top of page

The Problem with Finasteride

Click here to return to the previous article in this series.

Click here to watch the companion YouTube video for this series.


In the previous article in this series, we discussed the metabolism of neuroactive steroids and their cognitive and behavioral impacts. If you have not read it, this article will be less valuable.


In this article, we will review the modern evidence on finasteride’s side effects, with a particular focus on its cognitive and behavioral effects.


FINASTERIDE & ITS ENZYMATIC TARGET


Finasteride is an inhibitor of the enzyme 5α-reductase (5AR). It was developed in the 1980’s[1] for the treatment of benign prostatic hyperplasia (BPH), characterized by the growth of the prostate in response to androgenic hormones. BPH affects about 40 million men in the United States[2]. In addition to its use for BPH, finasteride is one of only two FDA-approved medications for the treatment of androgenic alopecia, the miniaturization of hair follicles that results from androgen exposure over a man’s lifetime[3].


Recall that steroids are synthesized in the human body from sterols in the adrenals, gonads, and brain. Cholesterol is first converted into pregnenolone by the cholesterol side-chain cleavage enzyme (P450scc), from which all other steroids are derived through various enzymes. The target of finasteride, 5AR, converts deoxycorticosterone into dihydrodeoxycorticosterone (DHDOC), progesterone into dihydroprogesterone (DHP), and testosterone into dihydrotestosterone (DHT)[4]. Consequently, inhibiting 5AR also inhibits the synthesis of the neurosteroids androsterone, androstanediol, THDOC, allopregnanolone, and isopregnanolone.


5AR has three isoforms, the second and third of which are strongly inhibited by finasteride. Dutasteride, a 5AR inhibitor with growing popularity, inhibits all three isoforms strongly[5]. The reader will remember that the type 1 isoform that finasteride inhibits less strongly than dutasteride is the main 5AR isoform in the brain. Consequently, dutasteride exhibits greater cognitive and behavioral side effects than finasteride. Nonetheless, neurosteroids are regularly imported into the brain through the blood-brain barrier, so any inhibition of 5AR will reduce the prevalence of neurosteroids in the brain.


Of all the natural steroids, DHT is the most powerful agonist of the androgen receptor (AR) and thus has the greatest potential to induce androgenic side effects in men and women. Androgens bind to the androgen receptor in the cell cytoplasm. The binding activates the receptor, allowing it to translocate into the nucleus, homodimerize, and act as a transcription factor, binding DNA response elements called androgen response elements (AREs) present in different target genes – essentially, changing gene expression in the body[6]. Androgen receptors in the nuclei can modulate about 500 androgen response elements (AREs) and 200 androgen receptor-responsive genes[7].


Because of their effect on the androgen receptor, both testosterone and DHT cause dermal papilla cell apoptosis in a dose-dependent and time-related manner[8]. Finasteride reduces the conversion of testosterone into the more potent androgen DHT, and can also bind lightly at the androgen receptor, reducing androgenic activity generally[9]. At a dose of 1 mg taken daily, finasteride can prevent the progression of androgenic alopecia and benign prostate hyperplasia[10].


For the first two decades of its use, finasteride was thought to have mostly benign side effects, as researchers were impressed with its marginal effect on semen parameters and testosterone levels in healthy patients. Between 2010 and 2015, the first literature describing persistent side effects from finasteride were published, and in 2010 Merck included in their drug sheet a note that, rarely, persistent issues with erection had been observed in post-finasteride users. In 2012 the FDA mandated that the list of persistent side effects be expanded further[11].


FERTILITY, METABOLIC SYNDROME, AND AGING PHALLUSES


The major unwanted effects of finasteride involve fertility changes, metabolic syndrome, and psychological effects. Though this review will focus on finasteride’s effects on cognition and behavior, it is worthwhile to briefly discuss the other effects.


Finasteride was originally thought not to affect semen parameters. The first major randomized, double-blind, placebo-controlled study indicated that 1 mg finasteride did not affect semen parameters[12], but it only studied men without prior fertility problems. Prompted by case reports of men experiencing severe reductions in sperm quality, a later study examined 27 men who reported to an infertility clinic. It concluded that 1 mg/day of finasteride does worsen semen parameters in men with pre-existing fertility issues[13]. It appears that while most men have minimal semen quality changes on finasteride, about 5% are hypersensitive to it and experience a dramatic reduction in sperm count and quality consequent to treatment[14].


In the last decade, finasteride has also been shown to produce symptoms of the metabolic syndrome in men and rodents[15]. Specifically, 5AR inhibition appears to worsen insulin sensitivity as measured by glucose metrics, liver health as measured by ALT and AST enzymes, and low-density lipoprotein cholesterol levels, in the long-term. Because a lot of the metabolic effect of 5AR inhibition appears to depend on the type 1 isoform of 5AR, the effect is more pronounced with dutasteride[16] use, though it is also evident with finasteride[17][18].


Though the role of steroids in metabolic and lipid health is less clear, physiologic levels of androgens appear necessary for the maintenance of liver health. It is established that androgen deprivation influences the development of liver disease[19], while androgen treatment reduces fatty liver disease[20][21][22]. Mice lacking hepatic androgen receptors exhibit greater hepatic steatosis[23], and hepatic steatosis is associated with low serum testosterone and high serum DHEA[24] in humans. On the other hand, super-physiologic doses of androgens are associated with the development of liver tumors and hepatocellular carcinoma[25].


Note that there are other, less well-known effects of androgen deprivation. For example, the observed reduction in erection quality in finasteride-treated patients has led academics to question whether the androgen-deprivation may be aging men prematurely. Consequent to androgen deprivation, post-finasteride patients exhibit upregulated androgen receptor density in penile skin tissue up to 8 years after discontinuation of the treatment[26], and androgen receptors were found to be upregulated in other tissues, including the prostate[27]. Hypoandrogenism is thought to cause fibrosis in the corpora cavernosa of penile tissue through collagen fiber deposition and the inhibition of nitric oxide synthases[28][29]. Moreover, 5AR inhibition induced smooth muscle cell apoptosis in the corpora cavernosa in rats[30], leading researchers to speculate that it could cause fibrosis and apoptosis in human penile tissue.


FINASTERIDE, THE PLASMA, AND CEREBROSPINAL FLUID


The most concerning side effects observed in finasteride-treated patients have been psychological and behavioral changes. Particularly, patients often exhibit an anxious or depressed[31] phenotype and report chronic fatigue, loss of libido, sleep problems, decreased initiative, and difficulty concentrating[32].


Though it is obvious that downstream effects of 5AR inhibition on steroid synthesis may be the culprit behind the observed cognitive and behavioral symptoms, most studies only examine plasma levels of steroids. For good reason, patients are less willing to have their spine’s punctured to donate cerebrospinal fluid (CSF) for academics. Nonetheless, a team of Italian researchers managed to convince three groups of patients to do exactly that. This series of three studies will provide the basis for our analysis of the effects of finasteride on neuroactive steroids. Because they are necessarily small due to the invasiveness of the procedure, because of their weak study design, and because the psychological symptoms exist in complex systems, we will have to compare the results with what we know of the biochemistry of finasteride and neuroactive steroids. Here, I report only results that were statistically significant.


In 2013, Melcangi et al.[33] began the series of studies with only 3 post-finasteride patient volunteers and compared their plasma and CSF results to 5 healthy volunteers. On average, the three patients had used finasteride for 3.6 years and ceased treatment 5.9 years before commencement of the study. Due to the small study size, the only statistically significant outcomes in plasma were undetectable DHP, raised androstanediol, and raised estrogen, while in CSF allopregnanolone and isopregnanolone were undetectable, testosterone was raised, and estrogen was raised.


In 2014, Caruso et al. [34] examined the plasma and CSF of 7 post-finasteride patients with persistent symptomology and compared their results to 12 healthy volunteers. On average, patients used finasteride for 2 years and discontinued treatment 4.4 years prior to the study. In this study, DHEA was unchanged in both CSF and plasma while estradiol was again raised in plasma. Androstanediol was raised in both plasma and CSF. Progesterone was reduced in CSF, its metabolite DHP was reduced in plasma and undetectable in CSF, allopregnanolone was undetectable in both CSF and plasma, and isopregnanolone was raised in plasma. Pregnenolone was raised in both CSF and plasma.


In 2017, Melcangi et al. [35] analyzed the plasma and CSF of 14 post-finasteride patients who reported persistent side effects and compared them to a control of 25 healthy subjects. The post-finasteride patients used finasteride for 2.8 years and had discontinued treatment 5.4 years earlier, on average. The 14 subjects exhibited increased DHEA and testosterone in both CSF and plasma and increased androstanediol in CSF. However, they exhibited lower CSF estradiol and progesterone, though plasma levels were unaffected. Of the progesterone metabolites, DHP was lowered in both CSF and plasma while allopregnanolone was undetectable in plasma. The post-finasteride patients also exhibited lower pregnenolone levels in the CSF, but higher pregnenolone levels in plasma.


Though there are major differences in the outcomes of the studies, there are also observable patterns. Unfortunately, this team of researchers did not measure THDOC, androsterone, or the sulfated forms of pregnenolone and DHEA. Nevertheless, progesterone and its critical neurosteroid metabolite allopregnanolone were consistently lower. Interestingly, DHEA and testosterone were either raised or unchanged, and androstanediol was consistently raised, though DHT was consistently lower.


CSF steroid measures are particularly relevant to behavior and cognition because they represent hormones that have either passed the blood-brain-spinal cord barrier or been synthesized in nervous or brain tissue. Progesterone and its metabolite allopregnanolone are consistently lowered in former finasteride users with persistent symptomology. This is in line with studies on current finasteride users, while dutasteride totally inhibits allopregnanolone synthesis as measured in plasma, even in women[36].


While the causal role of finasteride on progestogen levels cannot be determined, we know from the previous article in this series that improved progestogen activity improves the reported symptomology.


FINASTERIDE THWARTS THE ACTIVITY OF NEUROSTEROIDS


In 2016, a study examined 25 sexually symptomatic post-finasteride patients, asymptomatic post-finasteride patients, and a control. Finasteride was used for between a year and two years in the two groups and had been ceased between 3 and 3.5 years in the two groups. The authors performed fMRI scans of the patients brains and observed functional abnormalities among the sexually symptomatic patients that resembled functional abnormalities common to depressed patients[37], though the patients had been selected for sexual symptoms.


In adulthood, the hippocampus is one of only two regions to experience the birth of new neurons – neurogenesis[38]. Depressed people exhibit reduced hippocampal volume with reduced dendritic complexity, decreased neuronal soma size, and reduced hippocampal neurogenesis[39]. Beginning two decades ago, neuroscientists have grown to suspect that reduced neurogenesis is not a symptom of depression but a causal factor in its development[40]. Strikingly, finasteride has been shown to inhibit neurogenesis in the male rodent brain[41], while several neurosteroids appear to increase neurogenesis.


Selective serotonin reuptake inhibitors (SSRIs) are thought to improve depressive symptoms through their upregulation of brain growth factors like brain-derived neurotrophic factor (BDNF) that increase hippocampal neurogenesis[42]. Recall that SSRIs also raised CSF measures of the important progesterone metabolite allopregnanolone[43][44][45][46]. Allopregnanolone, which was decreased in all of the CSF studies on finasteride, is also diminished in women with post-traumatic stress disorder (PTSD)[47], in depressed pregnant women[48], in people with anxiety disorders and impulse aggression, and in schizophrenics with negative symptoms[49]. Consequently, causality is difficult to speculate on.


Caruso et al. observed only one elevated progesterone metabolite in plasma – isopregnanolone. Isopregnanolone is elevated in women with chronic fatigue syndrome[50], and in depressives, it is elevated in plasma while pregnanolone and allopregnanolone are reduced. SSRI treatment not only raises allopregnanolone[51], but it also raises plasma pregnanolone and decreases isopregnanolone after recovery, while pregnenolone, DHP, and progesterone are left unchanged[52]. Recall that isopregnanolone has an opposing modulatory effect on GABAA receptors, as compared to its sister metabolite allopregnanolone.


Finasteride has also been observed to reduce dopaminergic activity in the brain, in line with our previous discussion of THDOC and/or allopregnanolone’s modulation of dopamine receptors. In rats, finasteride reduces dopaminergic activity[53]. Dutasteride, but not finasteride, can protect mice from the development of Parkinson’s disease due to the neurotoxic effects of MPTP, a molecule used to replicable the etiology of Parkinson’s disease. Dutasteride is protective of the dopaminergic neurons through its inhibitory effect on dopaminergic processes[54]. In an animal study[55], finasteride was observed to decrease the dopaminergic effect of alcohol.


CONCLUSION


In this article, we reviewed the evidence for finasteride’s semen, sexual, metabolic, cognitive, and behavioral effects. We examined the most detailed studies on biomarkers of post-finasteride treatment patients, which revealed consistently lower progesterone and allopregnanolone levels. Though we cannot be certain finasteride lowered allopregnanolone, as it is broadly observed to be reduced in depressed patients, we have enough evidence to assume that progesterone and allopregnanolone contribute causally to cognitive performance and mental well-being.


In the concluding article of this series, we will speculate on a protocol to improve finasteride-dependent symptomology.


Click here to read the final article.

[1] Stoner, E. (1990). The clinical development of a 5α-reductase inhibitor, finasteride. The Journal of steroid biochemistry and molecular biology, 37(3), 375-378. [2] Vuichoud, C., & Loughlin, K. R. (2015). Benign prostatic hyperplasia: epidemiology, economics and evaluation. Can J Urol, 22(Suppl 1), 1-6. [3] Adil, A., & Godwin, M. (2017). The effectiveness of treatments for androgenetic alopecia: a systematic review and meta-analysis. Journal of the American Academy of Dermatology, 77(1), 136-141. [4] Schumacher, M., Hussain, R., Gago, N., Oudinet, J. P., Mattern, C., & Ghoumari, A. (2012). Progesterone synthesis in the nervous system: implications for myelination and myelin repair. Frontiers in neuroscience, 6, 10. [5] Skeldon, S. C., Macdonald, E. M., Law, M. R., Huang, A., Paterson, J. M., Mamdani, M. M., & Juurlink, D. (2017). The cardiovascular safety of dutasteride. The Journal of urology, 197(5), 1309-1314. [6] Lee, H. J., & Chang, C. (2003). Recent advances in androgen receptor action. Cellular and Molecular Life Sciences CMLS, 60(8), 1613-1622. [7] Bolton, E. C., So, A. Y., Chaivorapol, C., Haqq, C. M., Li, H., & Yamamoto, K. R. (2007). Cell-and gene-specific regulation of primary target genes by the androgen receptor. Genes & development, 21(16), 2005-2017. [8] Winiarska, A., Mandt, N., Kamp, H., Hossini, A., Seltmann, H., Zouboulis, C. C., & Blume-Peytavi, U. (2006). Effect of 5α-dihydrotestosterone and testosterone on apoptosis in human dermal papilla cells. Skin pharmacology and physiology, 19(6), 311-321. [9] Chhipa, R. R., Halim, D., Cheng, J., Zhang, H. Y., Mohler, J. L., Ip, C., & Wu, Y. (2013). The direct inhibitory effect of dutasteride or finasteride on androgen receptor activity is cell line specific. The Prostate, 73(14), 1483-1494. [10] Mella, J. M., Perret, M. C., Manzotti, M., Catalano, H. N., & Guyatt, G. (2010). Efficacy and safety of finasteride therapy for androgenetic alopecia: a systematic review. Archives of dermatology, 146(10), 1141-1150. [11] Di Loreto, C., La Marra, F., Mazzon, G., Belgrano, E., Trombetta, C., & Cauci, S. (2014). Immunohistochemical evaluation of androgen receptor and nerve structure density in human prepuce from patients with persistent sexual side effects after finasteride use for androgenetic alopecia. PLoS One, 9(6). [12] Overstreet, J. W., Fuh, V. L., Gould, J., Howards, S. S., Lieber, M. M., Hellstrom, W., ... & LEWIS, R. (1999). Chronic treatment with finasteride daily does not affect spermatogenesis or semen production in young men. The Journal of urology, 162(4), 1295-1300. [13] Samplaski, M. K., Lo, K., Grober, E., & Jarvi, K. (2013). Finasteride use in the male infertility population: effects on semen and hormone parameters. Fertility and sterility, 100(6), 1542-1546. [14] Amory, J. K., Wang, C., Swerdloff, R. S., Anawalt, B. D., Matsumoto, A. M., Bremner, W. J., ... & Clark, R. V. (2007). The effect of 5α-reductase inhibition with dutasteride and finasteride on semen parameters and serum hormones in healthy men. The Journal of Clinical Endocrinology & Metabolism, 92(5), 1659-1665. [15] Traish, A. M. (2020). Health Risks Associated with Long-Term Finasteride and Dutasteride Use: It's Time to Sound the Alarm. The World Journal of Men's Health, 38. [16] Traish, A., Haider, K. S., Doros, G., & Haider, A. (2017). Long-term dutasteride therapy in men with benign prostatic hyperplasia alters glucose and lipid profiles and increases severity of erectile dysfunction. Hormone molecular biology and clinical investigation, 30(3). [17] Doulabi, S. R. S. M., Kavoussi, H., Isapour, D., Hashemian, A., & Taheriniya, A. (2013). Effect of finasteride on lipid profile in individuals with androgenetic alopecia. African Journal of Pharmacy and Pharmacology, 7(6), 315-317. [18] Duskova, M., Hill, M., & Starka, L. (2010). Changes of metabolic profile in men treated for androgenetic alopecia with 1 mg finasteride. Endocr Regul, 44(1), 3-8. [19] Gild, P., Cole, A. P., Krasnova, A., Dickerman, B. A., von Landenberg, N., Sun, M., ... & Kibel, A. S. (2018). Liver disease in men undergoing androgen deprivation therapy for prostate cancer. The Journal of urology, 200(3), 573-581. [20] Mendenhall, C. L. (1968). Anabolic steroid therapy as an adjunct to diet in alcoholic hepatic steatosis. The American journal of digestive diseases, 13(9), 783-791. [21] Jacqueson, A., Thevenin, M., Warnet, J. M., Claude, J. R., & Truhaut, R. (1978). Comparative Study of the Protective Effect of an Anabolic Steroid. In Toxicological Aspects of Food Safety (pp. 193-196). Springer, Berlin, Heidelberg. [22] Kelly, D. M., Nettleship, J. E., Akhtar, S., Muraleedharan, V., Sellers, D. J., Brooke, J. C., ... & Jones, T. H. (2014). Testosterone suppresses the expression of regulatory enzymes of fatty acid synthesis and protects against hepatic steatosis in cholesterol-fed androgen deficient mice. Life sciences, 109(2), 95-103. [23] Lin, H. Y., Yu, I. C., Wang, R. S., Chen, Y. T., Liu, N. C., Altuwaijri, S., ... & Yeh, S. (2008). Increased hepatic steatosis and insulin resistance in mice lacking hepatic androgen receptor. Hepatology, 47(6), 1924-1935. [24] Völzke, H., Aumann, N., Krebs, A., Nauck, M., Steveling, A., Lerch, M. M., ... & Wallaschofski, H. (2010). Hepatic steatosis is associated with low serum testosterone and high serum DHEAS levels in men. International journal of andrology, 33(1), 45-53. [25] Velazquez, I., & Alter, B. P. (2004). Androgens and liver tumors: Fanconi's anemia and non‐Fanconi's conditions. American journal of hematology, 77(3), 257-267. [26] Di Loreto, C., La Marra, F., Mazzon, G., Belgrano, E., Trombetta, C., & Cauci, S. (2014). Immunohistochemical evaluation of androgen receptor and nerve structure density in human prepuce from patients with persistent sexual side effects after finasteride use for androgenetic alopecia. PLoS One, 9(6). [27] Hsieh, J. T., Chen, S. C., Yu, H. J., & Chang, H. C. (2011). Finasteride upregulates expression of androgen receptor in hyperplastic prostate and LNCaP cells: implications for chemoprevention of prostate cancer. The Prostate, 71(10), 1115-1121. [28] Pinsky, M. R., Gur, S., Tracey, A. J., Harbin, A., & Hellstrom, W. J. (2011). The effects of chronic 5‐alpha‐reductase inhibitor (dutasteride) treatment on rat erectile function. The journal of sexual medicine, 8(11), 3066-3074. [29] Iacono, F., Prezioso, D., Ruffo, A., Illiano, E., Romis, L., Di Lauro, G., ... & Amato, B. (2012). Testosterone deficiency causes penile fibrosis and organic erectile dysfunction in aging men. Evaluating association among Age. BMC surgery, 12(S1), S24. [30] Zhang, M. G., Wang, X. J., Shen, Z. J., & Gao, P. J. (2013). Long-term oral administration of 5α-reductase inhibitor attenuates erectile function by inhibiting autophagy and promoting apoptosis of smooth muscle cells in corpus cavernosum of aged rats. Urology, 82(3), 743-e9. [31] Melcangi, R. C., Caruso, D., Abbiati, F., Giatti, S., Calabrese, D., Piazza, F., & Cavaletti, G. (2013). Neuroactive steroid levels are modified in cerebrospinal fluid and plasma of post‐finasteride patients showing persistent sexual side effects and anxious/depressive symptomatology. The journal of sexual medicine, 10(10), 2598-2603. [32] Alanazi, N. (2016). Assessing the quality of life and health outcomes of Androgenic Alopecia patients using Propecia (Doctoral dissertation, Rutgers University-School of Health Related Professions). [33] Melcangi, R. C., Caruso, D., Abbiati, F., Giatti, S., Calabrese, D., Piazza, F., & Cavaletti, G. (2013). Neuroactive steroid levels are modified in cerebrospinal fluid and plasma of post‐finasteride patients showing persistent sexual side effects and anxious/depressive symptomatology. The journal of sexual medicine, 10(10), 2598-2603. [34] Caruso, D., Abbiati, F., Giatti, S., Romano, S., Fusco, L., Cavaletti, G., & Melcangi, R. C. (2015). Patients treated for male pattern hair with finasteride show, after discontinuation of the drug, altered levels of neuroactive steroids in cerebrospinal fluid and plasma. The Journal of steroid biochemistry and molecular biology, 146, 74-79. [35] Melcangi, R. C., Santi, D., Spezzano, R., Grimoldi, M., Tabacchi, T., Fusco, M. L., ... & Simoni, M. (2017). Neuroactive steroid levels and psychiatric and andrological features in post-finasteride patients. The Journal of steroid biochemistry and molecular biology, 171, 229-235. [36] Martinez, P. E., Rubinow, D. R., Nieman, L. K., Koziol, D. E., Morrow, A. L., Schiller, C. E., ... & Schmidt, P. J. (2016). 5α-reductase inhibition prevents the luteal phase increase in plasma allopregnanolone levels and mitigates symptoms in women with premenstrual dysphoric disorder. Neuropsychopharmacology, 41(4), 1093. [37] Basaria, S., Jasuja, R., Huang, G., Wharton, W., Pan, H., Pencina, K., ... & Labrie, F. (2016). Characteristics of men who report persistent sexual symptoms after finasteride use for hair loss. The Journal of Clinical Endocrinology & Metabolism, 101(12), 4669-4680. [38] Van Praag, H., Schinder, A. F., Christie, B. R., Toni, N., Palmer, T. D., & Gage, F. H. (2002). Functional neurogenesis in the adult hippocampus. Nature, 415(6875), 1030-1034. [39] Stockmeier, C. A., Mahajan, G. J., Konick, L. C., Overholser, J. C., Jurjus, G. J., Meltzer, H. Y., ... & Rajkowska, G. (2004). Cellular changes in the postmortem hippocampus in major depression. Biological psychiatry, 56(9), 640-650. [40] Jacobs, B. L., Van Praag, H., & Gage, F. H. (2000). Adult brain neurogenesis and psychiatry: a novel theory of depression. Molecular psychiatry, 5(3), 262-269. [41] Römer, B., Pfeiffer, N., Lewicka, S., Ben-Abdallah, N., Vogt, M. A., Deuschle, M., ... & Gass, P. (2010). Finasteride treatment inhibits adult hippocampal neurogenesis in male mice. Pharmacopsychiatry, 43(05), 174-178. [42] Samuels, B. A., & Hen, R. (2011). Neurogenesis and affective disorders. European Journal of Neuroscience, 33(6), 1152-1159. [43] Romeo, E., Ströhle, A., Spalletta, G., Michele, F. D., Hermann, B., Holsboer, F., ... & Rupprecht, R. (1998). Effects of antidepressant treatment on neuroactive steroids in major depression. American Journal of Psychiatry, 155(7), 910-913. [44] Uzunova, V., Sampson, L., & Uzunov, D. P. (2006). Relevance of endogenous 3α-reduced neurosteroids to depression and antidepressant action. Psychopharmacology, 186(3), 351-361. [45] Uzunova, V., Sheline, Y., Davis, J. M., Rasmusson, A., Uzunov, D. P., Costa, E., & Guidotti, A. (1998). Increase in the cerebrospinal fluid content of neurosteroids in patients with unipolar major depression who are receiving fluoxetine or fluvoxamine. Proceedings of the National Academy of Sciences, 95(6), 3239-3244. [46] Schüle, C., Nothdurfter, C., & Rupprecht, R. (2014). The role of allopregnanolone in depression and anxiety. Progress in neurobiology, 113, 79-87. [47] Rasmusson, A. M., Pinna, G., Paliwal, P., Weisman, D., Gottschalk, C., Charney, D., ... & Guidotti, A. (2006). Decreased cerebrospinal fluid allopregnanolone levels in women with posttraumatic stress disorder. Biological psychiatry, 60(7), 704-713. [48] Hellgren, C., Åkerud, H., Skalkidou, A., Bäckström, T., & Sundström-Poromaa, I. (2014). Low serum allopregnanolone is associated with symptoms of depression in late pregnancy. Neuropsychobiology, 69(3), 147-153. [49] Schüle, C., Nothdurfter, C., & Rupprecht, R. (2014). The role of allopregnanolone in depression and anxiety. Progress in neurobiology, 113, 79-87. [50] Murphy, B. P., Abbott, F. V., Allison, C. M., Watts, C., & Ghadirian, A. M. (2004). Elevated levels of some neuroactive progesterone metabolites, particularly isopregnanolone, in women with chronic fatigue syndrome. Psychoneuroendocrinology, 29(2), 245-268. [51] Uzunova, V., Sheline, Y., Davis, J. M., Rasmusson, A., Uzunov, D. P., Costa, E., & Guidotti, A. (1998). Increase in the cerebrospinal fluid content of neurosteroids in patients with unipolar major depression who are receiving fluoxetine or fluvoxamine. Proceedings of the National Academy of Sciences, 95(6), 3239-3244. [52] Romeo, E., Ströhle, A., Spalletta, G., Michele, F. D., Hermann, B., Holsboer, F., ... & Rupprecht, R. (1998). Effects of antidepressant treatment on neuroactive steroids in major depression. American Journal of Psychiatry, 155(7), 910-913. [53] Li, L., Kang, Y. X., Ji, X. M., Li, Y. K., Li, S. C., Zhang, X. J., ... & Shi, G. M. (2018). Finasteride inhibited brain dopaminergic system and open‐field behaviors in adolescent male rats. CNS neuroscience & therapeutics, 24(2), 115-125. [54] Litim, N., Bourque, M., Al Sweidi, S., Morissette, M., & Di Paolo, T. (2015). The 5α-reductase inhibitor Dutasteride but not Finasteride protects dopamine neurons in the MPTP mouse model of Parkinson's disease. Neuropharmacology, 97, 86-94. [55] Dazzi, L., Serra, M., Seu, E., Cherchi, G., Pisu, M. G., Purdy, R. H., & Biggio, G. (2002). Progesterone enhances ethanol‐induced modulation of mesocortical dopamine neurons: antagonism by finasteride. Journal of neurochemistry, 83(5), 1103-1109.

bottom of page