaborators. Global, regional, and national burden of Parkinson’s disease, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2018;17(11):939–53.CrossRef GBD 2016 Parkinson’s Disease Collaborators. Global, regional, and national burden of Parkinson’s disease, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2018;17(11):939–53.CrossRef
4.
go back to reference Jankovic J. Parkinson’s disease: clinical features and diagnosis. J Neurol Neurosurg Psychiatry. 2008;79(4):368–76.PubMedCrossRef Jankovic J. Parkinson’s disease: clinical features and diagnosis. J Neurol Neurosurg Psychiatry. 2008;79(4):368–76.PubMedCrossRef
5.
go back to reference Rodriguez-Oroz MC, Jahanshahi M, Krack P, Litvan I, Macias R, Bezard E, et al. Initial clinical manifestations of Parkinson’s disease: features and pathophysiological mechanisms. Lancet Neurol. 2009;8(12):1128–39.PubMedCrossRef Rodriguez-Oroz MC, Jahanshahi M, Krack P, Litvan I, Macias R, Bezard E, et al. Initial clinical manifestations of Parkinson’s disease: features and pathophysiological mechanisms. Lancet Neurol. 2009;8(12):1128–39.PubMedCrossRef
9.
go back to reference Berg D, Lang AE, Postuma RB, Maetzler W, Deuschl G, Gasser T, et al. Changing the research criteria for the diagnosis of Parkinson’s disease: obstacles and opportunities. Lancet Neurol. 2013;12(5):514–24.PubMedCrossRef Berg D, Lang AE, Postuma RB, Maetzler W, Deuschl G, Gasser T, et al. Changing the research criteria for the diagnosis of Parkinson’s disease: obstacles and opportunities. Lancet Neurol. 2013;12(5):514–24.PubMedCrossRef
11.
go back to reference Juri C, Rodríguez-Oroz MC, Burguera JA, Guridi J, Obeso JA. Pain and dyskinesia in Parkinson’s disease. Mov Disord. 2010;25(1):130–2.PubMedCrossRef Juri C, Rodríguez-Oroz MC, Burguera JA, Guridi J, Obeso JA. Pain and dyskinesia in Parkinson’s disease. Mov Disord. 2010;25(1):130–2.PubMedCrossRef
12.
go back to reference Sabatini U, Boulanouar K, Fabre N, Martin F, Carel C, Colonnese C, et al. Cortical motor reorganization in akinetic patients with Parkinson’s disease: a functional MRI study. Brain. 2000;123(Pt 2):394–403.PubMedCrossRef Sabatini U, Boulanouar K, Fabre N, Martin F, Carel C, Colonnese C, et al. Cortical motor reorganization in akinetic patients with Parkinson’s disease: a functional MRI study. Brain. 2000;123(Pt 2):394–403.PubMedCrossRef
13.
go back to reference Olanow CW. The pathogenesis of cell death in Parkinson’s disease—2007. Mov Disord. 2007;22(SUPPL. 17):S335–42.PubMedCrossRef Olanow CW. The pathogenesis of cell death in Parkinson’s disease—2007. Mov Disord. 2007;22(SUPPL. 17):S335–42.PubMedCrossRef
14.
go back to reference Braak H, Ghebremedhin E, Rüb U, Bratzke H, Del Tredici K. Stages in the development of Parkinson’s disease-related pathology. Cell Tissue Res. 2004;318(1):121–34.PubMedCrossRef Braak H, Ghebremedhin E, Rüb U, Bratzke H, Del Tredici K. Stages in the development of Parkinson’s disease-related pathology. Cell Tissue Res. 2004;318(1):121–34.PubMedCrossRef
15.
go back to reference Mink JW, Blumenschine RJ, Adams DB. Ratio of central nervous system to body metabolism in vertebrates: its constancy and functional basis. Am J Physiol. 1981;241(3):R203–12.PubMed Mink JW, Blumenschine RJ, Adams DB. Ratio of central nervous system to body metabolism in vertebrates: its constancy and functional basis. Am J Physiol. 1981;241(3):R203–12.PubMed
16.
go back to reference Connor JR, Benkovic SA. Iron regulation in the brain: histochemical, biochemical, and molecular considerations. Ann Neurol. 1992;32(Suppl):S51–61.PubMedCrossRef Connor JR, Benkovic SA. Iron regulation in the brain: histochemical, biochemical, and molecular considerations. Ann Neurol. 1992;32(Suppl):S51–61.PubMedCrossRef
17.
go back to reference Mills E, Dong X-P, Wang F, Xu H. Mechanisms of brain iron transport: insight into neurodegeneration and CNS disorders. Future Med Chem. 2010;2(1):51–64.PubMedCrossRef Mills E, Dong X-P, Wang F, Xu H. Mechanisms of brain iron transport: insight into neurodegeneration and CNS disorders. Future Med Chem. 2010;2(1):51–64.PubMedCrossRef
18.
go back to reference Patel NK, Pavese N, Javed S, Hotton GR, Brooks DJ, Gill SS. Benefits of putaminal GDNF infusion in Parkinson disease are maintained after GDNF cessation. Neurology. 2013;81(13):1176–8.PubMedPubMedCentralCrossRef Patel NK, Pavese N, Javed S, Hotton GR, Brooks DJ, Gill SS. Benefits of putaminal GDNF infusion in Parkinson disease are maintained after GDNF cessation. Neurology. 2013;81(13):1176–8.PubMedPubMedCentralCrossRef
19.
go back to reference Parain K, Murer MG, Yan Q, Faucheux B, Agid Y, Hirsch E, et al. Reduced expression of brain-derived neurotrophic factor protein in Parkinson’s disease substantia nigra. Neuroreport. 1999;10(3):557–61.PubMedCrossRef Parain K, Murer MG, Yan Q, Faucheux B, Agid Y, Hirsch E, et al. Reduced expression of brain-derived neurotrophic factor protein in Parkinson’s disease substantia nigra. Neuroreport. 1999;10(3):557–61.PubMedCrossRef
20.
go back to reference Pringsheim T, Jette N, Frolkis A, Steeves TDL. The prevalence of Parkinson’s disease: a systematic review and meta-analysis. Mov Disord. 2014;29(13):1583–90.PubMedCrossRef Pringsheim T, Jette N, Frolkis A, Steeves TDL. The prevalence of Parkinson’s disease: a systematic review and meta-analysis. Mov Disord. 2014;29(13):1583–90.PubMedCrossRef
21.
go back to reference Hirsch L, Jette N, Frolkis A, Steeves T, Pringsheim T. The incidence of Parkinson’s disease: a systematic review and meta-analysis. Neuroepidemiology. 2016;46(4):292–300.PubMedCrossRef Hirsch L, Jette N, Frolkis A, Steeves T, Pringsheim T. The incidence of Parkinson’s disease: a systematic review and meta-analysis. Neuroepidemiology. 2016;46(4):292–300.PubMedCrossRef
22.
go back to reference Van Den Eeden SK. Incidence of Parkinson’s disease: variation by age, gender, and race/ethnicity. Am J Epidemiol. 2003;157(11):1015–22.CrossRef Van Den Eeden SK. Incidence of Parkinson’s disease: variation by age, gender, and race/ethnicity. Am J Epidemiol. 2003;157(11):1015–22.CrossRef
23.
go back to reference Rodríguez-Violante M, Villar-Velarde A, Valencia-Ramos C, Cervantes-Arriaga A. Características epidemiológicas de pacientes con enfermedad de Parkinson Arch Neurocien (Mex) INNN. Arch Neurocien. 2011;16:64–8. Rodríguez-Violante M, Villar-Velarde A, Valencia-Ramos C, Cervantes-Arriaga A. Características epidemiológicas de pacientes con enfermedad de Parkinson Arch Neurocien (Mex) INNN. Arch Neurocien. 2011;16:64–8.
25.
go back to reference Song Y, Gu Z, An J, Chan P. Gender differences on motor and non-motor symptoms of de novo patients with early Parkinson’s disease. Neurol Sci. 2014;35(12):1991–6.PubMedCrossRef Song Y, Gu Z, An J, Chan P. Gender differences on motor and non-motor symptoms of de novo patients with early Parkinson’s disease. Neurol Sci. 2014;35(12):1991–6.PubMedCrossRef
26.
go back to reference Yamawaki M, Kusumi M, Kowa H, Nakashima K. Changes in prevalence and incidence of Parkinson’s disease in Japan during a quarter of a century. Neuroepidemiology. 2009;32(4):263–9.PubMedCrossRef Yamawaki M, Kusumi M, Kowa H, Nakashima K. Changes in prevalence and incidence of Parkinson’s disease in Japan during a quarter of a century. Neuroepidemiology. 2009;32(4):263–9.PubMedCrossRef
28.
go back to reference Alves G, Müller B, Herlofson K, HogenEsch I, Telstad W, Aarsland D, et al. Incidence of Parkinson’s disease in Norway: the Norwegian ParkWest study. J Neurol Neurosurg Psychiatry. 2009;80(8):851–7.PubMedCrossRef Alves G, Müller B, Herlofson K, HogenEsch I, Telstad W, Aarsland D, et al. Incidence of Parkinson’s disease in Norway: the Norwegian ParkWest study. J Neurol Neurosurg Psychiatry. 2009;80(8):851–7.PubMedCrossRef
29.
go back to reference Kuiper GGJM, Carlsson B, Grandien K, Enmark E, Häggblad J, Nilsson S, et al. Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors α and β. Endocrinology. 1997;138(3):863–70.PubMedCrossRef Kuiper GGJM, Carlsson B, Grandien K, Enmark E, Häggblad J, Nilsson S, et al. Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors α and β. Endocrinology. 1997;138(3):863–70.PubMedCrossRef
32.
go back to reference Vermeulen A, Kaufman JM, Goemaere S, Van Pottelberg I. Estradiol in elderly men. Aging Male. 2002;5(2):98–102.PubMedCrossRef Vermeulen A, Kaufman JM, Goemaere S, Van Pottelberg I. Estradiol in elderly men. Aging Male. 2002;5(2):98–102.PubMedCrossRef
34.
go back to reference Gaignard P, Liere P, Thérond P, Schumacher M, Slama A, Guennoun R. Role of sex hormones on brain mitochondrial function, with special reference to aging and neurodegenerative diseases. Front Aging Neurosci. 2017;9:406.PubMedPubMedCentralCrossRef Gaignard P, Liere P, Thérond P, Schumacher M, Slama A, Guennoun R. Role of sex hormones on brain mitochondrial function, with special reference to aging and neurodegenerative diseases. Front Aging Neurosci. 2017;9:406.PubMedPubMedCentralCrossRef
35.
go back to reference Mcmahon T, Van Zijl PCM, Gilad AA. NIH Public Access. 2015;27(3):320–31. Mcmahon T, Van Zijl PCM, Gilad AA. NIH Public Access. 2015;27(3):320–31.
36.
go back to reference González C, Díaz F, Alonso A. Neuroprotective effects of estrogens: cross-talk between estrogen and intracellular insulin signalling. Infect Disord Drug Targets. 2008;8(1):65–7.PubMedCrossRef González C, Díaz F, Alonso A. Neuroprotective effects of estrogens: cross-talk between estrogen and intracellular insulin signalling. Infect Disord Drug Targets. 2008;8(1):65–7.PubMedCrossRef
38.
go back to reference Labandeira-Garcia JL, Rodriguez-Perez AI, Valenzuela R, Costa-Besada MA, Guerra MJ. Menopause and Parkinson’s disease. Interaction between estrogens and brain renin–angiotensin system in dopaminergic degeneration. Front Neuroendocrinol. 2016;2016(43):44–59.CrossRef Labandeira-Garcia JL, Rodriguez-Perez AI, Valenzuela R, Costa-Besada MA, Guerra MJ. Menopause and Parkinson’s disease. Interaction between estrogens and brain renin–angiotensin system in dopaminergic degeneration. Front Neuroendocrinol. 2016;2016(43):44–59.CrossRef
39.
go back to reference Haaxma CA, Bloem BR, Borm GF, Oyen WJG, Leenders KL, Eshuis S, et al. Gender differences in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2007;78(8):819–24.PubMedCrossRef Haaxma CA, Bloem BR, Borm GF, Oyen WJG, Leenders KL, Eshuis S, et al. Gender differences in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2007;78(8):819–24.PubMedCrossRef
40.
go back to reference Hirohata M, Ono K, Morinaga A, Ikeda T, Yamada M. Anti-aggregation and fibril-destabilizing effects of sex hormones on α-synuclein fibrils in vitro. Exp Neurol. 2009;217(2):434–9.PubMedCrossRef Hirohata M, Ono K, Morinaga A, Ikeda T, Yamada M. Anti-aggregation and fibril-destabilizing effects of sex hormones on α-synuclein fibrils in vitro. Exp Neurol. 2009;217(2):434–9.PubMedCrossRef
43.
go back to reference Nussbaum RL, Ellis CE. Alzheimer’s disease and Parkinson’s Disease. N Engl J Med. 2003;348:1356–64.PubMedCrossRef Nussbaum RL, Ellis CE. Alzheimer’s disease and Parkinson’s Disease. N Engl J Med. 2003;348:1356–64.PubMedCrossRef
46.
go back to reference Von Campenhausen S, Bornschein B, Wick R, Bötzel K, Sampaio C, Poewe W, et al. Prevalence and incidence of Parkinson’s disease in Europe. Eur Neuropsychopharmacol. 2005;15(4):473–90.CrossRef Von Campenhausen S, Bornschein B, Wick R, Bötzel K, Sampaio C, Poewe W, et al. Prevalence and incidence of Parkinson’s disease in Europe. Eur Neuropsychopharmacol. 2005;15(4):473–90.CrossRef
47.
go back to reference Picillo M, Nicoletti A, Fetoni V, Garavaglia B, Barone P, Pellecchia MT. The relevance of gender in Parkinson’s disease: a review. J Neurol. 2017;264(8):1583–607.PubMedCrossRef Picillo M, Nicoletti A, Fetoni V, Garavaglia B, Barone P, Pellecchia MT. The relevance of gender in Parkinson’s disease: a review. J Neurol. 2017;264(8):1583–607.PubMedCrossRef
48.
go back to reference Smith KM, Dahodwala N. Sex differences in Parkinson’s disease and other movement disorders. Exp Neurol. 2014;259:44–56.PubMedCrossRef Smith KM, Dahodwala N. Sex differences in Parkinson’s disease and other movement disorders. Exp Neurol. 2014;259:44–56.PubMedCrossRef
49.
go back to reference Le Roith D. Seminars in medicine of the Beth Israel Deaconess Medical Center. Insulin-like growth factors. N Engl J Med. 1997;336(9):633–40.PubMedCrossRef Le Roith D. Seminars in medicine of the Beth Israel Deaconess Medical Center. Insulin-like growth factors. N Engl J Med. 1997;336(9):633–40.PubMedCrossRef
50.
go back to reference Martín-Estal I, De la Garza R, Castilla-Cortazar I. Intrauterine Growth Retardation (IUGR) as a novel condition deficiency of Insulin-like growth. Retard Intrauter Growth. 2015;1(1):1. Martín-Estal I, De la Garza R, Castilla-Cortazar I. Intrauterine Growth Retardation (IUGR) as a novel condition deficiency of Insulin-like growth. Retard Intrauter Growth. 2015;1(1):1.
51.
go back to reference D’Ercole AJ, Applewhite GT, Underwood LE. Evidence that somatomedin is synthesized by multiple tissues in the fetus. Dev Biol. 1980;75:315–28.PubMedCrossRef D’Ercole AJ, Applewhite GT, Underwood LE. Evidence that somatomedin is synthesized by multiple tissues in the fetus. Dev Biol. 1980;75:315–28.PubMedCrossRef
53.
go back to reference Martín-Estal I, de la Garza RG, Castilla-Cortázar I. Intrauterine growth retardation (IUGR) as a novel condition of insulin-like growth factor-1 (IGF-1) deficiency. Rev Physiol Biochem Pharmacol. 2016;4(170):1–35. Martín-Estal I, de la Garza RG, Castilla-Cortázar I. Intrauterine growth retardation (IUGR) as a novel condition of insulin-like growth factor-1 (IGF-1) deficiency. Rev Physiol Biochem Pharmacol. 2016;4(170):1–35.
54.
go back to reference Aguirre GA, De Ita JR, de la Garza RG, Castilla-Cortazar I. Insulin-like growth factor-1 deficiency and metabolic syndrome. J Transl Med. 2016;14(1):3.PubMedPubMedCentralCrossRef Aguirre GA, De Ita JR, de la Garza RG, Castilla-Cortazar I. Insulin-like growth factor-1 deficiency and metabolic syndrome. J Transl Med. 2016;14(1):3.PubMedPubMedCentralCrossRef
55.
go back to reference Bondy CA, Lee WH. Patterns of insulin-like growth factor and IGF receptor gene expression in the brain. Functional implications. Ann N Y Acad Sci. 1993;692:33–43.PubMedCrossRef Bondy CA, Lee WH. Patterns of insulin-like growth factor and IGF receptor gene expression in the brain. Functional implications. Ann N Y Acad Sci. 1993;692:33–43.PubMedCrossRef
56.
go back to reference Bach MA, Shen-Orr Z, Lowe WL, Roberts CT, LeRoith D. Insulin-like growth factor I mRNA levels are developmentally regulated in specific regions of the rat brain. Brain Res Mol Brain Res. 1991;10(1):43–8.PubMedCrossRef Bach MA, Shen-Orr Z, Lowe WL, Roberts CT, LeRoith D. Insulin-like growth factor I mRNA levels are developmentally regulated in specific regions of the rat brain. Brain Res Mol Brain Res. 1991;10(1):43–8.PubMedCrossRef
57.
go back to reference Popken GJ, Dechert-Zeger M, Ye P, D’Ercole AJ. Brain development. Adv Exp Med Biol. 2005;567:187–220.PubMedCrossRef Popken GJ, Dechert-Zeger M, Ye P, D’Ercole AJ. Brain development. Adv Exp Med Biol. 2005;567:187–220.PubMedCrossRef
58.
go back to reference Baron-Van Evercooren A, Olichon-Berthe C, Kowalski A, Visciano G, Van Obberghen E. Expression of IGF-I and insulin receptor genes in the rat central nervous system: a developmental, regional, and cellular analysis. J Neurosci Res. 1991;28(2):244–53.PubMedCrossRef Baron-Van Evercooren A, Olichon-Berthe C, Kowalski A, Visciano G, Van Obberghen E. Expression of IGF-I and insulin receptor genes in the rat central nervous system: a developmental, regional, and cellular analysis. J Neurosci Res. 1991;28(2):244–53.PubMedCrossRef
59.
go back to reference Reinhardt RR, Bondy CA. Insulin-like growth factors cross the blood-brain barrier. Endocrinology. 1994;135(5):1753–61.PubMedCrossRef Reinhardt RR, Bondy CA. Insulin-like growth factors cross the blood-brain barrier. Endocrinology. 1994;135(5):1753–61.PubMedCrossRef
60.
go back to reference Gong X, Ma M, Fan X, Li M, Liu Q, Liu X, et al. Down-regulation of IGF-1/IGF-1R in hippocampus of rats with vascular dementia. Neurosci Lett. 2012;513(1):20–4.PubMedCrossRef Gong X, Ma M, Fan X, Li M, Liu Q, Liu X, et al. Down-regulation of IGF-1/IGF-1R in hippocampus of rats with vascular dementia. Neurosci Lett. 2012;513(1):20–4.PubMedCrossRef
61.
go back to reference Sonntag WE, Ramsey M, Carter CS. Growth hormone and insulin-like growth factor-1 (IGF-1) and their influence on cognitive aging. Ageing Res Rev. 2005;4(2):195–212.PubMedCrossRef Sonntag WE, Ramsey M, Carter CS. Growth hormone and insulin-like growth factor-1 (IGF-1) and their influence on cognitive aging. Ageing Res Rev. 2005;4(2):195–212.PubMedCrossRef
62.
go back to reference Adem A, Jossan SS, D’Argy R, Gillberg PG, Nordberg A, Winblad B, et al. Insulin-like growth factor 1 (IGF-1) receptors in the human brain: quantitative autoradiographic localization. Brain Res. 1989;503(2):299–303.PubMedCrossRef Adem A, Jossan SS, D’Argy R, Gillberg PG, Nordberg A, Winblad B, et al. Insulin-like growth factor 1 (IGF-1) receptors in the human brain: quantitative autoradiographic localization. Brain Res. 1989;503(2):299–303.PubMedCrossRef
64.
go back to reference Tong M, Dong M, de la Monte SM. Brain insulin-like growth factor and neurotrophin resistance in Parkinson’s disease and dementia with Lewy bodies: potential role of manganese neurotoxicity. J Alzheimers Dis. 2009;16(3):585–99.PubMedPubMedCentralCrossRef Tong M, Dong M, de la Monte SM. Brain insulin-like growth factor and neurotrophin resistance in Parkinson’s disease and dementia with Lewy bodies: potential role of manganese neurotoxicity. J Alzheimers Dis. 2009;16(3):585–99.PubMedPubMedCentralCrossRef
66.
go back to reference Morris JK, Zhang H, Gupte AA, Bomhoff GL, Stanford JA, Geiger PC. Measures of striatal insulin resistance in a 6-hydroxydopamine model of Parkinson’s disease. Brain Res. 2008;1240:185–95.PubMedPubMedCentralCrossRef Morris JK, Zhang H, Gupte AA, Bomhoff GL, Stanford JA, Geiger PC. Measures of striatal insulin resistance in a 6-hydroxydopamine model of Parkinson’s disease. Brain Res. 2008;1240:185–95.PubMedPubMedCentralCrossRef
67.
go back to reference Talbot K, Wang H-Y, Kazi H, Han L-Y, Bakshi KP, Stucky A, et al. Demonstrated brain insulin resistance in Alzheimer’s disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J Clin Investig. 2012;122(4):1316–38.PubMedCrossRefPubMedCentral Talbot K, Wang H-Y, Kazi H, Han L-Y, Bakshi KP, Stucky A, et al. Demonstrated brain insulin resistance in Alzheimer’s disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J Clin Investig. 2012;122(4):1316–38.PubMedCrossRefPubMedCentral
68.
go back to reference Fernandez AM, Torres-Alemán I. The many faces of insulin-like peptide signalling in the brain. Nat Rev Neurosci. 2012;13(4):225–39.PubMedCrossRef Fernandez AM, Torres-Alemán I. The many faces of insulin-like peptide signalling in the brain. Nat Rev Neurosci. 2012;13(4):225–39.PubMedCrossRef
69.
go back to reference Bäckström M, Hall K, Sara V. Somatomedin levels in cerebrospinal fluid from adults with pituitary disorders. Acta Endocrinol. 1984;107(2):171–8.CrossRef Bäckström M, Hall K, Sara V. Somatomedin levels in cerebrospinal fluid from adults with pituitary disorders. Acta Endocrinol. 1984;107(2):171–8.CrossRef
70.
go back to reference Carro E, Trejo JL, Gomez-Isla T, LeRoith D, Torres-Aleman I. Serum insulin-like growth factor I regulates brain amyloid-beta levels. Nat Med. 2002;8:1390–7.PubMedCrossRef Carro E, Trejo JL, Gomez-Isla T, LeRoith D, Torres-Aleman I. Serum insulin-like growth factor I regulates brain amyloid-beta levels. Nat Med. 2002;8:1390–7.PubMedCrossRef
71.
go back to reference Kalmar B, Greensmith L. Induction of heat shock proteins for protection against oxidative stress. Adv Drug Deliv Rev. 2009;61(4):310–8.PubMedCrossRef Kalmar B, Greensmith L. Induction of heat shock proteins for protection against oxidative stress. Adv Drug Deliv Rev. 2009;61(4):310–8.PubMedCrossRef
72.
go back to reference Garcia-Fernandez M, Sierra I, Puche JE, Guerra L, Castilla-Cortazar I. Liver mitochondrial dysfunction is reverted by insulin-like growth factor II (IGF-II) in aging rats. J Transl Med. 2011;9:123.PubMedPubMedCentralCrossRef Garcia-Fernandez M, Sierra I, Puche JE, Guerra L, Castilla-Cortazar I. Liver mitochondrial dysfunction is reverted by insulin-like growth factor II (IGF-II) in aging rats. J Transl Med. 2011;9:123.PubMedPubMedCentralCrossRef
73.
go back to reference Castilla-Cortázar I, García-Fernández M, Delgado G, Puche JE, Sierra I, Barhoum R, et al. Hepatoprotection and neuroprotection induced by low doses of IGF-II in aging rats. J Transl Med. 2011;9:103.PubMedPubMedCentralCrossRef Castilla-Cortázar I, García-Fernández M, Delgado G, Puche JE, Sierra I, Barhoum R, et al. Hepatoprotection and neuroprotection induced by low doses of IGF-II in aging rats. J Transl Med. 2011;9:103.PubMedPubMedCentralCrossRef
74.
go back to reference Duffy KR, Pardridge WM, Rosenfeld RG. Human blood-brain barrier insulin-like growth factor receptor. Metabolism. 1988;37(2):136–40.PubMedCrossRef Duffy KR, Pardridge WM, Rosenfeld RG. Human blood-brain barrier insulin-like growth factor receptor. Metabolism. 1988;37(2):136–40.PubMedCrossRef
75.
go back to reference Dyer AH, Vahdatpour C, Sanfeliu A, Tropea D. The role of Insulin-Like Growth Factor 1 (IGF-1) in brain development, maturation and neuroplasticity. Neuroscience. 2016;325:89–99.PubMedCrossRef Dyer AH, Vahdatpour C, Sanfeliu A, Tropea D. The role of Insulin-Like Growth Factor 1 (IGF-1) in brain development, maturation and neuroplasticity. Neuroscience. 2016;325:89–99.PubMedCrossRef
76.
77.
go back to reference Ye P, Li L, Richards RG, DiAugustine RP, D’Ercole AJ. Myelination is altered in insulin-like growth factor-I null mutant mice. J Neurosci. 2002;22(14):6041–51.PubMedPubMedCentralCrossRef Ye P, Li L, Richards RG, DiAugustine RP, D’Ercole AJ. Myelination is altered in insulin-like growth factor-I null mutant mice. J Neurosci. 2002;22(14):6041–51.PubMedPubMedCentralCrossRef
78.
go back to reference Popken GJ, Hodge RD, Ye P, Zhang J, Ng W, O’Kusky JR, et al. In vivo effects of insulin-like growth factor-I (IGF-I) on prenatal and early postnatal development of the central nervous system. Eur J Neurosci. 2004;19(8):2056–68.PubMedCrossRef Popken GJ, Hodge RD, Ye P, Zhang J, Ng W, O’Kusky JR, et al. In vivo effects of insulin-like growth factor-I (IGF-I) on prenatal and early postnatal development of the central nervous system. Eur J Neurosci. 2004;19(8):2056–68.PubMedCrossRef
79.
go back to reference Ye P, D’Ercole AJ. Insulin-like growth factor actions during development of neural stem cells and progenitors in the central nervous system. J Neurosci Res. 2006;83(1):1–6.PubMedCrossRef Ye P, D’Ercole AJ. Insulin-like growth factor actions during development of neural stem cells and progenitors in the central nervous system. J Neurosci Res. 2006;83(1):1–6.PubMedCrossRef
81.
go back to reference Huat TJ, Khan AA, Pati S, Mustafa Z, Abdullah JM, Jaafar H. IGF-1 enhances cell proliferation and survival during early differentiation of mesenchymal stem cells to neural progenitor-like cells. BMC Neurosci. 2014;15:91.PubMedPubMedCentralCrossRef Huat TJ, Khan AA, Pati S, Mustafa Z, Abdullah JM, Jaafar H. IGF-1 enhances cell proliferation and survival during early differentiation of mesenchymal stem cells to neural progenitor-like cells. BMC Neurosci. 2014;15:91.PubMedPubMedCentralCrossRef
82.
go back to reference Aberg ND, Brywe KG, Isgaard J. Aspects of growth hormone and insulin-like growth factor-I related to neuroprotection, regeneration, and functional plasticity in the adult brain. Sci World J. 2006;6:53–80.CrossRef Aberg ND, Brywe KG, Isgaard J. Aspects of growth hormone and insulin-like growth factor-I related to neuroprotection, regeneration, and functional plasticity in the adult brain. Sci World J. 2006;6:53–80.CrossRef
83.
go back to reference O’Kusky JR, Ye P, D’Ercole AJ. Insulin-like growth factor-I promotes neurogenesis and synaptogenesis in the hippocampal dentate gyrus during postnatal development. J Neurosci. 2000;20(22):8435–42.PubMedPubMedCentralCrossRef O’Kusky JR, Ye P, D’Ercole AJ. Insulin-like growth factor-I promotes neurogenesis and synaptogenesis in the hippocampal dentate gyrus during postnatal development. J Neurosci. 2000;20(22):8435–42.PubMedPubMedCentralCrossRef
84.
go back to reference Hurtado-Chong A, Yusta-Boyo MJ, Vergaño-Vera E, Bulfone A, de Pablo F, Vicario-Abejón C. IGF-I promotes neuronal migration and positioning in the olfactory bulb and the exit of neuroblasts from the subventricular zone. Eur J Neurosci. 2009;30(5):742–55.PubMedCrossRef Hurtado-Chong A, Yusta-Boyo MJ, Vergaño-Vera E, Bulfone A, de Pablo F, Vicario-Abejón C. IGF-I promotes neuronal migration and positioning in the olfactory bulb and the exit of neuroblasts from the subventricular zone. Eur J Neurosci. 2009;30(5):742–55.PubMedCrossRef
85.
go back to reference Oishi K, Watatani K, Itoh Y, Okano H, Guillemot F, Nakajima K, et al. Selective induction of neocortical GABAergic neurons by the PDK1-Akt pathway through activation of Mash1. Proc Natl Acad Sci USA. 2009;106(31):13064–9.PubMedCrossRefPubMedCentral Oishi K, Watatani K, Itoh Y, Okano H, Guillemot F, Nakajima K, et al. Selective induction of neocortical GABAergic neurons by the PDK1-Akt pathway through activation of Mash1. Proc Natl Acad Sci USA. 2009;106(31):13064–9.PubMedCrossRefPubMedCentral
86.
go back to reference Trejo JL, Llorens-Martín MV, Torres-Alemán I. The effects of exercise on spatial learning and anxiety-like behavior are mediated by an IGF-I-dependent mechanism related to hippocampal neurogenesis. Mol Cell Neurosci. 2008;37(2):402–11.PubMedCrossRef Trejo JL, Llorens-Martín MV, Torres-Alemán I. The effects of exercise on spatial learning and anxiety-like behavior are mediated by an IGF-I-dependent mechanism related to hippocampal neurogenesis. Mol Cell Neurosci. 2008;37(2):402–11.PubMedCrossRef
87.
go back to reference Madathil SK, Evans HN, Saatman KE. Temporal and regional changes in IGF-1/IGF-1R signaling in the mouse brain after traumatic brain injury. J Neurotrauma. 2010;27(1):95–107.PubMedCrossRef Madathil SK, Evans HN, Saatman KE. Temporal and regional changes in IGF-1/IGF-1R signaling in the mouse brain after traumatic brain injury. J Neurotrauma. 2010;27(1):95–107.PubMedCrossRef
88.
go back to reference Sandberg Nordqvist AC, von Holst H, Holmin S, Sara VR, Bellander BM, Schalling M. Increase of insulin-like growth factor (IGF)-1, IGF binding protein-2 and -4 mRNAs following cerebral contusion. Brain Res Mol Brain Res. 1996;38(2):285–93.PubMedCrossRef Sandberg Nordqvist AC, von Holst H, Holmin S, Sara VR, Bellander BM, Schalling M. Increase of insulin-like growth factor (IGF)-1, IGF binding protein-2 and -4 mRNAs following cerebral contusion. Brain Res Mol Brain Res. 1996;38(2):285–93.PubMedCrossRef
89.
go back to reference Wang W, Li D, Li Q, Wang L, Bai G, Yang T, et al. Erythropoietin promotes peripheral nerve regeneration in rats by upregulating expression of insulin-like growth factor-1. Arch Med Sci. 2015;11(2):433–7.PubMedPubMedCentralCrossRef Wang W, Li D, Li Q, Wang L, Bai G, Yang T, et al. Erythropoietin promotes peripheral nerve regeneration in rats by upregulating expression of insulin-like growth factor-1. Arch Med Sci. 2015;11(2):433–7.PubMedPubMedCentralCrossRef
90.
go back to reference Isgaard J, Aberg D, Nilsson M. Protective and regenerative effects of the GH/IGF-I axis on the brain. Minerva Endocrinol. 2007;32(2):103–13.PubMed Isgaard J, Aberg D, Nilsson M. Protective and regenerative effects of the GH/IGF-I axis on the brain. Minerva Endocrinol. 2007;32(2):103–13.PubMed
91.
go back to reference Corvin AP, Molinos I, Little G, Donohoe G, Gill M, Morris DW, et al. Insulin-like growth factor 1 (IGF1) and its active peptide (1-3)IGF1 enhance the expression of synaptic markers in neuronal circuits through different cellular mechanisms. Neurosci Lett. 2012;520(1):51–6.PubMedCrossRef Corvin AP, Molinos I, Little G, Donohoe G, Gill M, Morris DW, et al. Insulin-like growth factor 1 (IGF1) and its active peptide (1-3)IGF1 enhance the expression of synaptic markers in neuronal circuits through different cellular mechanisms. Neurosci Lett. 2012;520(1):51–6.PubMedCrossRef
92.
go back to reference Llorens-Martín M, Torres-Alemán I, Trejo JL. Mechanisms mediating brain plasticity: IGF1 and adult hippocampal neurogenesis. Neuroscientist. 2009;15(2):134–48.PubMedCrossRef Llorens-Martín M, Torres-Alemán I, Trejo JL. Mechanisms mediating brain plasticity: IGF1 and adult hippocampal neurogenesis. Neuroscientist. 2009;15(2):134–48.PubMedCrossRef
93.
go back to reference Tropea D, Kreiman G, Lyckman A, Mukherjee S, Yu H, Horng S, et al. Gene expression changes and molecular pathways mediating activity-dependent plasticity in visual cortex. Nat Neurosci. 2006;9(5):660–8.PubMedCrossRef Tropea D, Kreiman G, Lyckman A, Mukherjee S, Yu H, Horng S, et al. Gene expression changes and molecular pathways mediating activity-dependent plasticity in visual cortex. Nat Neurosci. 2006;9(5):660–8.PubMedCrossRef
94.
go back to reference Torres-Aleman I. Insulin-like growth factors as mediators of functional plasticity in the adult brain. Horm Metab Res. 1999;31(2–3):114–9.PubMedCrossRef Torres-Aleman I. Insulin-like growth factors as mediators of functional plasticity in the adult brain. Horm Metab Res. 1999;31(2–3):114–9.PubMedCrossRef
96.
go back to reference Bonfanti L. PSA-NCAM in mammalian structural plasticity and neurogenesis. Prog Neurobiol. 2006;80(3):129–64.PubMedCrossRef Bonfanti L. PSA-NCAM in mammalian structural plasticity and neurogenesis. Prog Neurobiol. 2006;80(3):129–64.PubMedCrossRef
97.
go back to reference Monzo HJ, Park TIH, Dieriks BV, Jansson D, Faull RLM, Dragunow M, et al. Insulin and IGF1 modulate turnover of polysialylated neural cell adhesion molecule (PSA-NCAM) in a process involving specific extracellular matrix components. J Neurochem. 2013;126(6):758–70.PubMedCrossRef Monzo HJ, Park TIH, Dieriks BV, Jansson D, Faull RLM, Dragunow M, et al. Insulin and IGF1 modulate turnover of polysialylated neural cell adhesion molecule (PSA-NCAM) in a process involving specific extracellular matrix components. J Neurochem. 2013;126(6):758–70.PubMedCrossRef
98.
go back to reference McCusker RH, McCrea K, Zunich S, Dantzer R, Broussard SR, Johnson RW, et al. Insulin-like growth factor-I enhances the biological activity of brain-derived neurotrophic factor on cerebrocortical neurons. J Neuroimmunol. 2006;179(1–2):186–90.PubMedCrossRef McCusker RH, McCrea K, Zunich S, Dantzer R, Broussard SR, Johnson RW, et al. Insulin-like growth factor-I enhances the biological activity of brain-derived neurotrophic factor on cerebrocortical neurons. J Neuroimmunol. 2006;179(1–2):186–90.PubMedCrossRef
99.
go back to reference Mendez P, Azcoitia I, Garcia-Segura LM. Interdependence of oestrogen and insulin-like growth factor-I in the brain: potential for analysing neuroprotective mechanisms. J Endocrinol. 2005;185(1):11–7.PubMedCrossRef Mendez P, Azcoitia I, Garcia-Segura LM. Interdependence of oestrogen and insulin-like growth factor-I in the brain: potential for analysing neuroprotective mechanisms. J Endocrinol. 2005;185(1):11–7.PubMedCrossRef
100.
go back to reference Cardona-Gómez GP, Trejo JL, Fernandez AM, Garcia-Segura LM. Estrogen receptors and insulin-like growth factor-I receptors mediate estrogen-dependent synaptic plasticity. NeuroReport. 2000;11(8):1735–8.PubMedCrossRef Cardona-Gómez GP, Trejo JL, Fernandez AM, Garcia-Segura LM. Estrogen receptors and insulin-like growth factor-I receptors mediate estrogen-dependent synaptic plasticity. NeuroReport. 2000;11(8):1735–8.PubMedCrossRef
101.
go back to reference Trejo JL, Carro E, Lopez-Lopez C, Torres-Aleman I. Role of serum insulin-like growth factor I in mammalian brain aging. Growth Horm IGF Res. 2004;14(Suppl A):S39–43.PubMedCrossRef Trejo JL, Carro E, Lopez-Lopez C, Torres-Aleman I. Role of serum insulin-like growth factor I in mammalian brain aging. Growth Horm IGF Res. 2004;14(Suppl A):S39–43.PubMedCrossRef
102.
go back to reference Svensson J, Diez M, Engel J, Wass C, Tivesten A, Jansson J-O, et al. Endocrine, liver-derived IGF-I is of importance for spatial learning and memory in old mice. J Endocrinol. 2006;189(3):617–27.PubMedCrossRef Svensson J, Diez M, Engel J, Wass C, Tivesten A, Jansson J-O, et al. Endocrine, liver-derived IGF-I is of importance for spatial learning and memory in old mice. J Endocrinol. 2006;189(3):617–27.PubMedCrossRef
103.
go back to reference Lupien SB, Bluhm EJ, Ishii DN. Systemic insulin-like growth factor-I administration prevents cognitive impairment in diabetic rats, and brain IGF regulates learning/memory in normal adult rats. J Neurosci Res. 2003;74(4):512–23.PubMedCrossRef Lupien SB, Bluhm EJ, Ishii DN. Systemic insulin-like growth factor-I administration prevents cognitive impairment in diabetic rats, and brain IGF regulates learning/memory in normal adult rats. J Neurosci Res. 2003;74(4):512–23.PubMedCrossRef
104.
go back to reference Hoshaw BA, Malberg JE, Lucki I. Central administration of IGF-I and BDNF leads to long-lasting antidepressant-like effects. Brain Res. 2005;1037(1–2):204–8.PubMedCrossRef Hoshaw BA, Malberg JE, Lucki I. Central administration of IGF-I and BDNF leads to long-lasting antidepressant-like effects. Brain Res. 2005;1037(1–2):204–8.PubMedCrossRef
105.
go back to reference Castro-Alamancos MA, Torres-Aleman I. Learning of the conditioned eye-blink response is impaired by an antisense insulin-like growth factor I oligonucleotide. Proc Natl Acad Sci USA. 1994;91(21):10203–7.PubMedCrossRefPubMedCentral Castro-Alamancos MA, Torres-Aleman I. Learning of the conditioned eye-blink response is impaired by an antisense insulin-like growth factor I oligonucleotide. Proc Natl Acad Sci USA. 1994;91(21):10203–7.PubMedCrossRefPubMedCentral
106.
go back to reference Bluthé R-M, Kelley KW, Dantzer R. Effects of insulin-like growth factor-I on cytokine-induced sickness behavior in mice. Brain Behav Immun. 2006;20(1):57–63.PubMedPubMedCentralCrossRef Bluthé R-M, Kelley KW, Dantzer R. Effects of insulin-like growth factor-I on cytokine-induced sickness behavior in mice. Brain Behav Immun. 2006;20(1):57–63.PubMedPubMedCentralCrossRef
107.
go back to reference Ang LC, Bhaumick B, Juurlink BH. Neurite promoting activity of insulin, insulin-like growth factor I and nerve growth factor on spinal motoneurons is astrocyte dependent. Brain Res Dev Brain Res. 1993;74(1):83–8.PubMedCrossRef Ang LC, Bhaumick B, Juurlink BH. Neurite promoting activity of insulin, insulin-like growth factor I and nerve growth factor on spinal motoneurons is astrocyte dependent. Brain Res Dev Brain Res. 1993;74(1):83–8.PubMedCrossRef
108.
go back to reference Pomerance M, Gavaret JM, Breton M, Pierre M. Effects of growth factors on phosphatidylinositol-3 kinase in astroglial cells. J Neurosci Res. 1995;40(6):737–46.PubMedCrossRef Pomerance M, Gavaret JM, Breton M, Pierre M. Effects of growth factors on phosphatidylinositol-3 kinase in astroglial cells. J Neurosci Res. 1995;40(6):737–46.PubMedCrossRef
109.
go back to reference Zawada WM, Kirschman DL, Cohen JJ, Heidenreich KA, Freed CR. Growth factors rescue embryonic dopamine neurons from programmed cell death. Exp Neurol. 1996;140(1):60–7.PubMedCrossRef Zawada WM, Kirschman DL, Cohen JJ, Heidenreich KA, Freed CR. Growth factors rescue embryonic dopamine neurons from programmed cell death. Exp Neurol. 1996;140(1):60–7.PubMedCrossRef
110.
go back to reference Masuda S, Chikuma M, Sasaki R. Insulin-like growth factors and insulin stimulate erythropoietin production in primary cultured astrocytes. Brain Res. 1997;746(1–2):63–70.PubMedCrossRef Masuda S, Chikuma M, Sasaki R. Insulin-like growth factors and insulin stimulate erythropoietin production in primary cultured astrocytes. Brain Res. 1997;746(1–2):63–70.PubMedCrossRef
111.
go back to reference Tranque P, Naftolin F, Robbins R. Differential regulation of astrocyte plasminogen activators by insulin-like growth factor-I and epidermal growth factor. Endocrinology. 1994;134(6):2606–13.PubMedCrossRef Tranque P, Naftolin F, Robbins R. Differential regulation of astrocyte plasminogen activators by insulin-like growth factor-I and epidermal growth factor. Endocrinology. 1994;134(6):2606–13.PubMedCrossRef
112.
go back to reference Bradshaw SL, Han VK. Hormonal regulation of astroglial insulin-like growth factor (IGF)-binding protein gene expression by IGFs and insulin. Endocrinology. 1993;133(4):1767–77.PubMedCrossRef Bradshaw SL, Han VK. Hormonal regulation of astroglial insulin-like growth factor (IGF)-binding protein gene expression by IGFs and insulin. Endocrinology. 1993;133(4):1767–77.PubMedCrossRef
113.
go back to reference Aberg ND, Blomstrand F, Aberg MAI, Björklund U, Carlsson B, Carlsson-Skwirut C, et al. Insulin-like growth factor-I increases astrocyte intercellular gap junctional communication and connexin43 expression in vitro. J Neurosci Res. 2003;74(1):12–22.PubMedCrossRef Aberg ND, Blomstrand F, Aberg MAI, Björklund U, Carlsson B, Carlsson-Skwirut C, et al. Insulin-like growth factor-I increases astrocyte intercellular gap junctional communication and connexin43 expression in vitro. J Neurosci Res. 2003;74(1):12–22.PubMedCrossRef
114.
go back to reference Suzuki K, Ikegaya Y, Matsuura S, Kanai Y, Endou H, Matsuki N. Transient upregulation of the glial glutamate transporter GLAST in response to fibroblast growth factor, insulin-like growth factor and epidermal growth factor in cultured astrocytes. J Cell Sci. 2001;114(Pt 20):3717–25.PubMed Suzuki K, Ikegaya Y, Matsuura S, Kanai Y, Endou H, Matsuki N. Transient upregulation of the glial glutamate transporter GLAST in response to fibroblast growth factor, insulin-like growth factor and epidermal growth factor in cultured astrocytes. J Cell Sci. 2001;114(Pt 20):3717–25.PubMed
115.
go back to reference Sonnewald U, Wang AY, Petersen SB, Westergaard N, Schousboe A, Erikson R, et al. 13C NMR study of IGF-I- and insulin-effects on mitochondrial function in cultured brain cells. NeuroReport. 1995;6(6):878–80.PubMedCrossRef Sonnewald U, Wang AY, Petersen SB, Westergaard N, Schousboe A, Erikson R, et al. 13C NMR study of IGF-I- and insulin-effects on mitochondrial function in cultured brain cells. NeuroReport. 1995;6(6):878–80.PubMedCrossRef
116.
go back to reference Dringen R, Hamprecht B. Glucose, insulin, and insulin-like growth factor I regulate the glycogen content of astroglia-rich primary cultures. J Neurochem. 1992;58(2):511–7.PubMedCrossRef Dringen R, Hamprecht B. Glucose, insulin, and insulin-like growth factor I regulate the glycogen content of astroglia-rich primary cultures. J Neurochem. 1992;58(2):511–7.PubMedCrossRef
117.
go back to reference Masters BA, Werner H, Roberts CT, LeRoith D, Raizada MK. Developmental regulation of insulin-like growth factor-I-stimulated glucose transporter in rat brain astrocytes. Endocrinology. 1991;128(5):2548–57.PubMedCrossRef Masters BA, Werner H, Roberts CT, LeRoith D, Raizada MK. Developmental regulation of insulin-like growth factor-I-stimulated glucose transporter in rat brain astrocytes. Endocrinology. 1991;128(5):2548–57.PubMedCrossRef
118.
go back to reference Spina Purrello V, Cormaci G, Denaro L, Reale S, Costa A, Lalicata C, et al. Effect of growth factors on nuclear and mitochondrial ADP-ribosylation processes during astroglial cell development and aging in culture. Mech Ageing Dev. 2002;123(5):511–20.PubMedCrossRef Spina Purrello V, Cormaci G, Denaro L, Reale S, Costa A, Lalicata C, et al. Effect of growth factors on nuclear and mitochondrial ADP-ribosylation processes during astroglial cell development and aging in culture. Mech Ageing Dev. 2002;123(5):511–20.PubMedCrossRef
119.
go back to reference Matsuda T, Murata Y, Kawamura N, Hayashi M, Tamada K, Takuma K, et al. Selective induction of alpha 1 isoform of (Na++K +)-ATPase by insulin/insulin-like growth factor-I in cultured rat astrocytes. Arch Biochem Biophys. 1993;307(1):175–82.PubMedCrossRef Matsuda T, Murata Y, Kawamura N, Hayashi M, Tamada K, Takuma K, et al. Selective induction of alpha 1 isoform of (Na++K +)-ATPase by insulin/insulin-like growth factor-I in cultured rat astrocytes. Arch Biochem Biophys. 1993;307(1):175–82.PubMedCrossRef
120.
go back to reference Ridet JL, Malhotra SK, Privat A, Gage FH. Reactive astrocytes: cellular and molecular cues to biological function. Trends Neurosci. 1997;20(12):570–7.PubMedCrossRef Ridet JL, Malhotra SK, Privat A, Gage FH. Reactive astrocytes: cellular and molecular cues to biological function. Trends Neurosci. 1997;20(12):570–7.PubMedCrossRef
121.
go back to reference Walter HJ, Berry M, Hill DJ, Logan A. Spatial and temporal changes in the insulin-like growth factor (IGF) axis indicate autocrine/paracrine actions of IGF-I within wounds of the rat brain. Endocrinology. 1997;138(7):3024–34.PubMedCrossRef Walter HJ, Berry M, Hill DJ, Logan A. Spatial and temporal changes in the insulin-like growth factor (IGF) axis indicate autocrine/paracrine actions of IGF-I within wounds of the rat brain. Endocrinology. 1997;138(7):3024–34.PubMedCrossRef
122.
go back to reference Antoniades HN, Galanopoulos T, Neville-Golden J, Maxwell M. Expression of insulin-like growth factors I and II and their receptor mRNAs in primary human astrocytomas and meningiomas; in vivo studies using in situ hybridization and immunocytochemistry. Int J Cancer. 1992;50(2):215–22.PubMedCrossRef Antoniades HN, Galanopoulos T, Neville-Golden J, Maxwell M. Expression of insulin-like growth factors I and II and their receptor mRNAs in primary human astrocytomas and meningiomas; in vivo studies using in situ hybridization and immunocytochemistry. Int J Cancer. 1992;50(2):215–22.PubMedCrossRef
123.
go back to reference Lara-Diaz VJ, Castilla-Cortazar I, Martín-Estal I, García-Magariño M, Aguirre GA, Puche JE, et al. IGF-1 modulates gene expression of proteins involved in inflammation, cytoskeleton, and liver architecture. J Physiol Biochem. 2017;73:245–58.PubMedPubMedCentralCrossRef Lara-Diaz VJ, Castilla-Cortazar I, Martín-Estal I, García-Magariño M, Aguirre GA, Puche JE, et al. IGF-1 modulates gene expression of proteins involved in inflammation, cytoskeleton, and liver architecture. J Physiol Biochem. 2017;73:245–58.PubMedPubMedCentralCrossRef
124.
go back to reference Morales-Garza LA, Puche JE, de la Garza RG, Muñoz-Morón Ú, Castilla-Cortázar I. Experimental approach to IGF-1 therapy in acute liver damage: acute injury by carbon tetrachloride in controls and mice with partial IGF-1 deficiency. J Hepatol. 2017;66(1):1–14.CrossRef Morales-Garza LA, Puche JE, de la Garza RG, Muñoz-Morón Ú, Castilla-Cortázar I. Experimental approach to IGF-1 therapy in acute liver damage: acute injury by carbon tetrachloride in controls and mice with partial IGF-1 deficiency. J Hepatol. 2017;66(1):1–14.CrossRef
125.
go back to reference LeRoith D, Werner H, Beitner-Johnson D, Roberts CT. Molecular and cellular aspects of the insulin-like growth factor I receptor. In: Endocrine reviews, vol. 16. 1995. p. 143–63.PubMedCrossRef LeRoith D, Werner H, Beitner-Johnson D, Roberts CT. Molecular and cellular aspects of the insulin-like growth factor I receptor. In: Endocrine reviews, vol. 16. 1995. p. 143–63.PubMedCrossRef
126.
go back to reference Fernandez S, Fernandez AM, Lopez-Lopez C, Torres-Aleman I. Emerging roles of insulin-like growth factor-I in the adult brain. Growth Horm IGF Res. 2007;17(2):89–95.PubMedCrossRef Fernandez S, Fernandez AM, Lopez-Lopez C, Torres-Aleman I. Emerging roles of insulin-like growth factor-I in the adult brain. Growth Horm IGF Res. 2007;17(2):89–95.PubMedCrossRef
127.
go back to reference García-Fernández M, Delgado G, Puche JE, González-Barón S, Castilla Cortázar I. Low doses of insulin-like growth factor I improve insulin resistance, lipid metabolism, and oxidative damage in aging rats. Endocrinology. 2008;149(5):2433–42.PubMedCrossRef García-Fernández M, Delgado G, Puche JE, González-Barón S, Castilla Cortázar I. Low doses of insulin-like growth factor I improve insulin resistance, lipid metabolism, and oxidative damage in aging rats. Endocrinology. 2008;149(5):2433–42.PubMedCrossRef
128.
go back to reference Castilla-Cortazar I, Guerra L, Puche JE, Munoz U, Barhoum R, Escudero E, et al. An experimental model of partial insulin-like growth factor-1 deficiency in mice. J Physiol Biochem. 2013;70:129–39.PubMedCrossRef Castilla-Cortazar I, Guerra L, Puche JE, Munoz U, Barhoum R, Escudero E, et al. An experimental model of partial insulin-like growth factor-1 deficiency in mice. J Physiol Biochem. 2013;70:129–39.PubMedCrossRef
129.
go back to reference Puche JE, Muñoz Ú, García-Magariño M, Sádaba MC, Castilla-Cortázar I. Partial IGF-1 deficiency induces brain oxidative damage and edema, which are ameliorated by replacement therapy. Biofactors. 2016;42(1):60–79.PubMed Puche JE, Muñoz Ú, García-Magariño M, Sádaba MC, Castilla-Cortázar I. Partial IGF-1 deficiency induces brain oxidative damage and edema, which are ameliorated by replacement therapy. Biofactors. 2016;42(1):60–79.PubMed
130.
go back to reference Castilla-Cortázar I, Iturrieta I, García-Magariño M, Puche JE, Martín-Estal I, Aguirre GA, et al. Neurotrophic factors and their receptors are altered by the mere partial IGF-1 deficiency. Neuroscience. 2019;404:445–58.PubMedCrossRef Castilla-Cortázar I, Iturrieta I, García-Magariño M, Puche JE, Martín-Estal I, Aguirre GA, et al. Neurotrophic factors and their receptors are altered by the mere partial IGF-1 deficiency. Neuroscience. 2019;404:445–58.PubMedCrossRef
131.
go back to reference Gasperi M, Castellano AE. Growth hormone/insulin-like growth factor I axis in neurodegenerative diseases. J Endocrinol Investig. 2010;33(8):587–91.CrossRef Gasperi M, Castellano AE. Growth hormone/insulin-like growth factor I axis in neurodegenerative diseases. J Endocrinol Investig. 2010;33(8):587–91.CrossRef
132.
go back to reference Gubbi S, Quipildor GF, Barzilai N, Huffman DM, Milman S. 40 years of IGF1: IGF1: the Jekyll and Hyde of the aging brain. J Mol Endocrinol. 2018;61(1):T171–85.PubMedPubMedCentralCrossRef Gubbi S, Quipildor GF, Barzilai N, Huffman DM, Milman S. 40 years of IGF1: IGF1: the Jekyll and Hyde of the aging brain. J Mol Endocrinol. 2018;61(1):T171–85.PubMedPubMedCentralCrossRef
134.
go back to reference Watanabe T, Miyazaki A, Katagiri T, Yamamoto H, Idei T, Iguchi T. Relationship between serum insulin-like growth factor-1 levels and Alzheimer’s disease and vascular dementia. J Am Geriatr Soc. 2005;53(10):1748–53.PubMedCrossRef Watanabe T, Miyazaki A, Katagiri T, Yamamoto H, Idei T, Iguchi T. Relationship between serum insulin-like growth factor-1 levels and Alzheimer’s disease and vascular dementia. J Am Geriatr Soc. 2005;53(10):1748–53.PubMedCrossRef
135.
go back to reference Laron Z, Pertzelan A, Mannheimer S. Genetic pituitary dwarfism with high serum concentation of growth hormone—a new inborn error of metabolism? Isr J Med Sci. 1966;2(2):152–5.PubMed Laron Z, Pertzelan A, Mannheimer S. Genetic pituitary dwarfism with high serum concentation of growth hormone—a new inborn error of metabolism? Isr J Med Sci. 1966;2(2):152–5.PubMed
136.
go back to reference Laron Z. Short stature due to genetic defects affecting growth hormone activity. N Engl J Med. 1996;334(7):463–5.PubMedCrossRef Laron Z. Short stature due to genetic defects affecting growth hormone activity. N Engl J Med. 1996;334(7):463–5.PubMedCrossRef
137.
go back to reference Klinger B, Laron Z. Three year IGF-I treatment of children with Laron syndrome. J Pediatr Endocrinol Metab. 1995;8(3):149–58.PubMedCrossRef Klinger B, Laron Z. Three year IGF-I treatment of children with Laron syndrome. J Pediatr Endocrinol Metab. 1995;8(3):149–58.PubMedCrossRef
139.
go back to reference Wu A, Grant DB, Hambley J, Levi AJ. Reduced serum somatomedin activity in patients with chronic liver disease. Clin Sci Mol Med. 1974;47(4):359–66.PubMed Wu A, Grant DB, Hambley J, Levi AJ. Reduced serum somatomedin activity in patients with chronic liver disease. Clin Sci Mol Med. 1974;47(4):359–66.PubMed
140.
go back to reference Picardi A, de Oliveira AC, Muguerza B, Tosar A, Quiroga J, Castilla-Cortázar I, et al. Low doses of insulin-like growth factor-I improve nitrogen retention and food efficiency in rats with early cirrhosis. J Hepatol. 1997;26(1):191–202.PubMedCrossRef Picardi A, de Oliveira AC, Muguerza B, Tosar A, Quiroga J, Castilla-Cortázar I, et al. Low doses of insulin-like growth factor-I improve nitrogen retention and food efficiency in rats with early cirrhosis. J Hepatol. 1997;26(1):191–202.PubMedCrossRef
141.
go back to reference Pascual M, Castilla-Cortazar I, Urdaneta E, Quiroga J, Garcia M, Picardi A, et al. Altered intestinal transport of amino acids in cirrhotic rats: the effect of insulin-like growth factor-I. Am J Physiol Gastrointest Liver Physiol. 2000;279(2):G319–24.PubMedCrossRef Pascual M, Castilla-Cortazar I, Urdaneta E, Quiroga J, Garcia M, Picardi A, et al. Altered intestinal transport of amino acids in cirrhotic rats: the effect of insulin-like growth factor-I. Am J Physiol Gastrointest Liver Physiol. 2000;279(2):G319–24.PubMedCrossRef
142.
go back to reference Castilla-Cortázar I, Pascual M, Urdaneta E, Pardo J, Puche JE, Vivas B, et al. Jejunal microvilli atrophy and reduced nutrient transport in rats with advanced liver cirrhosis: improvement by Insulin-like Growth Factor I. BMC Gastroenterol. 2004;4:12.PubMedPubMedCentralCrossRef Castilla-Cortázar I, Pascual M, Urdaneta E, Pardo J, Puche JE, Vivas B, et al. Jejunal microvilli atrophy and reduced nutrient transport in rats with advanced liver cirrhosis: improvement by Insulin-like Growth Factor I. BMC Gastroenterol. 2004;4:12.PubMedPubMedCentralCrossRef
143.
go back to reference Castilla-Cortázar I, Picardi A, Tosar A, Ainzúa J, Urdaneta E, García M, et al. Effect of insulin-like growth factor I on in vivo intestinal absorption of d-galactose in cirrhotic rats. Am J Physiol. 1999;276(1 Pt 1):G37–42.PubMed Castilla-Cortázar I, Picardi A, Tosar A, Ainzúa J, Urdaneta E, García M, et al. Effect of insulin-like growth factor I on in vivo intestinal absorption of d-galactose in cirrhotic rats. Am J Physiol. 1999;276(1 Pt 1):G37–42.PubMed
144.
go back to reference Castilla-Cortazar I, Prieto J, Urdaneta E, Pascual M, Nuñez M, Zudaire E, et al. Impaired intestinal sugar transport in cirrhotic rats: correction by low doses of insulin-like growth factor I. Gastroenterology. 1997;113(4):1180–7.PubMedCrossRef Castilla-Cortazar I, Prieto J, Urdaneta E, Pascual M, Nuñez M, Zudaire E, et al. Impaired intestinal sugar transport in cirrhotic rats: correction by low doses of insulin-like growth factor I. Gastroenterology. 1997;113(4):1180–7.PubMedCrossRef
145.
go back to reference Cemborain A, Castilla-Cortázar I, García M, Quiroga J, Muguerza B, Picardi A, et al. Osteopenia in rats with liver cirrhosis: beneficial effects of IGF-I treatment. J Hepatol. 1998;28(1):122–31.PubMedCrossRef Cemborain A, Castilla-Cortázar I, García M, Quiroga J, Muguerza B, Picardi A, et al. Osteopenia in rats with liver cirrhosis: beneficial effects of IGF-I treatment. J Hepatol. 1998;28(1):122–31.PubMedCrossRef
146.
go back to reference Cemborain A, Castilla-Cortázar I, García M, Muguerza B, Delgado G, Díaz-Sánchez M, et al. Effects of IGF-I treatment on osteopenia in rats with advanced liver cirrhosis. J Physiol Biochem. 2000;56(2):91–9.PubMedCrossRef Cemborain A, Castilla-Cortázar I, García M, Muguerza B, Delgado G, Díaz-Sánchez M, et al. Effects of IGF-I treatment on osteopenia in rats with advanced liver cirrhosis. J Physiol Biochem. 2000;56(2):91–9.PubMedCrossRef
147.
go back to reference Castilla-Cortazar I, Quiroga J, Prieto J. Insulin-like growth factor-I, liver function, and hypogonadism in rats with experimentally induced cirrhosis. Hepatology. 2000;31(6):1379.PubMedCrossRef Castilla-Cortazar I, Quiroga J, Prieto J. Insulin-like growth factor-I, liver function, and hypogonadism in rats with experimentally induced cirrhosis. Hepatology. 2000;31(6):1379.PubMedCrossRef
148.
go back to reference Castilla-Cortazar I, Garcia M, Quiroga J, Diez N, Diez-Caballero F, Calvo A, et al. Insulin-like growth factor-I reverts testicular atrophy in rats with advanced cirrhosis. Hepatology. 2000;31(3):592–600.PubMedCrossRef Castilla-Cortazar I, Garcia M, Quiroga J, Diez N, Diez-Caballero F, Calvo A, et al. Insulin-like growth factor-I reverts testicular atrophy in rats with advanced cirrhosis. Hepatology. 2000;31(3):592–600.PubMedCrossRef
149.
go back to reference Castilla-Cortázar I, Aliaga-Montilla MA, Salvador J, García M, Delgado G, González-Barón S, Quiroga J, Prieto J. Insulin-like growth factor-I restores the reduced somatostatinergic tone controlling growth hormone secretion in cirrhotic rats. Liver. 2001;21:405–9.PubMedCrossRef Castilla-Cortázar I, Aliaga-Montilla MA, Salvador J, García M, Delgado G, González-Barón S, Quiroga J, Prieto J. Insulin-like growth factor-I restores the reduced somatostatinergic tone controlling growth hormone secretion in cirrhotic rats. Liver. 2001;21:405–9.PubMedCrossRef
150.
go back to reference Pérez R, García-Fernández M, Díaz-Sánchez M, Puche JE, Delgado G, Conchillo M, et al. Mitochondrial protection by low doses of insulin-like growth factor-I in experimental cirrhosis. World J Gastroenterol. 2008;14:2731–9.PubMedPubMedCentralCrossRef Pérez R, García-Fernández M, Díaz-Sánchez M, Puche JE, Delgado G, Conchillo M, et al. Mitochondrial protection by low doses of insulin-like growth factor-I in experimental cirrhosis. World J Gastroenterol. 2008;14:2731–9.PubMedPubMedCentralCrossRef
151.
go back to reference García-Fernández M, Castilla-Cortázar I, Díaz-Sanchez M, Navarro I, Puche JE, Castilla A, et al. Antioxidant effects of insulin-like growth factor-I (IGF-I) in rats with advanced liver cirrhosis. BMC Gastroenterol. 2005;5:7.PubMedPubMedCentralCrossRef García-Fernández M, Castilla-Cortázar I, Díaz-Sanchez M, Navarro I, Puche JE, Castilla A, et al. Antioxidant effects of insulin-like growth factor-I (IGF-I) in rats with advanced liver cirrhosis. BMC Gastroenterol. 2005;5:7.PubMedPubMedCentralCrossRef
152.
go back to reference Muguerza B, Castilla-Cortázar I, García M, Quiroga J, Santidrián S, Prieto J. Antifibrogenic effect in vivo of low doses of insulin-like growth factor-I in cirrhotic rats. Biochim Biophys Acta. 2001;1536(2–3):185–95.PubMedCrossRef Muguerza B, Castilla-Cortázar I, García M, Quiroga J, Santidrián S, Prieto J. Antifibrogenic effect in vivo of low doses of insulin-like growth factor-I in cirrhotic rats. Biochim Biophys Acta. 2001;1536(2–3):185–95.PubMedCrossRef
153.
go back to reference Castilla-Cortazar I, Garcia M, Muguerza B, Quiroga J, Perez R, Santidrian S, et al. Hepatoprotective effects of insulin-like growth factor I in rats with carbon tetrachloride-induced cirrhosis. Gastroenterology. 1997;113(5):1682–91.PubMedCrossRef Castilla-Cortazar I, Garcia M, Muguerza B, Quiroga J, Perez R, Santidrian S, et al. Hepatoprotective effects of insulin-like growth factor I in rats with carbon tetrachloride-induced cirrhosis. Gastroenterology. 1997;113(5):1682–91.PubMedCrossRef
154.
go back to reference Conchillo M, de Knegt RJ, Payeras M, Quiroga J, Sangro B, Herrero J-I, et al. Insulin-like growth factor I (IGF-I) replacement therapy increases albumin concentration in liver cirrhosis: results of a pilot randomized controlled clinical trial. J Hepatol. 2005;43(4):630–6.PubMedCrossRef Conchillo M, de Knegt RJ, Payeras M, Quiroga J, Sangro B, Herrero J-I, et al. Insulin-like growth factor I (IGF-I) replacement therapy increases albumin concentration in liver cirrhosis: results of a pilot randomized controlled clinical trial. J Hepatol. 2005;43(4):630–6.PubMedCrossRef
155.
go back to reference Ceda GP, Dall’Aglio E E, Maggio M, Lauretani F, Bandinelli S, Falzoi C, et al. Clinical implications of the reduced activity of the GH-IGF-I axis in older men. J Endocrinol Investig. 2005;28(11 Suppl Proceedings):96–100. Ceda GP, Dall’Aglio E E, Maggio M, Lauretani F, Bandinelli S, Falzoi C, et al. Clinical implications of the reduced activity of the GH-IGF-I axis in older men. J Endocrinol Investig. 2005;28(11 Suppl Proceedings):96–100.
156.
go back to reference Sun LY, Al-Regaiey K, Masternak MM, Wang J, Bartke A. Local expression of GH and IGF-1 in the hippocampus of GH-deficient long-lived mice. Neurobiol Aging. 2005;26(6):929–37.PubMedCrossRef Sun LY, Al-Regaiey K, Masternak MM, Wang J, Bartke A. Local expression of GH and IGF-1 in the hippocampus of GH-deficient long-lived mice. Neurobiol Aging. 2005;26(6):929–37.PubMedCrossRef
157.
go back to reference Sonntag WE, Lynch CD, Cooney PT, Hutchins PM. Decreases in cerebral microvasculature with age are associated with the decline in growth hormone and insulin-like growth factor 1. Endocrinology. 1997;138(8):3515–20.PubMedCrossRef Sonntag WE, Lynch CD, Cooney PT, Hutchins PM. Decreases in cerebral microvasculature with age are associated with the decline in growth hormone and insulin-like growth factor 1. Endocrinology. 1997;138(8):3515–20.PubMedCrossRef
158.
go back to reference Sonntag WE, Carter CS, Ikeno Y, Ekenstedt K, Carlson CS, Loeser RF, et al. Adult-onset growth hormone and insulin-like growth factor I deficiency reduces neoplastic disease, modifies age-related pathology, and increases life span. Endocrinology. 2005;146(7):2920–32.PubMedCrossRef Sonntag WE, Carter CS, Ikeno Y, Ekenstedt K, Carlson CS, Loeser RF, et al. Adult-onset growth hormone and insulin-like growth factor I deficiency reduces neoplastic disease, modifies age-related pathology, and increases life span. Endocrinology. 2005;146(7):2920–32.PubMedCrossRef
159.
go back to reference Shimokawa I, Higami Y, Tsuchiya T, Otani H, Komatsu T, Chiba T, et al. Life span extension by reduction of the growth hormone-insulin-like growth factor-1 axis: relation to caloric restriction. FASEB J. 2003;17(9):1108–9.PubMedCrossRef Shimokawa I, Higami Y, Tsuchiya T, Otani H, Komatsu T, Chiba T, et al. Life span extension by reduction of the growth hormone-insulin-like growth factor-1 axis: relation to caloric restriction. FASEB J. 2003;17(9):1108–9.PubMedCrossRef
160.
161.
go back to reference Sonntag WE, Bennett C, Ingram R, Donahue A, Ingraham J, Chen H, et al. Growth hormone and IGF-I modulate local cerebral glucose utilization and ATP levels in a model of adult-onset growth hormone deficiency. Am J Physiol Endocrinol Metab. 2006;291(3):E604–10.PubMedCrossRef Sonntag WE, Bennett C, Ingram R, Donahue A, Ingraham J, Chen H, et al. Growth hormone and IGF-I modulate local cerebral glucose utilization and ATP levels in a model of adult-onset growth hormone deficiency. Am J Physiol Endocrinol Metab. 2006;291(3):E604–10.PubMedCrossRef
162.
go back to reference Maggio M, Lauretani F, Ceda GP, Bandinelli S, Basaria S, Paolisso G, et al. Association of hormonal dysregulation with metabolic syndrome in older women: data from the InCHIANTI study. Am J Physiol Endocrinol Metab. 2007;292(1):E353–8.PubMedCrossRef Maggio M, Lauretani F, Ceda GP, Bandinelli S, Basaria S, Paolisso G, et al. Association of hormonal dysregulation with metabolic syndrome in older women: data from the InCHIANTI study. Am J Physiol Endocrinol Metab. 2007;292(1):E353–8.PubMedCrossRef
163.
go back to reference Groban L, Pailes NA, Bennett CDL, Carter CS, Chappell MC, Kitzman DW, et al. Growth hormone replacement attenuates diastolic dysfunction and cardiac angiotensin II expression in senescent rats. J Gerontol A Biol Sci Med Sci. 2006;61(1):28–35.PubMedCrossRef Groban L, Pailes NA, Bennett CDL, Carter CS, Chappell MC, Kitzman DW, et al. Growth hormone replacement attenuates diastolic dysfunction and cardiac angiotensin II expression in senescent rats. J Gerontol A Biol Sci Med Sci. 2006;61(1):28–35.PubMedCrossRef
164.
go back to reference Li Q, Wu S, Li S-Y, Lopez FL, Du M, Kajstura J, et al. Cardiac-specific overexpression of insulin-like growth factor 1 attenuates aging-associated cardiac diastolic contractile dysfunction and protein damage. Am J Physiol Heart Circ Physiol. 2007;292(3):H1398–403.PubMedCrossRef Li Q, Wu S, Li S-Y, Lopez FL, Du M, Kajstura J, et al. Cardiac-specific overexpression of insulin-like growth factor 1 attenuates aging-associated cardiac diastolic contractile dysfunction and protein damage. Am J Physiol Heart Circ Physiol. 2007;292(3):H1398–403.PubMedCrossRef
166.
go back to reference Tollet-Egnell P, Flores-Morales A, Odeberg J, Lundeberg J, Norstedt G. Differential cloning of growth hormone-regulated hepatic transcripts in the aged rat. Endocrinology. 2000;141(3):910–21.PubMedCrossRef Tollet-Egnell P, Flores-Morales A, Odeberg J, Lundeberg J, Norstedt G. Differential cloning of growth hormone-regulated hepatic transcripts in the aged rat. Endocrinology. 2000;141(3):910–21.PubMedCrossRef
167.
go back to reference Puche JE, García-Fernández M, Muntané J, Rioja J, González-Barón S, Cortazar IC. Low doses of insulin-like growth factor-I induce mitochondrial protection in aging rats. Endocrinology. 2008;149:2620–7.PubMedCrossRef Puche JE, García-Fernández M, Muntané J, Rioja J, González-Barón S, Cortazar IC. Low doses of insulin-like growth factor-I induce mitochondrial protection in aging rats. Endocrinology. 2008;149:2620–7.PubMedCrossRef
168.
go back to reference García-Fernández M, Delgado G, Puche JE, González-Barón S, Cortázar IC. Low doses of insulin-like growth factor I improve insulin resistance, lipid metabolism, and oxidative damage in aging rats. Endocrinology. 2008;149:2433–42.PubMedCrossRef García-Fernández M, Delgado G, Puche JE, González-Barón S, Cortázar IC. Low doses of insulin-like growth factor I improve insulin resistance, lipid metabolism, and oxidative damage in aging rats. Endocrinology. 2008;149:2433–42.PubMedCrossRef
169.
go back to reference Pellecchia MT, Pivonello R, Salvatore E, Faggiano A, Barone P, De Michele G, et al. Growth hormone response to arginine test distinguishes multiple system atrophy from Parkinson’s disease and idiopathic late-onset cerebellar ataxia. Clin Endocrinol. 2005;62(4):428–33.CrossRef Pellecchia MT, Pivonello R, Salvatore E, Faggiano A, Barone P, De Michele G, et al. Growth hormone response to arginine test distinguishes multiple system atrophy from Parkinson’s disease and idiopathic late-onset cerebellar ataxia. Clin Endocrinol. 2005;62(4):428–33.CrossRef
170.
go back to reference Volpi R, Caffarra P, Scaglioni A, Boni S, Saginario A, Chiodera P, et al. Defective 5-HT 1-receptor-mediated neurotransmission in the control of growth hormone secretion in Parkinson’s disease. Neuropsychobiology. 1997;35(2):79–83.PubMedCrossRef Volpi R, Caffarra P, Scaglioni A, Boni S, Saginario A, Chiodera P, et al. Defective 5-HT 1-receptor-mediated neurotransmission in the control of growth hormone secretion in Parkinson’s disease. Neuropsychobiology. 1997;35(2):79–83.PubMedCrossRef
171.
go back to reference Bernhard FP, Heinzel S, Binder G, Weber K, Apel A, Roeben B, et al. Insulin-like growth factor 1 (IGF-1) in Parkinson’s disease: potential as trait-, progression- and prediction marker and confounding factors. PLoS ONE. 2016;11(3):e0150552.PubMedPubMedCentralCrossRef Bernhard FP, Heinzel S, Binder G, Weber K, Apel A, Roeben B, et al. Insulin-like growth factor 1 (IGF-1) in Parkinson’s disease: potential as trait-, progression- and prediction marker and confounding factors. PLoS ONE. 2016;11(3):e0150552.PubMedPubMedCentralCrossRef
172.
go back to reference Godau J, Knauel K, Weber K, Brockmann K, Maetzler W, Binder G, et al. Serum insulinlike growth factor 1 as possible marker for risk and early diagnosis of Parkinson disease. Arch Neurol. 2011;68(7):925–31.PubMedCrossRef Godau J, Knauel K, Weber K, Brockmann K, Maetzler W, Binder G, et al. Serum insulinlike growth factor 1 as possible marker for risk and early diagnosis of Parkinson disease. Arch Neurol. 2011;68(7):925–31.PubMedCrossRef
173.
go back to reference Godau J, Herfurth M, Kattner B, Gasser T, Berg D. Increased serum insulin-like growth factor 1 in early idiopathic Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2010;81(5):536–8.PubMedCrossRef Godau J, Herfurth M, Kattner B, Gasser T, Berg D. Increased serum insulin-like growth factor 1 in early idiopathic Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2010;81(5):536–8.PubMedCrossRef
174.
go back to reference Mashayekhi F, Mirzajani E, Naji M, Azari M. Expression of insulin-like growth factor-1 and insulin-like growth factor binding proteins in the serum and cerebrospinal fluid of patients with Parkinson’s disease. J Clin Neurosci. 2010;17(5):623–7.PubMedCrossRef Mashayekhi F, Mirzajani E, Naji M, Azari M. Expression of insulin-like growth factor-1 and insulin-like growth factor binding proteins in the serum and cerebrospinal fluid of patients with Parkinson’s disease. J Clin Neurosci. 2010;17(5):623–7.PubMedCrossRef
175.
go back to reference Numao A, Suzuki K, Miyamoto M, Miyamoto T, Hirata K. Clinical correlates of serum insulin-like growth factor-1 in patients with Parkinson’s disease, multiple system atrophy and progressive supranuclear palsy. Park Relat Disord. 2014;20(2):212–6.CrossRef Numao A, Suzuki K, Miyamoto M, Miyamoto T, Hirata K. Clinical correlates of serum insulin-like growth factor-1 in patients with Parkinson’s disease, multiple system atrophy and progressive supranuclear palsy. Park Relat Disord. 2014;20(2):212–6.CrossRef
176.
go back to reference Fiszer U, Michałowska M, Baranowska B, Wolińska-Witort E, Jeske W, Jethon M, et al. Leptin and ghrelin concentrations and weight loss in Parkinson’s disease. Acta Neurol Scand. 2010;121(4):230–6.PubMedCrossRef Fiszer U, Michałowska M, Baranowska B, Wolińska-Witort E, Jeske W, Jethon M, et al. Leptin and ghrelin concentrations and weight loss in Parkinson’s disease. Acta Neurol Scand. 2010;121(4):230–6.PubMedCrossRef
177.
go back to reference Tuncel D, Inanc Tolun F, Toru I. Serum insulin-like growth factor-1 and nitric oxide levels in Parkinson’s disease. Mediators Inflamm. 2009;2009:132464.PubMedPubMedCentralCrossRef Tuncel D, Inanc Tolun F, Toru I. Serum insulin-like growth factor-1 and nitric oxide levels in Parkinson’s disease. Mediators Inflamm. 2009;2009:132464.PubMedPubMedCentralCrossRef
178.
go back to reference Pellecchia MT, Santangelo G, Picillo M, Pivonello R, Longo K, Pivonello C, et al. Insulin-like growth factor-1 predicts cognitive functions at 2-year follow-up in early, drug-naïve Parkinson’s disease. Eur J Neurol. 2014;21(5):802–7.PubMedCrossRef Pellecchia MT, Santangelo G, Picillo M, Pivonello R, Longo K, Pivonello C, et al. Insulin-like growth factor-1 predicts cognitive functions at 2-year follow-up in early, drug-naïve Parkinson’s disease. Eur J Neurol. 2014;21(5):802–7.PubMedCrossRef
179.
go back to reference Picillo M, Erro R, Santangelo G, Pivonello R, Longo K, Pivonello C, et al. Insulin-like growth factor-1 and progression of motor symptoms in early, drug-naïve Parkinson’s disease. J Neurol. 2013;260(7):1724–30.PubMedCrossRef Picillo M, Erro R, Santangelo G, Pivonello R, Longo K, Pivonello C, et al. Insulin-like growth factor-1 and progression of motor symptoms in early, drug-naïve Parkinson’s disease. J Neurol. 2013;260(7):1724–30.PubMedCrossRef
180.
go back to reference Picillo M, Pivonello R, Santangelo G, Pivonello C, Savastano R, Auriemma R, et al. Serum IGF-1 is associated with cognitive functions in early, drug-naive Parkinson’s disease. PLoS ONE. 2017;12(10):e0186508.PubMedPubMedCentralCrossRef Picillo M, Pivonello R, Santangelo G, Pivonello C, Savastano R, Auriemma R, et al. Serum IGF-1 is associated with cognitive functions in early, drug-naive Parkinson’s disease. PLoS ONE. 2017;12(10):e0186508.PubMedPubMedCentralCrossRef
181.
go back to reference Li D-H, He Y-C, Quinn TJ, Liu J. Serum insulin-like growth factor-1 in patients with de novo, drug naïve Parkinson’s disease: a meta-analysis. PLoS ONE. 2015;10(12):e0144755.PubMedPubMedCentralCrossRef Li D-H, He Y-C, Quinn TJ, Liu J. Serum insulin-like growth factor-1 in patients with de novo, drug naïve Parkinson’s disease: a meta-analysis. PLoS ONE. 2015;10(12):e0144755.PubMedPubMedCentralCrossRef
182.
go back to reference Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, et al. alpha-Synuclein locus triplication causes Parkinson’s disease. Science. 2003;302(5646):841.CrossRefPubMed Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, et al. alpha-Synuclein locus triplication causes Parkinson’s disease. Science. 2003;302(5646):841.CrossRefPubMed
183.
go back to reference Kumari U, Tan EK. LRRK2 in Parkinson’s disease: genetic and clinical studies from patients. FEBS J. 2009;276(22):6455–63.PubMedCrossRef Kumari U, Tan EK. LRRK2 in Parkinson’s disease: genetic and clinical studies from patients. FEBS J. 2009;276(22):6455–63.PubMedCrossRef
184.
go back to reference Licker V, Kövari E, Hochstrasser DF, Burkhard PR. Proteomics in human Parkinson’s disease research. J Proteomics. 2009;73(1):10–29.PubMedCrossRef Licker V, Kövari E, Hochstrasser DF, Burkhard PR. Proteomics in human Parkinson’s disease research. J Proteomics. 2009;73(1):10–29.PubMedCrossRef
185.
go back to reference Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388(6645):839–40.CrossRefPubMed Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388(6645):839–40.CrossRefPubMed
186.
go back to reference Chung J-Y, Lee S-J, Lee S-H, Jung YS, Ha N-C, Seol W, et al. Direct interaction of α-synuclein and AKT regulates IGF-1 signaling: implication of Parkinson disease. Neurosignals. 2011;19(2):86–96.PubMedCrossRef Chung J-Y, Lee S-J, Lee S-H, Jung YS, Ha N-C, Seol W, et al. Direct interaction of α-synuclein and AKT regulates IGF-1 signaling: implication of Parkinson disease. Neurosignals. 2011;19(2):86–96.PubMedCrossRef
187.
go back to reference Kao S-Y. Rescue of α-synuclein cytotoxicity by insulin-like growth factors. Biochem Biophys Res Commun. 2009;385(3):434–8.PubMedCrossRef Kao S-Y. Rescue of α-synuclein cytotoxicity by insulin-like growth factors. Biochem Biophys Res Commun. 2009;385(3):434–8.PubMedCrossRef
188.
go back to reference Kim W, Lee Y, McKenna ND, Yi M, Simunovic F, Wang Y, et al. miR-126 contributes to Parkinson’s disease by dysregulating the insulin-like growth factor/phosphoinositide 3-kinase signaling. Neurobiol Aging. 2014;35(7):1712–21.PubMedPubMedCentralCrossRef Kim W, Lee Y, McKenna ND, Yi M, Simunovic F, Wang Y, et al. miR-126 contributes to Parkinson’s disease by dysregulating the insulin-like growth factor/phosphoinositide 3-kinase signaling. Neurobiol Aging. 2014;35(7):1712–21.PubMedPubMedCentralCrossRef
189.
go back to reference Dulloo AG, Montani J-P. Obesity in Parkinson’s disease patients on electrotherapy: collateral damage, adiposity rebound or secular trends? Br J Nutr. 2005;93(4):417–9.PubMedCrossRef Dulloo AG, Montani J-P. Obesity in Parkinson’s disease patients on electrotherapy: collateral damage, adiposity rebound or secular trends? Br J Nutr. 2005;93(4):417–9.PubMedCrossRef
190.
go back to reference Chen H, Zhang SM, Schwarzschild MA, Hernán MA, Willett WC, Ascherio A. Obesity and the risk of Parkinson’s disease. Am J Epidemiol. 2004;159(6):547–55.PubMedCrossRef Chen H, Zhang SM, Schwarzschild MA, Hernán MA, Willett WC, Ascherio A. Obesity and the risk of Parkinson’s disease. Am J Epidemiol. 2004;159(6):547–55.PubMedCrossRef
191.
go back to reference Santiago JA, Potashkin JA. Shared dysregulated pathways lead to Parkinson’s disease and diabetes. Trends Mol Med. 2013;19(3):176–86.PubMedCrossRef Santiago JA, Potashkin JA. Shared dysregulated pathways lead to Parkinson’s disease and diabetes. Trends Mol Med. 2013;19(3):176–86.PubMedCrossRef
192.
go back to reference Zaganas I, Kapetanaki S, Mastorodemos V, Kanavouras K, Colosio C, Wilks MF, et al. Linking pesticide exposure and dementia: what is the evidence? Toxicology. 2013;307:3–11.PubMedCrossRef Zaganas I, Kapetanaki S, Mastorodemos V, Kanavouras K, Colosio C, Wilks MF, et al. Linking pesticide exposure and dementia: what is the evidence? Toxicology. 2013;307:3–11.PubMedCrossRef
193.
go back to reference Freire C, Koifman S. Pesticide exposure and Parkinson’s disease: epidemiological evidence of association. Neurotoxicology. 2012;33(5):947–71.PubMedCrossRef Freire C, Koifman S. Pesticide exposure and Parkinson’s disease: epidemiological evidence of association. Neurotoxicology. 2012;33(5):947–71.PubMedCrossRef
194.
go back to reference Di Monte DA. The environment and Parkinson’s disease: is the nigrostriatal system preferentially targeted by neurotoxins? Lancet Neurol. 2003;2(9):531–8.PubMedCrossRef Di Monte DA. The environment and Parkinson’s disease: is the nigrostriatal system preferentially targeted by neurotoxins? Lancet Neurol. 2003;2(9):531–8.PubMedCrossRef
195.
go back to reference Aguirre GA, González-Guerra JL, Espinosa L, Castilla-Cortazar I. Insulin-like growth factor 1 in the cardiovascular system. In: Rev Physiol Biochem Pharmacol. 2018. Aguirre GA, González-Guerra JL, Espinosa L, Castilla-Cortazar I. Insulin-like growth factor 1 in the cardiovascular system. In: Rev Physiol Biochem Pharmacol. 2018.
196.
go back to reference Zuo L, Motherwell MS. The impact of reactive oxygen species and genetic mitochondrial mutations in Parkinson’s disease. Gene. 2013;532(1):18–23.PubMedCrossRef Zuo L, Motherwell MS. The impact of reactive oxygen species and genetic mitochondrial mutations in Parkinson’s disease. Gene. 2013;532(1):18–23.PubMedCrossRef
Metadata
Title
Is insulin-like growth factor-1 involved in Parkinson’s disease development?
Authors
Inma Castilla-Cortázar
Gabriel A. Aguirre
Giovana Femat-Roldán
Irene Martín-Estal
Luis Espinosa
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Journal of Translational Medicine / Issue 1/2020
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-020-02223-0

Other articles of this Issue 1/2020

Journal of Translational Medicine 1/2020 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