Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Expert Review
  • Published:

Mechanisms affecting brain remodeling in depression: do all roads lead to impaired fibrinolysis?

Abstract

Fibrinolysis occurs when plasminogen activators, such as tissue plasminogen activator (tPA), convert plasminogen to plasmin, which dissolves the fibrin clot. The proteolytic activity of tPA and plasmin is not restricted to fibrin degradation. In the extravascular space, these two proteases modify a variety of substrates other than fibrin, playing a crucial role in physiological and pathological tissue remodeling. In the brain, for example, tPA and plasmin mediate the conversion of brain-derived neurotrophic factor precursor (proBDNF) to mature brain-derived neurotrophic factor precursor (BDNF). Thus, the fibrinolytic system influences processes reported to be dysfunctional in depression, including neurogenesis, synaptic plasticity, and reward processing. The hypothesis that decreased fibrinolytic activity is an important element in the pathogenesis of depression is supported by the association between depression and increased levels of plasminogen activator inhibitor (PAI)−1, the main inhibitor of tPA. Also, various biochemical markers of depression induce PAI-1 synthesis, including hypercortisolism, hyperinsulinemia, hyperleptinemia, increased levels of cytokines, and hyperhomocysteinemia. Moreover, hypofibrinolysis provides a link between depression and emotional eating, binge eating, vegetarianism, and veganism. This paper discusses the role of reduced fibrinolytic activity in the bidirectional interplay between depression and its somatic manifestations and complications. It also reviews evidence that abnormal fibrinolysis links heterogeneous conditions associated with treatment-resistant depression. Understanding the role of hypofibrinolysis in depression may open new avenues for its treatment.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Multiple roles of tPA and plasmin in brain remodeling.
Fig. 2: The food-mood connection.

Similar content being viewed by others

References

  1. Han MH, Russo SJ, Nestler EJ. Molecular, cellular, and circuit basis of depression susceptibility and resilience. In: Quevedo J, Carvalho AF, Zarate CA editors. Neurobiology of depression. Academic Press; 2019. p. 123–36.

  2. Kessler RC, Sampson NA, Berglund P, Gruber MJ, Al-Hamzawi A, Andrade L, et al. Anxious and non-anxious major depressive disorder in the World Health Organization. World Ment Health Surv Epidemiol Psychiatr Sci. 2015;24:210–26.

    Article  CAS  Google Scholar 

  3. McEwen BS, Bowles NP, Gray JD, Hill MN, Hunter RG, Karatsoreos IN, et al. Mechanisms of stress in the brain. Nat Neurosci. 2015;18:1353–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Duman RS. Pathophysiology of depression: the concept of synaptic plasticity. Eur Psychiatry. 2002;17:306–10.

    Article  PubMed  Google Scholar 

  5. Campbell S, Marriott M, Nahmias C, MacQueen GM. Lower hippocampal volume in patients suffering from depression: a meta-analysis. Am J Psychiatry. 2004;161:598–607.

    Article  PubMed  Google Scholar 

  6. Bora E, Harrison BJ, Davey CG, Yücel M, Pantelis C. Meta analysis of volumetric abnormalities in cortico-striatal-pallidal-thalamic circuits in major depressive disorder. Psychol Med. 2012;42:671–81.

    Article  CAS  PubMed  Google Scholar 

  7. Sugimoto K, Kakeda S, Watanabe K, Katsuki A, Ueda I, Igata N, et al. Relationship between white matter integrity and serum inflammatory cytokine levels in drug-naïve patients with major depressive disorder: diffusion tensor imaging study using tract-based spatial statistics. Transl Psychiatry. 2018;8:1–8.

    Article  Google Scholar 

  8. Zhang FF, Peng W, Sweeney JA, Jia ZY, Gong QY. Brain structure alterations in depression: Psychoradiological evidence. CNS Neurosci Ther. 2018;24:994–1003.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Christian KM, Song H, Ming GL. Functions and dysfunctions of adult hippocampal neurogenesis. Annu Rev Neurosci. 2014;37:243–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. De Luca C, Colangelo AM, Virtuoso A, Alberghina L, Papa M. Neurons, glia, extracellular matrix and neurovascular unit: a systems biology approach to the complexity of synaptic plasticity in health and disease. Intern J Mol Sci. 2020;21:1539.

    Article  Google Scholar 

  11. Ito M, Nagai T, Kamei H, Nakamichi N, Nabeshima T, Takuma K, et al. Involvement of tissue plasminogen activator-plasmin system in depolarization-evoked dopamine release in the nucleus accumbens of mice. Mol Pharm. 2006;70:1720–5.

    Article  CAS  Google Scholar 

  12. Malemud CJ. Matrix metalloproteinases (MMPs) in health and disease: an overview. Front Biosci. 2006;11:1696–701.

    Article  CAS  PubMed  Google Scholar 

  13. Trotter JH, Lussier AL, Psilos KE, Mahoney HL, Sponaugle AE, Hoe HS, et al. Extracellular proteolysis of reelin by tissue plasminogen activator following synaptic potentiation. Neuroscience. 2014;274:299–307.

    Article  CAS  PubMed  Google Scholar 

  14. Matys T, Strickland S. Tissue plasminogen activator and NMDA receptor cleavage. Nat Med. 2003;9:371–2.

    Article  CAS  PubMed  Google Scholar 

  15. Lu B, Pang PT, Woo NH. The yin and yang of neurotrophin action. Nat Rev Neurosci. 2005;6:603–14.

    Article  CAS  PubMed  Google Scholar 

  16. Parmer RJ, Gong Y, Yoo SH, Miles LA. Neuroendocrine targeting of tissue plasminogen activator (t-PA). J Neurol Disord Stroke. 2020;7:1153.

    PubMed  PubMed Central  Google Scholar 

  17. Visentin C, Broggini L, Sala BM, Russo R, Barbiroli A, Santambrogio C, et al. Glycosylation tunes neuroserpin physiological and pathological properties. Int J Mol Sci. 2020;21:3235.

    Article  CAS  PubMed Central  Google Scholar 

  18. Girard RA, Chauhan PS, Tucker TA, Allen T, Kaur J, Jeffers A, et al. Increased expression of plasminogen activator inhibitor-1 (PAI-1) is associated with depression and depressive phenotype in C57Bl/6J mice. Exp Brain Res. 2019;237:3419–30.

    Article  CAS  PubMed  Google Scholar 

  19. Jiang H, Li X, Chen S, Lu N, Yue Y, Liang J, et al. Plasminogen activator inhibitor-1 in depression: results from animal and clinical studies. Sci Rep. 2016;6:30464.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Lahlou-Laforet K, Alhenc-Gelas M, Pornin M, Bydlowski S, Seigneur E, Benetos A, et al. Relation of depressive mood to plasminogen activator inhibitor, tissue plasminogen activator, and fibrinogen levels in patients with versus without coronary heart disease. Am J Cardiol. 2006;97:1287–91.

    Article  CAS  PubMed  Google Scholar 

  21. Chan MK, Cooper JD, Bot M, Birkenhager TK, Bergink V, Drexhage HA, et al. Blood-based immune-endocrine biomarkers of treatment response in depression. J Psychiatr Res. 2016;83:249–59.

    Article  PubMed  Google Scholar 

  22. Alessi MC, Juhan-Vague I. PAI-1 and the metabolic syndrome: links, causes, and consequences. Arterioscler Thromb Vasc Biol. 2006;26:2200–7.

    Article  CAS  PubMed  Google Scholar 

  23. Van Zonneveld AJ, Curriden SA, Loskutoff DJ. Type 1 plasminogen activator inhibitor gene: functional analysis and glucocorticoid regulation of its promoter. Proc Natl Acad Sci USA. 1988;85:5525–9.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Nordt TK, Bode C, Sobel BE. Stimulation in vivo of expression of intra-abdominal adipose tissue plasminogen activator inhibitor type I by proinsulin. Diabetologia. 2001;44:1121–4.

    Article  CAS  PubMed  Google Scholar 

  25. Singh P, Peterson TE, Barber KR, Kuniyoshi FS, Jensen A, Hoffmann M, et al. Leptin upregulates the expression of plasminogen activator inhibitor-1 in human vascular endothelial cells. Biochem Biophys Res Commun. 2010;392:47–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Eriksson P, Nilsson L, Karpe F, Hamsten A. Very-low-density lipoprotein response element in the promoter region of the human plasminogen activator inhibitor-1 gene implicated in the impaired fibrinolysis of hypertriglyceridemia. Arterioscl Thromb Vasc Biol. 1998;18:20–6.

    Article  CAS  PubMed  Google Scholar 

  27. Vaughan DT. PAI‐1 and atherothrombosis. J Thromb Haemost. 2005;3:1879–83.

    Article  CAS  PubMed  Google Scholar 

  28. Dong J, Fujii S, Imagawa S, Matsumoto S, Matsushita M, Todo S, et al. IL-1 and IL-6 induce hepatocyte plasminogen activator inhibitor-1 expression through independent signaling pathways converging on C/EBPδ. Am J Physiol Cell Physiol. 2017;292:209–15.

    Article  Google Scholar 

  29. Pandey M, Loskutoff DJ, Samad F. Molecular mechanisms of tumor necrosis factor-alpha-mediated plasminogen activator inhibitor-1 expression in adipocytes. FASEB J. 2005;19:1317–9.

    Article  CAS  PubMed  Google Scholar 

  30. Midorikawa S, Sanada H, Hashimoto S, Watanabe T. Enhancement by homocysteine of plasminogen activator inhibitor-1 gene expression and secretion from vascular endothelial and smooth muscle cells. Biochem Biophys Res Commun. 2000;272:182–5.

    Article  CAS  PubMed  Google Scholar 

  31. Søndergaard SR, Ostrowski K, Ullum H, Pedersen BK. Changes in plasma concentrations of interleukin-6 and interleukin-1 receptor antagonists in response to adrenaline infusion in humans. Eur J Appl Physiol. 2000;83:95–98.

    Article  PubMed  Google Scholar 

  32. Mastorakos G, Chrousos GP, Weber JS. Recombinant interleukin-6 activates the hypothalamic-pituitary-adrenal axis in humans. J Clin Endocrinol Metab. 1993;77:1690–4.

    CAS  PubMed  Google Scholar 

  33. Bornstein SR, Engeland WC, Ehrhart-Bornstein M, Herman JP. Dissociation of ACTH and glucocorticoids. Trends Endocrinol Metab. 2008;19:175–80.

    Article  CAS  PubMed  Google Scholar 

  34. Boonen E, Vervenne H, Meersseman P, Andrew R, Mortier L, Declercq PE, et al. Reduced cortisol metabolism during critical illness. N. Engl J Med. 2013;368:1477–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Gold PW, Chrousos GP. Organization of the stress system and its dysregulation in melancholic and atypical depression: high vs low CRH/NE states. Mol Psychiatry. 2002;7:254–75.

    Article  CAS  PubMed  Google Scholar 

  36. Wright KP Jr, Drake AL, Frey DJ, Fleshner M, Desouza CA, Gronfier C, et al. Influence of sleep deprivation and circadian misalignment on cortisol, inflammatory markers, and cytokine balance. Brain Behav Immunol. 2015;47:24–34.

    Article  CAS  Google Scholar 

  37. Hoirisch-Clapauch S, Brenner B. The role of the fibrinolytic system in female reproductive disorders and depression. Thromb Update. 2020;1:100004.

    Article  Google Scholar 

  38. Magariños AM, McEwen BS, Saboureau M, Pevet P. Rapid and reversible changes in intrahippocampal connectivity during the course of hibernation in European hamsters. Proc Natl Acad Sci USA. 2006;103:18775–80.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Bahi A, Dreyer JC. Hippocampus-specific deletion of tissue plasminogen activator “tPA” in adult mice impairs depression- and anxiety-like behaviors. Eur Neuropsychopharmacol. 2012;22:672–82.

    Article  CAS  PubMed  Google Scholar 

  40. Jiang H, Chen S, Li C, Lu N, Yue Y, Yin Y, et al. The serum protein levels of the tPA–BDNF pathway are implicated in depression and antidepressant treatment. Transl Psychiatry. 2017;7:e1079.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Çakici N, Sutterland AL, Penninx BW, Dalm VA, de Haan L, van Beveren NJ. Altered peripheral blood compounds in drug-naïve first-episode patients with either schizophrenia or major depressive disorder: a meta-analysis. Brain Behav Immun. 2020;88:547–58.

    Article  PubMed  Google Scholar 

  42. Leighton SP, Nerurkar L, Krishnadas R, Johnman C, Graham GJ, Cavanagh J. Chemokines in depression in health and in inflammatory illness: a systematic review and meta-analysis. Mol Psychiatry. 2018;23:48–58.

    Article  CAS  PubMed  Google Scholar 

  43. Kappelmann N, Lewis G, Dantzer R, Jones PB, Khandaker GM. Antidepressant activity of anti-cytokine treatment: a systematic review and meta-analysis of clinical trials of chronic inflammatory conditions. Mol Psychiatry. 2018;23:335–43.

    Article  CAS  PubMed  Google Scholar 

  44. Frank MG, Fonken LK, Watkins LR, Maier SF. Microglia: neuroimmune-sensors of stress. Semin Cell. Dev Biol. 2019;94:176–85.

    CAS  Google Scholar 

  45. Norris JG, Benveniste EN. Interleukin-6 production by astrocytes: induction by the neurotransmitter norepinephrine. J Neuroimmunol. 1993;45:137–45.

    Article  CAS  PubMed  Google Scholar 

  46. Del Giudice M, Gangestad SW. Rethinking IL-6 and CRP: Why they are more than inflammatory biomarkers, and why it matters. Brain Behav Immun. 2018;70:61–75.

    Article  PubMed  Google Scholar 

  47. Scheller J, Chalaris A, Schmidt-Arras D, Rose-John S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta. 2011;1813:878–88.

    Article  CAS  PubMed  Google Scholar 

  48. Miller AH, Raison CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol. 2016;16:22–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Brymer KJ, Romay-Tallon R, Allen J, Caruncho HJ, Kalynchuk LE. Exploring the potential antidepressant mechanisms of TNF-α antagonists. Front Neurosci. 2019;13:98.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Patel A. Review: the role of inflammation in depression. Psychiatr Danub. 2013;25:S216–23.

    PubMed  Google Scholar 

  51. Lamers F, Vogelzangs N, Merikangas KR, De Jonge P, Beekman AT, Penninx BW. Evidence for a differential role of HPA-axis function, inflammation and metabolic syndrome in melancholic versus atypical depression. Mol Psychiatry. 2013;18:692–9.

    Article  CAS  PubMed  Google Scholar 

  52. Sawdey M, Podor TJ, Loskutoff DJ. Regulation of type 1 plasminogen activator inhibitor gene expression in cultured bovine aortic endothelial cells. Induction by transforming growth factor‐beta, lipopolysaccharide, and tumor necrosis factor‐alpha. J Biol Chem. 1989;264:10396–401.

    Article  CAS  PubMed  Google Scholar 

  53. Arnoldussen IA, Kiliaan AJ, Gustafson DR. Obesity and dementia: adipokines interact with the brain. Eur Neuropsychopharmacol. 2014;24:1982–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Thorens B, Mueckler M. Glucose transporters in the 21st Century. Am J Physiol Endocrinol Metab. 2010;298:E141–145.

    Article  CAS  PubMed  Google Scholar 

  55. McEwen BS, Reagan LP. Glucose transporter expression in the central nervous system: relationship to synaptic function. Eur J Pharm. 2004;490:13–24.

    Article  CAS  Google Scholar 

  56. Sylow L, Kleinert M, Richter EA, Jensen TE. Exercise-stimulated glucose uptake—regulation and implications for glycaemic control. Nat Rev Endocrinol. 2017;13:133–48.

    Article  CAS  PubMed  Google Scholar 

  57. Knudsen JR, Steenberg DE, Hingst JR, Hodgson LR, Henriquez-Olguin C, Li Z, et al. Prior exercise in humans redistributes intramuscular GLUT4 and enhances insulin-stimulated sarcolemmal and endosomal GLUT4 translocation. Mol Metab. 2020;39:100998.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Pilon G, Charbonneau A, White PJ, Dallaire P, Perreault M, Kapur S, et al. Endotoxin mediated-iNOS induction causes insulin resistance via ONOO−induced tyrosine nitration of IRS-1 in skeletal muscle. PLoS ONE. 2010;5:e15912.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Crescenzo R, Mazzoli A, Di Luccia B, Bianco F, Cancelliere R, Cigliano L, et al. Dietary fructose causes defective insulin signalling and ceramide accumulation in the liver that can be reversed by gut microbiota modulation. Food Nutr Res. 2017;61:1331657.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Ding S, Chi MM, Scull BP, Rigby R, Schwerbrock NM, Magness S, et al. High-fat diet: bacteria interactions promote intestinal inflammation which precedes and correlates with obesity and insulin resistance in mouse. PLoS ONE. 2010;5:e12191.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Basciano H, Federico L, Adeli K. Fructose, insulin resistance, and metabolic dyslipidemia. Nutr Metab (Lond). 2005;2:1–4.

    Article  Google Scholar 

  62. van Loon LJ, Goodpaster BH. Increased intramuscular lipid storage in the insulin-resistant and endurance-trained state. Pflügers Arch. 2006;451:606–16.

    Article  CAS  PubMed  Google Scholar 

  63. Nielsen J, Mogensen M, Vind BF, Sahlin K, Højlund K, Schrøder HD, et al. Increased subsarcolemmal lipids in type 2 diabetes: effect of training on localization of lipids, mitochondria, and glycogen in sedentary human skeletal muscle. Am J Physiol Endocrinol Metab. 2010;298:E706–13.

    Article  CAS  PubMed  Google Scholar 

  64. Ketterer C, Tschritter O, Preissl H, Heni M, Häring HU, Fritsche A. Insulin sensitivity of the human brain. Diabetes Res Clin Pr. 2011;93:S47–51.

    Article  CAS  Google Scholar 

  65. Tang BL. Leptin as a neuroprotective agent. Biochem Biophys Res Commun. 2008;368:181–5.

    Article  CAS  PubMed  Google Scholar 

  66. Grillo CA, Woodruff JL, Macht VA, Reagan LP. Insulin resistance and hippocampal dysfunction: Disentangling peripheral and brain causes from consequences. Exp Neurol. 2019;318:71–77.

    Article  CAS  PubMed  Google Scholar 

  67. Petersen MC, Shulman GI. Mechanisms of insulin action and insulin resistance. Physiol Rev. 2018;98:2133–223.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Pérez-Pérez A, Sánchez-Jiménez F, Vilariño-García T, Sánchez-Margalet V. Role of leptin in inflammation and vice versa. Int J Mol Sci. 2020;21:5887.

    Article  PubMed Central  Google Scholar 

  69. Milaneschi Y, Lamers F, Bot M, Drent ML, Penninx BW. Leptin dysregulation is specifically associated with major depression with atypical features: evidence for a mechanism connecting obesity and depression. Biol Psychiatry. 2017;81:807–14.

    Article  CAS  PubMed  Google Scholar 

  70. Hosaka S, Yamada T, Takahashi K, Dan T, Kaneko K, Kodama S, et al. Inhibition of plasminogen activator inhibitor-1 activation suppresses high fat diet-induced weight gain via alleviation of hypothalamic leptin resistance. Front Pharm. 2020;11:943.

    Article  CAS  Google Scholar 

  71. Domenici E, Willé DR, Tozzi F, Prokopenko I, Miller S, McKeown A, et al. Plasma protein biomarkers for depression and schizophrenia by multi analyte profiling of case-control collections. PLoS ONE. 2010;5:e9166.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Paans NP, Bot M, van Strien T, Brouwer IA, Visser M, Penninx BW. Eating styles in major depressive disorder: Results from a large-scale study. J Psychiatr Res. 2018;97:38–46.

    Article  PubMed  Google Scholar 

  73. Woldeyohannes HO, Soczynska JK, Maruschak NA, Syeda K, Wium-Andersen IK, Lee Y, et al. Binge eating in adults with mood disorders: Results from the International Mood Disorders Collaborative Project. Obes Res Clin Pr. 2016;10:531–43.

    Article  Google Scholar 

  74. Iguacel I, Huybrechts I, Moreno LA, Michels N. Vegetarianism and veganism compared with mental health and cognitive outcomes: a systematic review and meta-analysis. Nutr Rev. 2021;79:361–81.

    Article  PubMed  Google Scholar 

  75. Wurtman JJ, Wurtman RJ. Depression can beget obesity can beget depression. J Clin Psychiatry. 2015;76:1619–21.

    Article  Google Scholar 

  76. Wurtman J, Wurtman R. The trajectory from mood to obesity. Curr Obes Rep. 2018;7:1–5.

    Article  PubMed  Google Scholar 

  77. Tryon MS, Laugero KD. Stress and food intake: what’s the deal with your meal. CAB Rev. 2011;6:1–11.

    Article  Google Scholar 

  78. Lustig RH. Fructose: metabolic, hedonic, and societal parallels with ethanol. J Am Diet Assoc. 2010;110:1307–21.

    Article  CAS  PubMed  Google Scholar 

  79. Magnusson KR, Hauck L, Jeffrey BM, Elias V, Humphrey A, Nath R, et al. Relationships between diet-related changes in the gut microbiome and cognitive flexibility. Neuroscience. 2015;300:128–40.

    Article  CAS  PubMed  Google Scholar 

  80. Castro MC, Massa ML, Arbeláez LG, Schinella G, Gagliardino JJ, Francini F. Fructose-induced inflammation, insulin resistance and oxidative stress: A liver pathological triad effectively disrupted by lipoic acid. Life Sci. 2015;137:1–6.

    Article  CAS  PubMed  Google Scholar 

  81. Jiménez-Maldonado A, Ying Z, Byun HR, Gomez-Pinilla F. Short-term fructose ingestion affects the brain independently from establishment of metabolic syndrome. Biochim Biophys Acta Mol Basis Dis. 2018;1864:24–33.

    Article  PubMed  Google Scholar 

  82. Arnold SE, Lucki I, Brookshire BR, Carlson GC, Browne CA, Kazi H, et al. High fat diet produces brain insulin resistance, synaptodendritic abnormalities and altered behavior in mice. Neurobiol Dis. 2014;67:79–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Scherer T, Buettner CYin. and Yang of hypothalamic insulin and leptin signaling in regulating white adipose tissue metabolism. Rev Endocr Metab Disord. 2011;12:235–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Chireh B, Li M, D’Arcy C. Diabetes increases the risk of depression: a systematic review, meta-analysis and estimates of population attributable fractions based on prospective studies. Prev Med Rep. 2019;14:100822.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Silva DA, Coutinho ED, Ferriani LO, Viana MC. Depression subtypes and obesity in adults: A systematic review and meta‐analysis. Obes Rev. 2020;21:e12966.

    Article  PubMed  Google Scholar 

  86. Mannan M, Mamun A, Doi S, Clavarino A. Is there a bi-directional relationship between depression and obesity among adult men and women? Systematic review and bias-adjusted meta analysis. Asian J Psychiatry. 2016;21:51–66.

    Article  Google Scholar 

  87. Festa A, D’Agostino R Jr, Mykkänen L, Tracy RP, Zaccaro DJ, Hales CN, et al. Relative contribution of insulin and its precursors to fibrinogen and PAI-1 in a large population with different states of glucose tolerance: the Insulin Resistance Atherosclerosis Study (IRAS). Arterioscl Thromb Vasc Biol. 1999;19:562–8.

    Article  CAS  PubMed  Google Scholar 

  88. Wang F, Wang S, Zong QQ, Zhang Q, Ng CH, Ungvari GS, et al. Prevalence of comorbid major depressive disorder in Type 2 diabetes: a meta‐analysis of comparative and epidemiological studies. Diabet Med. 2019;36:961–9.

    Article  CAS  PubMed  Google Scholar 

  89. Graham EA, Deschenes SS, Khalil MN, Danna S, Filion KB, Schmitz N. Measures of depression and risk of type 2 diabetes: A systematic review and meta-analysis. J Affect Dis. 2020;265:224–32.

    Article  PubMed  Google Scholar 

  90. Lin CE, Chung CH, Chen LF, Chien WC. Increased risk for venous thromboembolism among patients with concurrent depressive, bipolar, and schizophrenic disorders. Gen Hosp Psychiatry. 2019;61:34–40.

    Article  PubMed  Google Scholar 

  91. von Känel R, Margani A, Stauber S, Meyer FA, Biasiutti FD, Vökt F, et al. Depressive symptoms as a novel risk factor for recurrent venous thromboembolism: a longitudinal observational study in patients referred for thrombophilia investigation. PLoS ONE. 2015;10:e0125858.

    Article  Google Scholar 

  92. May HT, Horne BD, Knight S, Knowlton KU, Bair TL, Lappé DL, et al. The association of depression at any time to the risk of death following coronary artery disease diagnosis. Eur Heart J Qual Care Clin Outcomes. 2017;3:296–302.

    Article  PubMed  Google Scholar 

  93. Meijer A, Conradi HJ, Bos EH, Thombs BD, van Melle JP, de Jonge P. Prognostic association of depression following myocardial infarction with mortality and cardiovascular events: a meta-analysis of 25 years of research. Gen Hosp Psychiatry. 2011;33:203–16.

    Article  PubMed  Google Scholar 

  94. Wu Q, Kling JM. Depression and the risk of myocardial infarction and coronary death: a meta-analysis of prospective cohort studies. Med (Baltim). 2016;95:e2815.

    Article  Google Scholar 

  95. Wang Y, Meng Z, Pei J, Qian L, Mao B, Li Y, et al. Anxiety and depression are risk factors for recurrent pregnancy loss: a nested case–control study. Health Qual Life Outcomes. 2021;19:1–9.

    Article  Google Scholar 

  96. Jarde A, Morais M, Kingston D, Giallo R, MacQueen GM, Giglia L, et al. Neonatal outcomes in women with untreated antenatal depression compared with women without depression: a systematic review and meta-analysis. JAMA Psychiatry. 2016;73:826–37.

    Article  PubMed  Google Scholar 

  97. Geiser F, Conrad R, Imbierowicz K, Meier C, Liedtke R, Klingmüller D, et al. Coagulation activation and fibrinolysis impairment are reduced in patients with anxiety and depression when medicated with serotonergic antidepressants. Psychiatry Clin Neurosci. 2011;65:518–25.

    Article  CAS  PubMed  Google Scholar 

  98. Hoirisch-Clapauch S, Nardi AE. Antidepressants: bleeding or thrombosis? Thromb Res. 2019;181:23–28.

    Article  Google Scholar 

  99. Fernandes N, Prada L, Rosa MM, Ferreira JJ, Costa J, Pinto FJ, et al. The impact of SSRIs on mortality and cardiovascular events in patients with coronary artery disease and depression: systematic review and meta-analysis. Clin Res Cardiol. 2021;110:183–93.

    Article  PubMed  Google Scholar 

  100. Schoenfeld TJ, Cameron HA. Adult neurogenesis and mental illness. Neuropsychopharmacology. 2015;40:113–28.

    Article  PubMed  Google Scholar 

  101. Sartori CR, Vieira AS, Ferrari EM, Langone F, Tongiorgi E, Parada CA. The antidepressive effect of the physical exercise correlates with increased levels of mature BDNF, and proBDNF proteolytic cleavage-related genes, p11 and tPA. Neuroscience. 2011;180:9–18.

    Article  CAS  PubMed  Google Scholar 

  102. Fang W, Zhang J, Hong L, Huang W, Dai X, Ye Q, et al. Metformin ameliorates stress-induced depression-like behaviors via enhancing the expression of BDNF by activating AMPK/CREB-mediated histone acetylation. J Affect Disord. 2020;260:302–13.

    Article  CAS  PubMed  Google Scholar 

  103. Segawa M, Morinobu S, Matsumoto T, Fuchikami M, Yamawaki S. Electroconvulsive seizure, but not imipramine, rapidly up-regulates pro-BDNF and t-PA, leading to mature BDNF production, in the rat hippocampus. Int J Neuropsychopharmacol. 2013;16:339–50.

    Article  CAS  PubMed  Google Scholar 

  104. Fidalgo TM, Morales-Quezada L, Muzy GS, Chiavetta NM, Mendonca ME, Santana MV, et al. Biological markers in non-invasive brain stimulation trials in major depressive disorder: a systematic review. J ECT. 2014;30:47–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Almeida FB, Nin MS, Barros HM. The role of allopregnanolone in depressive-like behaviors: Focus on neurotrophic proteins. Neurobiol Stress. 2020;12:100218.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Zhang F, Luo J, Zhu X. Ketamine ameliorates depressive-like behaviors by tPA-mediated conversion of proBDNF to mBDNF in the hippocampus of stressed rats. Psychiatry Res. 2018;269:646–51.

    Article  CAS  PubMed  Google Scholar 

  107. Abdallah MS, Mosalam EM, Zidan AA, Elattar KS, Zaki SA, Ramadan AN, et al. The antidiabetic metformin as an adjunct to antidepressants in patients with major depressive disorder: a proof-of-concept, randomized, double-blind, placebo-controlled trial. Neurotherapeutics. 2020;17:1897–906.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Stahl SM. Placebo-controlled comparison of the selective serotonin reuptake inhibitors citalopram and sertraline. Biol Psychiatry. 2000;48:894–901.

    Article  CAS  PubMed  Google Scholar 

  109. Bolton JM, Robinson J, Sareen J. Self-medication of mood disorders with alcohol and drugs in the National Epidemiologic Survey on Alcohol and Related Conditions. J Affect Disord. 2009;115:367–75.

    Article  PubMed  Google Scholar 

  110. Janes AC, Zegel M, Ohashi K, Betts J, Molokotos E, Olson D, et al. Nicotine normalizes cortico-striatal connectivity in non-smoking individuals with major depressive disorder. Neuropsychopharmacology. 2018;43:2445–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Kandel DB, Huang FY, Davies M. Comorbidity between patterns of substance use dependence and psychiatric syndromes. Drug Alcohol Depend. 2001;64:233–4.

    Article  CAS  PubMed  Google Scholar 

  112. Fukakusa A, Nagai T, Mizoguchi H, Otsuka N, Kimura H, Kamei H, et al. Role of tissue plasminogen activator in the sensitization of methamphetamine‐induced dopamine release in the nucleus accumbens. J Neurochem. 2008;105:436–44.

    Article  CAS  PubMed  Google Scholar 

  113. Beloate LN, Kalivas PW. Role of the extracellular matrix in addiction. In: Torregrossa M, editor. Neural mechanisms of addiction. Academic Press; 2019, p. 247–58.

  114. Jin H, Lin J, Fu L, Mei YF, Peng G, Tan X, et al. Physiological testosterone stimulates tissue plasminogen activator and tissue factor pathway inhibitor and inhibits plasminogen activator inhibitor type 1 release in endothelial cells. Biochem Cell Biol. 2007;85:246–51.

    Article  CAS  PubMed  Google Scholar 

  115. Pirl WF, Siegel GI, Goode MJ, Smith MR. Depression in men receiving androgen deprivation therapy for prostate cancer: a pilot study. Psycho‐Oncol. 2002;11:518–23.

    Article  Google Scholar 

  116. Walther A, Breidenstein J, Miller R. Association of testosterone treatment with alleviation of depressive symptoms in men: a systematic review and meta-analysis. JAMA Psychiatry. 2019;76:31–40.

    Article  PubMed  Google Scholar 

  117. VanLandingham JW, Cekic M, Cutler SM, Hoffman SW, Washington ER, et al. Progesterone and its metabolite allopregnanolone differentially regulate hemostatic proteins after traumatic brain injury. J Cereb Blood Flow Metab. 2008;28:1786–94.

    Article  CAS  PubMed  Google Scholar 

  118. Moradi F, Lotfi K, Armin M, Clark CC, Askari G, Rouhani MH. The association between serum homocysteine and depression: a systematic review and meta‐analysis of observational studies. Eur J Clin Invest. 2021;51:e13486.

    Article  CAS  PubMed  Google Scholar 

  119. Cesarman-Maus C, Hajjar KA. Molecular mechanisms of fibrinolysis. Br J Haematol. 2015;129:307–21.

    Article  Google Scholar 

  120. Feelders RA, Pulgar SJ, Kempel A, Pereira AM. The burden of Cushing’s disease: clinical and health-related quality of life aspects. Eur J Endocrinol. 2012;167:311–26.

    Article  CAS  PubMed  Google Scholar 

  121. Brown ES, Varghese FP, McEwen BS. Association of depression with medical illness: does cortisol play a role? Biol Psychiatry. 2004;55:1–9.

    Article  CAS  PubMed  Google Scholar 

  122. Isidori AM, Balercia G, Calogero AE, Corona G, Ferlin A, Francavilla S, et al. Outcomes of androgen replacement therapy in adult male hypogonadism: recommendations from the Italian Society of Endocrinology. J Endocrinol Invest. 2015;38:103–12.

    Article  CAS  PubMed  Google Scholar 

  123. Meroni M, Longo M, Dongiovanni P. Alcohol or gut microbiota: who is the guilty? Intern J Mol Sci. 2019;20:4568.

    Article  CAS  Google Scholar 

  124. Walther A, Rice T, Kufert Y, Ehlert U. Neuroendocrinology of a male-specific pattern for depression linked to alcohol use disorder and suicidal behavior. Front Psychiatry. 2017;7:206.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Tilg H, Diehl AM. Cytokines in alcoholic and nonalcoholic steatohepatitis. N Engl J Med. 2000;343:1467–76.

    Article  CAS  PubMed  Google Scholar 

  126. Wieërs G, Belkhir L, Enaud R, Leclercq S, de Foy JMP, Dequenne I, et al. How probiotics affect the microbiota. Front Cell Infect Microbiol. 2020;9:454.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Vrieze A, Van Nood E, Holleman F, Salojärvi J, Kootte RS, Bartelsman JF, et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology. 2012;143:913–6.

    Article  CAS  PubMed  Google Scholar 

  128. Kootte RS, Levin E, Salojärvi J, Smits LP, Hartstra AV, Udayappan SD, et al. Improvement of insulin sensitivity after lean donor feces in metabolic syndrome is driven by baseline intestinal microbiota composition. Cell Metab. 2017;26:611–9.

    Article  CAS  PubMed  Google Scholar 

  129. Messaoudi M, Lalonde R, Violle N, Javelot H, Desor D, Nejdi A, et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br J Nutr. 2011;105:755–64.

    Article  CAS  PubMed  Google Scholar 

  130. Meyyappan AC, Forth E, Wallace CJ, Milev R. Effect of fecal microbiota transplant on symptoms of psychiatric disorders: a systematic review. BMC Psychiatry. 2020;20:1–9.

    Google Scholar 

Download references

Acknowledgements

The author is deeply grateful to Dr. Jacqueline A. Menezes, Professor Olavo Bohrer Amaral, Dr. Sylvio S. Neves Provenzano, and Professor Ian A. Greer for their invaluable suggestions.

Author information

Authors and Affiliations

Authors

Contributions

The author’s contributions include conception and design of the hypothesis, drafting the manuscript, revising the manuscript critically for important intellectual content, and approval of the version of the manuscript to be published.

Corresponding author

Correspondence to Silvia Hoirisch-Clapauch.

Ethics declarations

Competing interest

The author declares no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hoirisch-Clapauch, S. Mechanisms affecting brain remodeling in depression: do all roads lead to impaired fibrinolysis?. Mol Psychiatry 27, 525–533 (2022). https://doi.org/10.1038/s41380-021-01264-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-021-01264-1

This article is cited by

Search

Quick links