Elsevier

Psychoneuroendocrinology

Volume 99, January 2019, Pages 206-215
Psychoneuroendocrinology

Treatment of male rats with finasteride, an inhibitor of 5alpha-reductase enzyme, induces long-lasting effects on depressive-like behavior, hippocampal neurogenesis, neuroinflammation and gut microbiota composition

https://doi.org/10.1016/j.psyneuen.2018.09.021Get rights and content

Highlights

  • One month of finasteride withdrawal induced depressive-like behavior.

  • Decrease of neurogenesis occurred after one month of finasteride withdrawal.

  • GFAP immunoreactivity was increased after one month of finasteride withdrawal.

  • Finasteride treatment and its withdrawal induced neuroinflammation in hippocampus.

  • Gut microbiota composition was affected by finasteride treatment and its withdrawal.

Abstract

Persistent alteration of plasma neuroactive steroid levels associated with major depression has been recently reported in men after the suspension of the treatment for androgenetic alopecia with finasteride, an inhibitor of the enzyme 5alpha-reductase. Observations in male rats confirmed persistent alterations in neuroactive steroid levels also in the brain. In the present study, we have ascertained possible effects on depressive-like behavior, neurogenesis, gliosis, neuroinflammation and gut microbiota in male rats after subchronic treatment for 20 days with finasteride and after one month of its withdrawal. At the end of treatment there was an increase in the number of pH3 immunoreactive cells in the subgranular zone of the dentate gyrus together with an increase in the mRNA levels of TNF-α in the hippocampus. By one month after the end of finasteride treatment, rats showed depressive-like behavior coupled with a decrease in the number of pH3 immunoreactive cells in the subgranular zone of the dentate gyrus, a decrease in granule cell density in the granule cell layer and an increase in the number of GFAP immunoreactive astrocytes in the dentate gyrus. Finally, alteration of gut microbiota (i.e., an increase in Bacteroidetes phylum and in Prevotellaceae family at the end of the treatment and a decrease in Ruminococcaceae family, Oscillospira and Lachnospira genus at the end of the withdrawal period) was detected.

In conclusion, finasteride treatment in male rats has long term effects on depressive-like behavior, hippocampal neurogenesis and neuroinflammation and gut microbiota composition.

Introduction

Neuroactive steroids are those steroids that modulate the function of the nervous system. They are important physiological neuromodulators and include steroids synthesized in the peripheral glands as well as those directly produced by the nervous tissue. Neuroactive steroids are involved in the neuroendocrine control of reproduction and regulate synaptic plasticity, neuronal and glial morphology, adult neurogenesis, myelination and cognition (Galea, 2008; Melcangi et al., 2016). Accordingly, psychiatric disorders, such as depression and anxiety, are associated with altered levels of neuroactive steroids, and particularly of 5α-reduced metabolites of testosterone and progesterone, in plasma and/or cerebrospinal fluid (CSF) (Backstrom et al., 2014; Schule et al., 2014).

Finasteride, an inhibitor of the enzyme 5α-reductase (5α-R), blocks the conversion of testosterone and progesterone, into their 5α-reduced metabolites (dihydrotestosterone, DHT, and dihydroprogesterone, DHP, respectively). For this reason, this drug is used for the treatment of benign prostatic hyperplasia (BPH) and androgenetic alopecia (AGA). Indeed, these disorders are associated with high levels of DHT in prostate and in hair follicles of BPH and AGA patients, respectively. By inhibiting 5α-R activity, finasteride reduces DHT levels in prostate and in hair follicles and improve the conditions of BPH and AGA (Traish et al., 2015).

Despite of the wide therapeutic use of finasteride in humans, its effects in the nervous system have been poorly explored. That is particularly important, because a subset of men taking finasteride for AGA show during the treatment a wide symptomatology including depression, erectile dysfunction, endocrine alterations and musculoskeletal manifestations (Giatti et al., 2018). Moreover, as self-reported by the patients (Altomare and Capella, 2002; Irwig, 2012; Rahimi-Ardabili et al., 2006; Traish et al., 2015) and as recently ascertained by two different clinical studies, these important side effects may persist even after discontinuation of the treatment (Basaria et al., 2016; Melcangi et al., 2017). Thus, these patients are affected by the so called post-finasteride syndrome (PFS). In particular, as reported by Basaria and collaborators, PFS patients have impaired sexual function as well as high depression scores (Basaria et al., 2016). In sixteen PFS male patients, we recently observed that 1) all patients showed erectile dysfunction, 2) four of them had altered peripheral neurogenic control of erection and 3) eight of them suffered from a DSM-IV major depressive disorder (Melcangi et al., 2017).

In addition, in plasma and CSF of PFS patients, suspension of drug treatment did not only lead, as expected, to an alteration of 5α-reduced metabolites of testosterone and progesterone, but also to a global alteration of neuroactive steroid levels (Caruso et al., 2015; Melcangi et al., 2013, 2017). Interestingly, observations obtained in male rats indicate that finasteride treatment and its suspension not only alters the levels of neuroactive steroids in plasma and CSF, but also in the brain tissue. However, it is important to note that the effects in plasma or CSF are different in comparison to what observed in brain areas. For instance, subchronic treatment with finasteride (i.e., for 20 days) induced only an increase of progesterone in the hippocampus, while in plasma only an increase in isopregnanolone (i.e., a metabolite of DHP) associated with a decrease in DHT were observed (Giatti et al., 2016). At the withdrawal (i.e., after one month of suspension) a decrease of pregnenolone (i.e., the first neuroactive steroid synthesize from cholesterol) and progesterone associated with an increase of DHP were observed in the hippocampus, while a decrease in progesterone, tetrahydroprogesterone, THP (i.e., a metabolite of DHP), DHT and 3α-diol (i.e., a metabolite of DHT) was observed in plasma (Giatti et al., 2016). Interestingly, neither subchronic treatment with finasteride nor its withdrawal were able to affect the levels of DHT in the hippocampus (Giatti et al., 2016). In addition, the brain changes in neuroactive steroid levels were coupled with altered expression of their receptors, suggesting an altered brain steroid signaling (Giatti et al., 2016).

Depression is a multifaceted psychiatric condition, where genetic susceptibility and environmental factors crosstalk to generate a distressing condition. As reported in humans and in animal models that mimic this condition, depression is characterized by decreased content of neurotransmitters and neurotrophic factors, increased inflammation and gliosis, decreased neurogenesis and neuron survival and mitochondrial impairment (Blier, 2013; Masi and Brovedani, 2011).

Gut microbiota is an exclusive combination of different organisms, such as bacteria, viruses, archaea, protozoa and fungi that interact in a bidirectional way with the central nervous system forming the so call microbiome-gut-brain axis (Mayer, 2011; Sharon et al., 2016). Immune, neural, endocrine and metabolic pathways are included in this axis. Among them, steroid hormones have also an important role. Indeed, several observations indicate that steroid hormones influence gut microbiota niches (Tetel et al., 2018). For instance, gonadectomy or steroid treatment affect the composition of gut microbiota in rodents (Org et al., 2016). Several recent observations seem also to suggest a link between gut microbiota and development and/or manifestation of neuropsychiatric disorders, such as depression and anxiety (Borgo et al., 2017; Foster and McVey Neufeld, 2013). Indeed, microbiota composition is altered both in animal models and in patients with this symptomatology (Hoban et al., 2016; Kelly et al., 2016).

On this basis, our main hypothesis is that treatment with finasteride and/or its withdrawal in male rat may induce depressive-like behavior related with cellular and/or molecular alterations in hippocampus and/or gut microbiota composition. Therefore, in the present study we will explore: (1), whether subchronic treatment with this inhibitor of the enzyme 5α-R induces depressive-like behavior in male rats; (2), whether this alteration persists by 1 month after the end of the treatment and (3), whether finasteride treatment and/or withdrawal is associated with modifications in neurogenesis, gliosis and neuroinflammatory parameters in the hippocampus and with changes in gut microbiota.

Section snippets

Animals and treatments

Male Sprague Dawley rats (250–275 g at arrival, Janvier Labs, Le Genest- Saint-Isle, France) were housed in the animal care facility of the Instituto Cajal, C.S.I.C., Madrid, Spain. All animals were kept in standard rat cages with food and tap water available ad libitum and under controlled humidity and temperature. The rats were acclimated to the new environment for 7 days before being randomly assigned to one of the experimental groups described below. All procedures for handling and killing

Forced swim test

Two-way ANOVA revealed a significant effect of treatment (F = 4.22, p =  0.0474) (Fig. 1). At the end of the treatment, no significant differences were detected in the immobility time in the Porsolt test between animals treated with vehicle or finasteride, neither in pre-test session (day 1, data not shown) or test session (day 2). However, as reported in Fig. 1, one month after withdrawal animals treated with finasteride showed a significantly higher immobility time in the Porsolt test

Discussion

The findings of the present study indicate that finasteride treatment has persistent effects on depressive-like behavior in young adult male rats. To assess depressive-like behavior we used the Porsolt forced swim test (Castagne et al., 2011). Immobility floating behavior in this test is decreased by antidepressant drugs and is considered a manifestation of helplessness (Castagne et al., 2011). In the forced swim test (as the tail suspension test, the foot shock or any other learned

Conclusion

In summary, our findings indicate that finasteride treatment causes several alterations in the hippocampus that are detected at the end of the treatment, such as increased proliferation in the subgranular zone of the dentate gyrus, increased number of astrocytes in the hilus and increased mRNA levels of TNF-α. In addition, other changes are detected by one month after finasteride withdrawal, including decreased proliferation in the subgranular zone, increased astrogliosis in the hilus and a

Contributors

Silvia Diviccaro performed the treatment of animals, behavioral analysis, immunohistochemistry, morphometric analysis, granule cell density analysis.

Silvia Giatti performed assessment of cytokines by Real time PCR

Francesca Borgo, Elisa Borghi and Matteo Barcella responsible for gut microbiota analysis

Silvia Diviccaro, Francesca Borgo, José Luis Trejo, Luis Miguel Garcia-Segura and Roberto Cosimo Melcangi were responsible for study design and wrote the manuscript.

Financial support

This research was supported by grants from MIUR Progetto Eccellenza. We also thank the Post-Finasteride Foundation for the financial support to R.C. Melcangi; Ministerio de Economía, Industria y Competitividad (MINECO), Spain (grant number BFU2017-82754-R), Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain and Fondos Feder for the financial support to L.M. Garcia-Segura.

Conflicts of Interest

None.

References (59)

  • J.R. Kelly et al.

    Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat

    J. Psychiatr. Res.

    (2016)
  • R.C. Melcangi et al.

    Neuroactive Steroid Levels are Modified in Cerebrospinal Fluid and Plasma of Post-Finasteride Patients Showing Persistent Sexual Side Effects and Anxious/Depressive Symptomatology

    J. Sex Med.

    (2013)
  • R.C. Melcangi et al.

    Levels and actions of neuroactive steroids in the nervous system under physiological and pathological conditions: sex-specific features

    Neurosci. Biobehav. Rev.

    (2016)
  • R.C. Melcangi et al.

    Neuroactive steroid levels and psychiatric and andrological features in post-finasteride patients

    J. Steroid Biochem. Mol. Biol.

    (2017)
  • L. Mohle et al.

    Ly6C(hi) Monocytes Provide a Link between Antibiotic-Induced Changes in Gut Microbiota and Adult Hippocampal Neurogenesis

    Cell Rep.

    (2016)
  • R.J. Post et al.

    Depression: the search for separable behaviors and circuits

    Curr. Opin. Neurobiol.

    (2018)
  • K. Rea et al.

    The microbiome: A key regulator of stress and neuroinflammation

    Neurobiol. Stress

    (2016)
  • J.M. Ridlon et al.

    Clostridium scindens: a human gut microbe with a high potential to convert glucocorticoids into androgens

    J. Lipid Res.

    (2013)
  • C. Schule et al.

    The role of allopregnanolone in depression and anxiety

    Prog. Neurobiol.

    (2014)
  • G. Sharon et al.

    The central nervous system and the gut microbiome

    Cell

    (2016)
  • C.A. Stockmeier et al.

    Cellular changes in the postmortem hippocampus in major depression

    Biol. Psychiatry

    (2004)
  • Y.L. Wang et al.

    Microglial activation mediates chronic mild stress-induced depressive- and anxiety-like behavior in adult rats

    J. Neuroinflammation

    (2018)
  • R. Yirmiya et al.

    Depression as a microglial disease

    Trends Neurosci.

    (2015)
  • M. Yu et al.

    Variations in gut microbiota and fecal metabolic phenotype associated with depression by 16S rRNA gene sequencing and LC/MS-based metabolomics

    J. Pharm. Biomed. Anal.

    (2017)
  • G. Altomare et al.

    Depression circumstantially related to the administration of finasteride for androgenetic alopecia

    J. Dermatol.

    (2002)
  • I.S. Baek et al.

    Chronic antidepressant treatment in normal mice induces anxiety and impairs stress-coping ability

    Exp. Neurobiol.

    (2015)
  • E. Barrett et al.

    gamma-Aminobutyric acid production by culturable bacteria from the human intestine

    J. Appl. Microbiol.

    (2012)
  • S. Basaria et al.

    Characteristics of men who report persistent sexual symptoms after finasteride use for hair loss

    J. Clin. Endocrinol. Metab.

    (2016)
  • P. Blier

    Neurotransmitter targeting in the treatment of depression

    J Clin Psychiatry

    (2013)
  • Cited by (0)

    View full text