1
|
Scheggi S, Concas L, Corsi S, Carta M, Melis M, Frau R. Expanding the therapeutic potential of neuro(active)steroids: a promising strategy for hyperdopaminergic behavioral phenotypes. Neurosci Biobehav Rev 2024; 164:105842. [PMID: 39103066 DOI: 10.1016/j.neubiorev.2024.105842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/10/2024] [Accepted: 08/01/2024] [Indexed: 08/07/2024]
Abstract
Imbalances in dopamine activity significantly contribute to the pathophysiology of several neuropsychiatric disorders, including addiction, ADHD, schizophrenia, impulse control disorders, and Parkinson's Disease. Neuro(active)steroids, comprising endogenous steroids that finely modulate neuronal activity, are considered crucial regulators of brain function and behavior, with implications in various physiological processes and pathological conditions. Specifically, subclasses of Neuro(active)steroids belonging to the 5α reductase pathway are prominently involved in brain disorders characterized by dopaminergic signaling imbalances. This review highlights the neuromodulatory effects of Neuro(active)steroids on the dopamine system and related aberrant behavioral phenotypes. We critically appraise the role of pregnenolone, progesterone, and allopregnanolone on dopamine signaling. Additionally, we discuss the impact of pharmacological interventions targeting 5α reductase activity in neuropsychiatric conditions characterized by excessive activation of the dopaminergic system, ranging from psychotic (endo)phenotypes and motor complications to decision-making problems and addiction.
Collapse
Affiliation(s)
- Simona Scheggi
- Dept. of Molecular and Developmental Medicine, University of Siena, Italy
| | - Luca Concas
- Dept. Of Biomedical Sciences, University of Cagliari, Italy
| | - Sara Corsi
- Dept. of Developmental and Regenerative Neurobiology, Lund University, Sweden
| | - Manolo Carta
- Dept. Of Biomedical Sciences, University of Cagliari, Italy
| | - Miriam Melis
- Dept. Of Biomedical Sciences, University of Cagliari, Italy
| | - Roberto Frau
- Dept. Of Biomedical Sciences, University of Cagliari, Italy; Guy Everett Laboratory, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
2
|
Branca C, Bortolato M. The role of neuroactive steroids in tic disorders. Neurosci Biobehav Rev 2024; 160:105637. [PMID: 38519023 PMCID: PMC11121756 DOI: 10.1016/j.neubiorev.2024.105637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/03/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Tics are sudden, repetitive movements or vocalizations. Tic disorders, such as Tourette syndrome (TS), are contributed by the interplay of genetic risk factors and environmental variables, leading to abnormalities in the functioning of the cortico-striatal-thalamo-cortical (CSTC) circuitry. Various neurotransmitter systems, such as gamma-aminobutyric acid (GABA) and dopamine, are implicated in the pathophysiology of these disorders. Building on the evidence that tic disorders are predominant in males and exacerbated by stress, emerging research is focusing on the involvement of neuroactive steroids, including dehydroepiandrosterone sulfate (DHEAS) and allopregnanolone, in the ontogeny of tics and other phenotypes associated with TS. Emerging evidence indicates that DHEAS levels are significantly elevated in the plasma of TS-affected boys, and the clinical onset of this disorder coincides with the period of adrenarche, the developmental stage characterized by a surge in DHEAS synthesis. On the other hand, allopregnanolone has garnered particular attention for its potential to mediate the adverse effects of acute stress on the exacerbation of tic severity and frequency. Notably, both neurosteroids act as key modulators of GABA-A receptors, suggesting a pivotal role of these targets in the pathophysiology of various clinical manifestations of tic disorders. This review explores the potential mechanisms by which these and other neuroactive steroids may influence tic disorders and discusses the emerging therapeutic strategies that target neuroactive steroids for the management of tic disorders.
Collapse
Affiliation(s)
- Caterina Branca
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
3
|
Cadeddu R, Van Zandt M, Santovito LS, Odeh K, Anderson CJ, Flanagan D, Nordkild P, Pinna G, Pittenger C, Bortolato M. Prefrontal allopregnanolone mediates the adverse effects of acute stress in a mouse model of tic pathophysiology. Neuropsychopharmacology 2023; 48:1288-1299. [PMID: 37198434 PMCID: PMC10354086 DOI: 10.1038/s41386-023-01603-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 04/24/2023] [Accepted: 05/05/2023] [Indexed: 05/19/2023]
Abstract
Ample evidence suggests that acute stress can worsen symptom severity in Tourette syndrome (TS); however, the neurobiological underpinnings of this phenomenon remain poorly understood. We previously showed that acute stress exacerbates tic-like and other TS-associated responses via the neurosteroid allopregnanolone (AP) in an animal model of repetitive behavioral pathology. To verify the relevance of this mechanism to tic pathophysiology, here we tested the effects of AP in a mouse model recapitulating the partial depletion of dorsolateral cholinergic interneurons (CINs) seen in post-mortem studies of TS. Mice underwent targeted depletion of striatal CINs during adolescence and were tested in young adulthood. Compared with controls, partially CIN-depleted male mice exhibited several TS-relevant abnormalities, including deficient prepulse inhibition (PPI) and increased grooming stereotypies after a 30-min session of spatial confinement - a mild acute stressor that increases AP levels in the prefrontal cortex (PFC). These effects were not seen in females. Systemic and intra-PFC AP administration dose-dependently worsened grooming stereotypies and PPI deficits in partially CIN-depleted males. Conversely, both AP synthesis inhibition and pharmacological antagonism reduced the effects of stress. These results further suggest that AP in the PFC mediates the adverse effects of stress on the severity of tics and other TS-related manifestations. Future studies will be necessary to confirm these mechanisms in patients and define the circuitry responsible for the effects of AP on tics.
Collapse
Affiliation(s)
- Roberto Cadeddu
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Meghan Van Zandt
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
| | - Luca Spiro Santovito
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Karen Odeh
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Collin J Anderson
- Department of Neurology, School of Medicine, University of Utah, Salt Lake City, UT, USA
- School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
- School of Biomedical Engineering, University of Sydney, Camperdown, NSW, Australia
| | - Deirdre Flanagan
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
| | | | - Graziano Pinna
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- UI Center on Depression and Resilience (UICDR), Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Center for Alcohol Research in Epigenetics (CARE), Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Christopher Pittenger
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA.
- Department of Psychology, School of Arts and Sciences, Yale University, New Haven, CT, USA.
- Child Study Center, School of Medicine, Yale University, New Haven, CT, USA.
- Center for Brain and Mind Health, School of Medicine, Yale University, New Haven, CT, USA.
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
4
|
Frau R, Traccis F, Concas L, Cadeddu R, Mosher LJ, Nordkild P, Gaikwad NW, Bortolato M. Prefrontal allopregnanolone synergizes with D 1 receptor activation to disrupt sensorimotor gating in male Sprague-Dawley rats. Psychopharmacology (Berl) 2023; 240:1359-1372. [PMID: 37129616 DOI: 10.1007/s00213-023-06375-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/26/2023] [Indexed: 05/03/2023]
Abstract
RATIONALE The prepulse inhibition (PPI) of the startle reflex is the best-established index of sensorimotor gating. We documented that the neurosteroid allopregnanolone (AP) is necessary to reduce PPI in response to D1 dopamine receptor agonists. Since Sprague-Dawley (SD) rats are poorly sensitive to the PPI-disrupting effects of these drugs, we hypothesized that AP might increase this susceptibility. OBJECTIVES We tested whether AP is sufficient to increase the vulnerability of SD rats to PPI deficits in response to the D1 receptor full agonist SKF82958. METHODS SD rats were tested for PPI after treatment with SKF82958 (0.05-0.3 mg/kg, SC) in combination with either intraperitoneal (1-10 mg/kg) or intracerebral (0.5 μg/μl/side) AP administration into the medial prefrontal cortex (mPFC) or nucleus accumbens shell. To rule out potential confounds, we measured whether SKF82958 affected the endogenous mPFC levels of AP. RESULTS SD rats exhibited marked PPI deficits in response to the combination of systemic and intra-mPFC AP with SKF82958 but not with the D2 receptor agonist quinpirole (0.3-0.6 mg/kg, SC). SKF82958 did not elevate mPFC levels of AP but enhanced the content of its precursor progesterone. The PPI deficits caused by SKF82958 in combination with AP were opposed by the AP antagonist isoallopregnanolone (10 mg/kg, IP) and the glutamate NMDA receptor positive modulator CIQ (5 mg/kg, IP). CONCLUSION These results suggest that AP enables the detrimental effects of D1 receptor activation on sensorimotor gating. AP antagonism or glutamatergic modulation counters these effects and may have therapeutic potential for neuropsychiatric disorders characterized by gating deficits.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy.
- Guy Everett Laboratory, University of Cagliari, Cagliari, Italy.
| | - Francesco Traccis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Luca Concas
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Roberto Cadeddu
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | - Laura J Mosher
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | | | | | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
5
|
Corsi S, Scheggi S, Pardu A, Braccagni G, Caruso D, Cioffi L, Diviccaro S, Gentile M, Fanni S, Stancampiano R, Gambarana C, Melcangi RC, Frau R, Carta M. Pregnenolone for the treatment of L-DOPA-induced dyskinesia in Parkinson's disease. Exp Neurol 2023; 363:114370. [PMID: 36878398 DOI: 10.1016/j.expneurol.2023.114370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/02/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023]
Abstract
Growing preclinical and clinical evidence highlights neurosteroid pathway imbalances in Parkinson's Disease (PD) and L-DOPA-induced dyskinesias (LIDs). We recently reported that 5α-reductase (5AR) inhibitors dampen dyskinesias in parkinsonian rats; however, unraveling which specific neurosteroid mediates this effect is critical to optimize a targeted therapy. Among the 5AR-related neurosteroids, striatal pregnenolone has been shown to be increased in response to 5AR blockade and decreased after 6-OHDA lesions in the rat PD model. Moreover, this neurosteroid rescued psychotic-like phenotypes by exerting marked antidopaminergic activity. In light of this evidence, we investigated whether pregnenolone might dampen the appearance of LIDs in parkinsonian drug-naïve rats. We tested 3 escalating doses of pregnenolone (6, 18, 36 mg/kg) in 6-OHDA-lesioned male rats and compared the behavioral, neurochemical, and molecular outcomes with those induced by the 5AR inhibitor dutasteride, as positive control. The results showed that pregnenolone dose-dependently countered LIDs without affecting L-DOPA-induced motor improvements. Post-mortem analyses revealed that pregnenolone significantly prevented the increase of validated striatal markers of dyskinesias, such as phospho-Thr-34 DARPP-32 and phospho-ERK1/2, as well as D1-D3 receptor co-immunoprecipitation in a fashion similar to dutasteride. Moreover, the antidyskinetic effect of pregnenolone was paralleled by reduced striatal levels of BDNF, a well-established factor associated with the development of LIDs. In support of a direct pregnenolone effect, LC/MS-MS analyses revealed that striatal pregnenolone levels strikingly increased after the exogenous administration, with no significant alterations in downstream metabolites. All these data suggest pregnenolone as a key player in the antidyskinetic properties of 5AR inhibitors and highlight this neurosteroid as an interesting novel tool to target LIDs in PD.
Collapse
Affiliation(s)
- Sara Corsi
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy
| | - Simona Scheggi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, SI, Italy
| | - Alessandra Pardu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy
| | - Giulia Braccagni
- Department of Molecular and Developmental Medicine, University of Siena, Siena, SI, Italy
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Lucia Cioffi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Silvia Diviccaro
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Mauro Gentile
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy
| | - Silvia Fanni
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy; Basal Ganglia Pathophysiology Unit, Department Experimental Medical Science, Lund University, Sweden
| | | | - Carla Gambarana
- Department of Molecular and Developmental Medicine, University of Siena, Siena, SI, Italy
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy; "Guy Everett Laboratory", Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.
| | - Manolo Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy.
| |
Collapse
|
6
|
Acute stress impairs sensorimotor gating via the neurosteroid allopregnanolone in the prefrontal cortex. Neurobiol Stress 2022; 21:100489. [DOI: 10.1016/j.ynstr.2022.100489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/07/2022] [Accepted: 09/20/2022] [Indexed: 11/21/2022] Open
|
7
|
Tics: neurological disorders determined by a deficit in sensorimotor gating processes. Neurol Sci 2022; 43:5839-5850. [PMID: 35781754 PMCID: PMC9474467 DOI: 10.1007/s10072-022-06235-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/20/2022] [Indexed: 11/21/2022]
Abstract
Tic related disorders affect 4–20% of the population, mostly idiopathic, can be grouped in a wide spectrum of severity, where the most severe end is Tourette Syndrome (TS). Tics are arrhythmic hyperkinesias to whom execution the subject is forced by a “premonitory urge” that can be classified as sensory tic, just-right experience or urge without obsession. If an intact volitional inhibition allows patients to temporarily suppress tics, a lack or deficit in automatic inhibition is involved in the genesis of the disorder. Studies have assessed the presence of intrinsic microscopic and macroscopic anomalies in striatal circuits and relative cortical areas in association with a hyperdopaminergic state in the basal forebrain. Prepulse inhibition (PPI) of the startle reflex is a measure of inhibitory functions by which a weak sensory stimulus inhibits the elicitation of a startle response determined by a sudden intense stimulus. It is considered an operation measure of sensorimotor gating, a neural process by which unnecessary stimuli are eliminated from awareness. Evidence points out that the limbic domain of the CSTC loops, dopamine and GABA receptors within the striatum play an important role in PPI modulation. It is conceivable that a sensorimotor gating deficit may be involved in the genesis of premonitory urge and symptoms. Therefore, correcting the sensorimotor gating deficit may be considered a target for tic-related disorders therapies; in such case PPI (as well as other indirect estimators of sensorimotor gating) could represent therapeutic impact predictors.
Collapse
|
8
|
Santoni M, Frau R, Pistis M. Transgenerational Sex-dependent Disruption of Dopamine Function Induced by Maternal Immune Activation. Front Pharmacol 2022; 13:821498. [PMID: 35211019 PMCID: PMC8861303 DOI: 10.3389/fphar.2022.821498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/17/2022] [Indexed: 11/28/2022] Open
Abstract
Several epidemiological studies suggest an association between maternal infections during pregnancy and the emergence of neurodevelopmental disorders in the offspring, such as autism and schizophrenia. Animal models broadened the knowledge about the pathophysiological mechanisms that develop from prenatal infection to the onset of psychopathological phenotype. Mounting evidence supports the hypothesis that detrimental effects of maternal immune activation might be transmitted across generations. Here, we explored the transgenerational effects on the dopamine system of a maternal immune activation model based on the viral mimetic polyriboinosinic-polyribocytidilic acid. We assessed dopamine neurons activity in the ventral tegmental area by in vivo electrophysiology. Furthermore, we studied two behavioral tests strictly modulated by the mesolimbic dopamine system, i.e., the open field in response to amphetamine and the prepulse inhibition of the startle reflex in response to the D2 agonist apomorphine. Second-generation adult male rats did not display any deficit in sensorimotor gating; however, they displayed an altered activity of ventral tegmental area dopamine neurons, indexed by a reduced spontaneous firing rate and a heightened motor activation in response to amphetamine administration in the open field. On the other hand, second-generation female rats were protected from ancestors' polyriboinosinic-polyribocytidilic acid treatment, as they did not show any alteration in dopamine cell activity or in behavioral tests. These results confirm that maternal immune activation negatively influences, in a sex-dependent manner, neurodevelopmental trajectories of the dopamine system across generations.
Collapse
Affiliation(s)
- Michele Santoni
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
- “Guy Everett” Laboratory, University of Cagliari, Cagliari, Italy
| | - Marco Pistis
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy
- Unit of Clinical Pharmacology, University Hospital, Cagliari, Italy
| |
Collapse
|
9
|
Floris G, Scheggi S, Pes R, Bortolato M. The steroidogenic inhibitor finasteride reverses pramipexole-induced alterations in probability discounting. Brain Res Bull 2022; 181:157-166. [PMID: 35122898 PMCID: PMC9012661 DOI: 10.1016/j.brainresbull.2022.01.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/21/2022] [Accepted: 01/29/2022] [Indexed: 12/28/2022]
Abstract
Pramipexole is a potent agonist of D3 and D2 dopamine receptors, currently approved for clinical use in Parkinson's disease (PD) and restless leg syndrome. Several studies have shown that pramipexole significantly increases the risk of pathological gambling and impulse-control disorders. While these iatrogenic complications can impose a severe social and financial burden, their treatment poses serious clinical challenges. Our group previously reported that the steroidogenic inhibitor finasteride reduced pathological gambling severity in PD patients who developed this complication following pramipexole treatment. To study the mechanisms underlying these effects, here we tested the impact of finasteride in a rat model of pramipexole-induced alterations of probability discounting. We previously showed that, in rats exposed to low doses of the monoamine-depleting agent reserpine (1mg/kg/day, SC), pramipexole (0.3mg/kg/day, SC) increased the propensity to engage in disadvantageous choices. This effect was paralleled by a marked D3 receptor upregulation in the nucleus accumbens. First, we tested how finasteride (25-50mg/kg, IP) intrinsically affects probability discounting. While the highest dose of finasteride produced a marked lack of interest in lever pressing (manifested as a significant increase in omissions), the 25mg/kg (IP) dose did not intrinsically modify probability discounting. However, this finasteride regimen significantly reduced the adverse effects of reserpine and pramipexole in probability discounting by diminishing rats' propensity to engage in highly disadvantageous probabilistic choices. The same regimen also reversed the upregulation of D3 receptors in the nucleus accumbens induced by reserpine and pramipexole. These findings confirm that finasteride opposes the impulsivity caused by pramipexole and suggest that this effect may be underpinned by a normalizing effect on D3 receptor expression in the nucleus accumbens.
Collapse
Affiliation(s)
- Gabriele Floris
- Dept. of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City (UT), USA.
| | - Simona Scheggi
- Dept. of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City (UT), USA; Dept. of Molecular and Developmental Medicine, School of Medicine, University of Siena, ITALY
| | - Romina Pes
- Dept. of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence (KS), USA
| | - Marco Bortolato
- Dept. of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City (UT), USA; Dept. of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence (KS), USA.
| |
Collapse
|
10
|
Bortolato M, Coffey BJ, Gabbay V, Scheggi S. Allopregnanolone: The missing link to explain the effects of stress on tic exacerbation? J Neuroendocrinol 2022; 34:e13022. [PMID: 34423500 PMCID: PMC8800948 DOI: 10.1111/jne.13022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/19/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022]
Abstract
The neurosteroid allopregnanolone (3α-hydroxy-5α-pregnan-20-one; AP) elicits pleiotropic effects in the central nervous system, ranging from neuroprotective and anti-inflammatory functions to the regulation of mood and emotional responses. Several lines of research show that the brain rapidly produces AP in response to acute stress to reduce the allostatic load and enhance coping. These effects not only are likely mediated by GABAA receptor activation but also result from the contributions of other mechanisms, such as the stimulation of membrane progesterone receptors. In keeping with this evidence, AP has been shown to exert rapid, potent antidepressant properties and has been recently approved for the therapy of moderate-to-severe postpartum depression. In addition to depression, emerging evidence points to the potential of AP as a therapy for other neuropsychiatric disorders, including anxiety, seizures, post-traumatic stress disorder and cognitive problems. Although this evidence has spurred interest in further therapeutic applications of AP, some investigations suggest that this neurosteroid may also be associated with adverse events in specific disorders. For example, our group has recently documented that AP increases tic-like manifestations in several animal models of tic disorders; furthermore, our results indicate that inhibiting AP synthesis and signalling reduces the exacerbation of tic severity associated with acute stress. Although the specific mechanisms of these effects remain partially elusive, our findings point to the possibility that the GABAergic activation by AP may also lead to disinhibitory effects, which could interfere with the ability of patients to suppress their tics. Future studies will be necessary to verify whether these mechanisms may apply to other externalising manifestations, such as impulse-control problems and manic symptoms.
Collapse
Affiliation(s)
- Marco Bortolato
- Department of Pharmacology and ToxicologyCollege of PharmacyUniversity of UtahSalt Lake CityUTUSA
- Research Consortium on NeuroEndocrine Causes of Tics (ReConNECT)
| | - Barbara J. Coffey
- Research Consortium on NeuroEndocrine Causes of Tics (ReConNECT)
- Department of Psychiatry and Behavioral ScienceMiller School of MedicineUniversity of MiamiMiamiFLUSA
| | - Vilma Gabbay
- Research Consortium on NeuroEndocrine Causes of Tics (ReConNECT)
- Department of Psychiatry and Behavioral SciencesAlbert Einstein College of MedicineBronxNYUSA
| | - Simona Scheggi
- Department of Molecular and Developmental MedicineSchool of MedicineUniversity of SienaSienaItaly
| |
Collapse
|
11
|
Saengmearnuparp T, Lojanapiwat B, Chattipakorn N, Chattipakorn S. The connection of 5-alpha reductase inhibitors to the development of depression. Biomed Pharmacother 2021; 143:112100. [PMID: 34479019 DOI: 10.1016/j.biopha.2021.112100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022] Open
Abstract
Recent literature connects 5-alpha reductase inhibitors (5-ARIs) with neuropsychiatric adverse effects. Several clinical studies have indicated that former 5-ARIs users had a higher incidence of depressive symptoms and neuropsychiatric side effects than non-users. However, the underlying mechanisms involved in the depression in former 5-ARIs patients, a condition known as "post finasteride syndrome (PFS)", are not thoroughly understood. This review aims to summarize and discuss the association between 5-ARIs and depression as well as possible mechanisms. We used PubMed search terms including "depression", "depressive symptoms", "MDD", "anxiety", or "suicidal idea", and "5-alpha reductase inhibitors", "finasteride", "dutasteride", "5-ARIs". All relevant articles from in vivo and clinical studies from 2002 to 2021 were carefully reviewed. Any contradictory findings were included and debated. The potential mechanisms that link 5-ARIs and depression include alteration in neuroactive steroids, dopaminergic dysfunction, reduced hippocampal neurogenesis, increased neuroinflammation, alteration of the HPA axis, and epigenetic modifications. From this review, we hope to provide information for future studies based on animal experiments, and potential therapeutic strategies for depressive patients with PFS.
Collapse
Affiliation(s)
| | - Bannakij Lojanapiwat
- Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
12
|
The α6 GABA A Receptor Positive Allosteric Modulator DK-I-56-1 Reduces Tic-Related Behaviors in Mouse Models of Tourette Syndrome. Biomolecules 2021; 11:biom11020175. [PMID: 33525455 PMCID: PMC7912006 DOI: 10.3390/biom11020175] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 12/22/2022] Open
Abstract
Tourette syndrome (TS) is a disabling neurodevelopmental disorder characterized by multiple, recurrent tics. The pharmacological treatment of TS is currently based on dopaminergic antagonists; however, these drugs are associated with extrapyramidal symptoms and other serious adverse events. Recent evidence suggests that positive allosteric modulators (PAMs) of GABAA receptors containing α6 subunits (α6 GABAARs) oppose the behavioral effects of dopamine. Building on this evidence, in the present study, we tested the efficacy of DK-I-56-1, a highly selective PAM for α6 GABAARs, in mouse models of TS exhibiting tic-related responses. DK-I-56-1 significantly reduced tic-like jerks and prepulse inhibition (PPI) deficits in D1CT-7 transgenic mice, a well-documented mouse model of TS. DK-I-56-1 also prevented the exacerbation of spontaneous eyeblink reflex induced by the potent dopamine D1 receptor agonist SKF 82958, a proxy for tic-like responses. We also showed that both systemic and prefrontal cortical administration of DK-I-56-1 countered the PPI disruption caused by SKF 82958. Although the effects of DK-I-56-1 were akin to those elicited by dopaminergic antagonists, this drug did not elicit extrapyramidal effects, as measured by catalepsy. These results point to α6 GABAAR PAMs as promising TS therapies with a better safety profile than dopaminergic antagonists.
Collapse
|
13
|
Cadeddu R, Bäckström T, Floris G, Nordkild P, Segerdahl M, Bortolato M. Isoallopregnanolone reduces tic-like behaviours in the D1CT-7 mouse model of Tourette syndrome. J Neuroendocrinol 2020; 32:e12754. [PMID: 31175669 DOI: 10.1111/jne.12754] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/16/2019] [Accepted: 06/04/2019] [Indexed: 12/29/2022]
Abstract
Tourette syndrome (TS) is a neuropsychiatric disorder characterised by multiple, persistent tics. These semi-voluntary motor and phonic manifestations are typically aggravated by exposure to acute stress, yet the mechanisms underlying this exacerbation remain unclear. Using a well-characterised animal model of TS, the D1CT-7 mouse, we recently showed that acute stress increases tic-like responses and causes sensorimotor gating deficits, as measured by the prepulse inhibition of the startle. We showed that these effects are promoted by the brain synthesis of the neurosteroid allopregnanolone (AP). In line with this idea, inhibition of AP synthesis by finasteride was found to suppress the tic-exacerbating effects of stress; conversely, AP administration resulted in a marked enhancement of the number of tic-like motor bursts. Given that the primary mechanism of AP is based on the positive allosteric modulation of GABAA receptors, in the present study, we hypothesised that the enhancement in tic-like behaviours induced by either stress or AP may be countered by isoallopregnanolone (isoAP), the natural 3β-epimer of AP that acts as an antagonist to the AP-binding site within GABAA receptors. In agreement with our hypothesis, isoAP (5-10 mg kg-1 , s.c.) dose-dependently reduced the number of tic-like behaviours induced by stress in D1CT-7 mice. These effects were comparable to those elicited by both the benchmark TS therapy haloperidol (0.3 mg kg-1 , i.p.), as well as finasteride (25 mg kg-1 , i.p.). IsoAP also countered the prepulse inhibition deficits secondary to stress in D1CT-7 mice. Finally, isoAP opposed the enhancement of tic-like behaviours induced by AP (15 mg kg-1 , i.p.). Given that isoAP is well-tolerated and has an optimal safety profile, these data suggest that this steroid may have therapeutic properties in TS.
Collapse
Affiliation(s)
- Roberto Cadeddu
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Torbjörn Bäckström
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, Umeå, Sweden
- Asarina Pharma, Copenhagen, Denmark
| | - Gabriele Floris
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | | | | | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
14
|
Frau R, Traccis F, Bortolato M. Neurobehavioural complications of sleep deprivation: Shedding light on the emerging role of neuroactive steroids. J Neuroendocrinol 2020; 32:e12792. [PMID: 31505075 PMCID: PMC6982588 DOI: 10.1111/jne.12792] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/06/2019] [Accepted: 09/05/2019] [Indexed: 01/05/2023]
Abstract
Sleep deprivation (SD) is associated with a broad spectrum of cognitive and behavioural complications, including emotional lability and enhanced stress reactivity, as well as deficits in executive functions, decision making and impulse control. These impairments, which have profound negative consequences on the health and productivity of many individuals, reflect alterations of the prefrontal cortex (PFC) and its connectivity with subcortical regions. However, the molecular underpinnings of these alterations remain elusive. Our group and others have begun examining how the neurobehavioural outcomes of SD may be influenced by neuroactive steroids, a family of molecules deeply implicated in sleep regulation and the stress response. These studies have revealed that, similar to other stressors, acute SD leads to increased synthesis of the neurosteroid allopregnanolone in the PFC. Whereas this up-regulation is likely aimed at counterbalancing the detrimental impact of oxidative stress induced by SD, the increase in prefrontal allopregnanolone levels contributes to deficits in sensorimotor gating and impulse control, signalling a functional impairment of PFC. This scenario suggests that the synthesis of neuroactive steroids during acute SD may be enacted as a neuroprotective response in the PFC; however, such compensation may in turn set off neurobehavioural complications by interfering with the corticolimbic connections responsible for executive functions and emotional regulation.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato (CA), Italy
- National Institute of Neuroscience (INN), University of Cagliari, Monserrato (CA), Italy
| | - Francesco Traccis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato (CA), Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City (UT), USA
| |
Collapse
|
15
|
Diviccaro S, Melcangi RC, Giatti S. Post-finasteride syndrome: An emerging clinical problem. Neurobiol Stress 2019; 12:100209. [PMID: 32435662 PMCID: PMC7231981 DOI: 10.1016/j.ynstr.2019.100209] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/11/2019] [Accepted: 12/23/2019] [Indexed: 12/14/2022] Open
Abstract
The presence of side effects during pharmacological treatment is unfortunately a quite common problem. In this review, we focused our attention on adverse events related to 5 alpha-reductase (5α-R) inhibitors (i.e., finasteride and dutasteride), approved for the treatment of benign prostatic hyperplasia and androgenetic alopecia (AGA). Although these drugs are generally well tolerated, many reports described adverse effects in men during treatment, such as sexual dysfunction and mood alteration. In addition, it has been also reported that persistent side effects may occur in some AGA patients. This condition, termed post-finasteride syndrome (PFS) is characterized by sexual side effects (i.e., low libido, erectile dysfunction, decreased arousal and difficulty in achieving orgasm), depression, anxiety and cognitive complaints that are still present despite drug withdrawal. Indeed, some national agencies (e.g., Swedish Medical Products Agency, the Medicines and Healthcare Products Regulatory Agency of UK and the U.S. Food and Drug Administration) required to include multiple persistent side effects within the finasteride labels. As here reported, these observations are mainly based on self-reporting of the symptomatology by the patients and few clinical studies have been performed so far. In addition, molecular mechanisms and/or genetic determinants behind such adverse effects have been poorly explored both in patients and animal models. Therefore, results here discussed indicate that PFS is an emerging clinical problem that needs to be further elucidated.
Collapse
Affiliation(s)
- Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milano, Italy
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milano, Italy
| | - Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milano, Italy
| |
Collapse
|
16
|
Godar SC, Cadeddu R, Floris G, Mosher LJ, Mi Z, Jarmolowicz DP, Scheggi S, Walf AA, Koonce CJ, Frye CA, Muma NA, Bortolato M. The Steroidogenesis Inhibitor Finasteride Reduces the Response to Both Stressful and Rewarding Stimuli. Biomolecules 2019; 9:biom9110749. [PMID: 31752360 PMCID: PMC6920809 DOI: 10.3390/biom9110749] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/14/2019] [Accepted: 11/18/2019] [Indexed: 01/15/2023] Open
Abstract
Finasteride (FIN) is the prototypical inhibitor of steroid 5α-reductase (5αR), the enzyme that catalyzes the rate-limiting step of the conversion of progesterone and testosterone into their main neuroactive metabolites. FIN is clinically approved for the treatment of benign prostatic hyperplasia and male baldness; while often well-tolerated, FIN has also been shown to cause or exacerbate psychological problems in vulnerable subjects. Evidence on the psychological effects of FIN, however, remains controversial, in view of inconsistent clinical reports. Here, we tested the effects of FIN in a battery of tests aimed at capturing complementary aspects of mood regulation and stress reactivity in rats. FIN reduced exploratory, incentive, prosocial, and risk-taking behavior; furthermore, it decreased stress coping, as revealed by increased immobility in the forced-swim test (FST). This last effect was also observed in female and orchiectomized male rats, suggesting that the mechanism of action of FIN does not primarily reflect changes in gonadal steroids. The effects of FIN on FST responses were associated with a dramatic decrease in corticotropin release hormone (CRH) mRNA and adrenocorticotropic hormone (ACTH) levels. These results suggest that FIN impairs stress reactivity and reduces behavioral activation and impulsive behavior by altering the function of the hypothalamus-pituitary-adrenal (HPA) axis.
Collapse
Affiliation(s)
- Sean C. Godar
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake, UT 84112, USA; (S.C.G.); (R.C.); (G.F.); (L.J.M.); (S.S.)
| | - Roberto Cadeddu
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake, UT 84112, USA; (S.C.G.); (R.C.); (G.F.); (L.J.M.); (S.S.)
| | - Gabriele Floris
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake, UT 84112, USA; (S.C.G.); (R.C.); (G.F.); (L.J.M.); (S.S.)
| | - Laura J. Mosher
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake, UT 84112, USA; (S.C.G.); (R.C.); (G.F.); (L.J.M.); (S.S.)
- Department of Pharmacology and Toxicology, School of Pharmacy; Lawrence, KS 66045, USA; (Z.M.); (N.A.M.)
| | - Zhen Mi
- Department of Pharmacology and Toxicology, School of Pharmacy; Lawrence, KS 66045, USA; (Z.M.); (N.A.M.)
| | - David P. Jarmolowicz
- Department of Applied Behavioral Science; University of Kansas, Lawrence, KS 66045, USA;
- Cofrin Logan Center for Addiction Research and Treatment; University of Kansas, Lawrence, KS 66045, USA
| | - Simona Scheggi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake, UT 84112, USA; (S.C.G.); (R.C.); (G.F.); (L.J.M.); (S.S.)
| | - Alicia A. Walf
- Department of Cognitive Science, Rensselaer Polytechnic Institute, Troy, NY 12180, USA;
- Department of Psychology; The University at Albany-SUNY, Albany, NY 12222, USA; (C.J.K.); (C.A.F.)
| | - Carolyn J. Koonce
- Department of Psychology; The University at Albany-SUNY, Albany, NY 12222, USA; (C.J.K.); (C.A.F.)
| | - Cheryl A. Frye
- Department of Psychology; The University at Albany-SUNY, Albany, NY 12222, USA; (C.J.K.); (C.A.F.)
- Department of Biological Sciences; The University at Albany-SUNY, Albany, NY 12222, USA
- Center for Neuroscience, The University at Albany-SUNY, Albany, NY 12222, USA
- Comprehensive Neuropsychological Services, Albany, NY 12203, USA
| | - Nancy A. Muma
- Department of Pharmacology and Toxicology, School of Pharmacy; Lawrence, KS 66045, USA; (Z.M.); (N.A.M.)
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake, UT 84112, USA; (S.C.G.); (R.C.); (G.F.); (L.J.M.); (S.S.)
- Correspondence:
| |
Collapse
|
17
|
Owens SJ, Purves-Tyson TD, Webster MJ, Shannon Weickert C. Evidence for enhanced androgen action in the prefrontal cortex of people with bipolar disorder but not schizophrenia or major depressive disorder. Psychiatry Res 2019; 280:112503. [PMID: 31446215 DOI: 10.1016/j.psychres.2019.112503] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 01/24/2023]
Abstract
Anxiety and depressive disorders are more prevalent in hypogonadal men. Low testosterone levels are associated with greater negative symptoms and impaired cognition in men with schizophrenia. Thus, androgens may contribute to brain pathophysiology in psychiatric disorders. We investigated androgen-related mRNAs in post-mortem dorsolateral prefrontal cortex of psychiatric disorders. We also assessed androgen receptor (AR) CAG trinucleotide repeat length, a functional AR gene variant associated with AR gene expression, receptor activity, and circulating testosterone. AR CAG repeat length was determined from genomic DNA and AR and 5α-reductase mRNAs measured using quantitative PCR in schizophrenia, bipolar disorder and control cases [n = 35/group; Stanley Medical Research Institute (SMRI) Array collection]. Layer-specific AR gene expression was determined using in situ hybridisation in schizophrenia, bipolar disorder, major depressive disorder and control cases (n = 15/group; SMRI Neuropathology Consortium). AR mRNA was increased in bipolar disorder, but was unchanged in schizophrenia, relative to controls. AR and 5α-reductase mRNAs were significantly positively correlated in bipolar disorder. AR CAG repeat length was significantly shorter in bipolar disorder relative to schizophrenia. AR mRNA expression was highest in cortical layers IV and V, but no layer-specific diagnostic differences were detected. Together, our results suggest enhanced cortical androgen action in people with bipolar disorder.
Collapse
Affiliation(s)
- Samantha J Owens
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick NSW 2031, Australia; School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney NSW 2052, Australia.
| | - Tertia D Purves-Tyson
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick NSW 2031, Australia; School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney NSW 2052, Australia
| | - Maree J Webster
- Laboratory of Brain Research, Stanley Medical Research Institute, MD 20815, USA
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick NSW 2031, Australia; School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney NSW 2052, Australia; Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY 13210, USA.
| |
Collapse
|
18
|
Mosher LJ, Cadeddu R, Yen S, Staudinger JL, Traccis F, Fowler SC, Maguire JL, Bortolato M. Allopregnanolone is required for prepulse inhibition deficits induced by D 1 dopamine receptor activation. Psychoneuroendocrinology 2019; 108:53-61. [PMID: 31228750 PMCID: PMC6773911 DOI: 10.1016/j.psyneuen.2019.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 06/08/2019] [Accepted: 06/12/2019] [Indexed: 10/26/2022]
Abstract
INTRODUCTION The extraction of salient information from the environment is modulated by the activation of dopamine receptors. Using rodent models, we previously reported that gating deficits caused by dopamine receptor activation - as measured by the prepulse inhibition (PPI) of startle - are effectively opposed by inhibitors of the steroidogenic enzyme 5α-reductase (5αR). The specific 5αR isoenzyme and steroids implicated in these effects, however, remain unknown. METHODS The effects of the selective D1 dopamine receptor agonist SKF-82958 (SKF, 0.3 mg/kg, IP) and D2 receptor agonist quinpirole (QUIN, 0.5 mg/kg, IP) were tested in the startle reflex and PPI of knockout (KO) mice for either 5αR type 1 (5αR1) or type 2 (5αR2). Furthermore, we established whether these effects may be modified by the 5α-reduced steroids dihydroprogesterone (DHP), allopregnanolone (AP), dihydrotestosterone (DHT), 5α-androstane-3α,17β-diol (3α-diol), or androsterone. To test the mechanisms whereby 5αR products may alter the PPI-disrupting properties of D1 agonists, we studied the involvement of GABA-A and PXR, two receptors targeted by neuroactive steroids. Specifically, we tested the effects of SKF in combination with the GABA-A antagonist bicuculline, as well as in KO mice for the GABA-A δ subunit and PXR. RESULTS 5αR1, but not 5αR2, knockout (KO) mice were insensitive to the PPI-disrupting effects of SKF. This sensitivity was reinstated by AP (3 mg/kg, IP), but not other 5α-reduced steroids. The PPI deficits induced by SKF were not modified by bicuculline, δ-subunit KO mice and PXR KO mice. CONCLUSIONS These results collectively suggest that 5αR1 enables the negative effects of D1 dopamine receptor activation on information processing via production of AP. The contribution of AP to the PPI-disrupting mechanisms of D1 receptor agonists, however, do not appear to be mediated by either GABA-A or PXR receptors.
Collapse
Affiliation(s)
- Laura J Mosher
- Dept. of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City (UT),Dept. of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence (KS)
| | - Roberto Cadeddu
- Dept. of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City (UT)
| | - Sabrina Yen
- Department of Neuroscience, School of Medicine, Tufts University, Boston (MA)
| | - Jeffrey L Staudinger
- Department of Basic Science, Kansas City University, School of Osteopathic Medicine, Joplin (MO)
| | - Francesco Traccis
- Department of Biomedical Sciences, University of Cagliari, Monserrato (CA), Italy
| | - Stephen C Fowler
- Dept. of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence (KS)
| | - Jamie L Maguire
- Department of Neuroscience, School of Medicine, Tufts University, Boston (MA)
| | - Marco Bortolato
- Dept. of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City UT United States.
| |
Collapse
|
19
|
Fanni S, Scheggi S, Rossi F, Tronci E, Traccis F, Stancampiano R, De Montis MG, Devoto P, Gambarana C, Bortolato M, Frau R, Carta M. 5alpha-reductase inhibitors dampen L-DOPA-induced dyskinesia via normalization of dopamine D1-receptor signaling pathway and D1-D3 receptor interaction. Neurobiol Dis 2018; 121:120-130. [PMID: 30261284 DOI: 10.1016/j.nbd.2018.09.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/06/2018] [Accepted: 09/18/2018] [Indexed: 12/20/2022] Open
Abstract
Although 1-3,4-dihydroxyphenylalanine (L-DOPA) is the mainstay therapy for treating Parkinson's disease (PD), its long-term administration is accompanied by the development of motor complications, particularly L-DOPA induced dyskinesia (LID), that dramatically affects patients' quality of life. LID has consistently been related to an excessive dopamine receptor transmission, particularly at the down-stream signaling of the striatal D1 receptors (D1R), resulting in an exaggerated stimulation of cAMP-dependent protein kinase and extracellular signal-regulated kinase (ERK) pathway. We previously reported that pharmacological blockade of 5alpha-reductase (5AR), the rate-limiting enzyme in neurosteroids synthesis, attenuates the severity of a broad set of behavioral alterations induced by D1R and D3R activation, without inducing extrapyramidal symptoms. In line with this evidence, in a recent study, we found that inhibition of 5AR by finasteride (FIN) produced a significant reduction of dyskinesia induced by L-DOPA and direct dopaminergic agonists in 6-OHDA-lesioned rats. In the attempt to further investigate the effect of 5AR inhibitors on dyskinesia and shed light on the mechanism of action, in the present study we compared the effect of FIN and dutasteride (DUTA), a potent dual 5AR inhibitor, on the development of LID, on the therapeutic efficacy of L-DOPA, on the molecular alterations downstream to the D1R, as well as on D1R-D3R interaction. The results indicated that both FIN and DUTA administration significantly reduced development and expression of LID; however, DUTA appeared more effective than FIN at a lower dose and produced its antidyskinetic effect without impacting the ability of L-DOPA to increase motor activation, or ameliorate forelimb use in parkinsonian rats. Moreover, this study demonstrates for the first time that 5AR inhibitors are able to prevent key events in the appearance of dyskinesia, such as L-DOPA-induced upregulation of striatal D1R-related cAMP/PKA/ERK signaling pathways and D1R-D3R coimmunoprecipitation, an index of heteromer formation. These findings are relevant as they confirm the 5AR enzyme as a potential therapeutic target for treatment of dyskinesia in PD, suggesting the first ever evidence that neurosteroidogenesis may affect functional interaction between dopamine D1R and D3R.
Collapse
Affiliation(s)
- Silvia Fanni
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Simona Scheggi
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 4, 53100 Siena, Italy
| | - Francesca Rossi
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Elisabetta Tronci
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Francesco Traccis
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Roberto Stancampiano
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Maria Graziella De Montis
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 4, 53100 Siena, Italy
| | - Paola Devoto
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Carla Gambarana
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 4, 53100 Siena, Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT 84112, USA
| | - Roberto Frau
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy; Tourette Syndrome Center, University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy; Sleep Medicine Center, University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy; National Institute of Neuroscience (INN), University of Cagliari, Monserrato, CA, Italy.
| | - Manolo Carta
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy.
| |
Collapse
|
20
|
Giatti S, Diviccaro S, Panzica G, Melcangi RC. Post-finasteride syndrome and post-SSRI sexual dysfunction: two sides of the same coin? Endocrine 2018; 61:180-193. [PMID: 29675596 DOI: 10.1007/s12020-018-1593-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/05/2018] [Indexed: 12/27/2022]
Abstract
Sexual dysfunction is a clinical condition due to different causes including the iatrogenic origin. For instance, it is well known that sexual dysfunction may occur in patients treated with antidepressants like selective serotonin reuptake inhibitors (SSRI). A similar side effect has been also reported during treatment with finasteride, an inhibitor of the enzyme 5alpha-reductase, for androgenetic alopecia. Interestingly, sexual dysfunction persists in both cases after drug discontinuation. These conditions have been named post-SSRI sexual dysfunction (PSSD) and post-finasteride syndrome (PFS). In particular, feeling of a lack of connection between the brain and penis, loss of libido and sex drive, difficulty in achieving an erection and genital paresthesia have been reported by patients of both conditions. It is interesting to note that the incidence of these diseases is probably so far underestimated and their etiopathogenesis is not sufficiently explored. To this aim, the present review will report the state of art of these two different pathologies and discuss, on the basis of the role exerted by three different neuromodulators such as dopamine, serotonin and neuroactive steroids, whether the persistent sexual dysfunction observed could be determined by common mechanisms.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Giancarlo Panzica
- Dipartimento di Neuroscienze "Rita Levi Montalcini", Università degli studi di Torino, Neuroscience Institute Cavallieri Ottolenghi (NICO), Orbassano, Italy
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
21
|
Traish AM. The Post-finasteride Syndrome: Clinical Manifestation of Drug-Induced Epigenetics Due to Endocrine Disruption. CURRENT SEXUAL HEALTH REPORTS 2018. [DOI: 10.1007/s11930-018-0161-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
22
|
Frau R, Bortolato M. Repurposing steroidogenesis inhibitors for the therapy of neuropsychiatric disorders: Promises and caveats. Neuropharmacology 2018; 147:55-65. [PMID: 29907425 DOI: 10.1016/j.neuropharm.2018.05.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/07/2018] [Accepted: 05/10/2018] [Indexed: 12/29/2022]
Abstract
Steroids exert a profound influence on behavioral reactivity, by modulating the functions of most neurotransmitters and shaping the impact of stress and sex-related variables on neural processes. This background - as well as the observation that most neuroactive steroids (including sex hormones, glucocorticoids and neurosteroids) are synthetized and metabolized by overlapping enzymatic machineries - points to steroidogenic pathways as a powerful source of targets for neuropsychiatric disorders. Inhibitors of steroidogenic enzymes have been developed and approved for a broad range of genitourinary and endocrine dysfunctions, opening to new opportunities to repurpose these drugs for the treatment of mental problems. In line with this idea, preliminary clinical and preclinical results from our group have shown that inhibitors of key steroidogenic enzymes, such as 5α-reductase and 17,20 desmolase-lyase, may have therapeutic efficacy in specific behavioral disorders associated with dopaminergic hyperfunction. While the lack of specificity of these effects raises potential concerns about endocrine adverse events, these initial findings suggest that steroidogenesis modulators with greater brain specificity may hold significant potential for the development of alternative therapies for psychiatric problems. This article is part of the Special Issue entitled 'Drug Repurposing: old molecules, new ways to fast track drug discovery and development for CNS disorders'.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato CA, Italy; Tourette Syndrome Center, University of Cagliari, Monserrato CA, Italy; Sleep Medicine Center, University of Cagliari, Monserrato CA, Italy; National Institute of Neuroscience (INN), University of Cagliari, Monserrato CA, Italy.
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
23
|
Mosher LJ, Godar SC, Morissette M, McFarlin KM, Scheggi S, Gambarana C, Fowler SC, Di Paolo T, Bortolato M. Steroid 5α-reductase 2 deficiency leads to reduced dominance-related and impulse-control behaviors. Psychoneuroendocrinology 2018; 91:95-104. [PMID: 29544191 PMCID: PMC5901899 DOI: 10.1016/j.psyneuen.2018.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/09/2018] [Accepted: 02/13/2018] [Indexed: 01/10/2023]
Abstract
The enzyme steroid 5α-reductase 2 (5αR2) catalyzes the conversion of testosterone into the potent androgen 5α-dihydrotestosterone. Previous investigations showed that 5αR2 is expressed in key brain areas for emotional and socio-affective reactivity, yet the role of this enzyme in behavioral regulation remains mostly unknown. Here, we profiled the behavioral characteristics of 5αR2 heterozygous (HZ) and knockout (KO) mice, as compared with their wild-type (WT) littermates. While male 5αR2 KO mice displayed no overt alterations in motoric, sensory, information-processing and anxiety-related behaviors, they exhibited deficits in neurobehavioral correlates of dominance (including aggression against intruders, mating, and tube dominance) as well as novelty-seeking and risk-taking responses. Furthermore, male 5αR2 KO mice exhibited reduced D2-like dopamine receptor binding in the shell of the nucleus accumbens - a well-recognized molecular signature of social dominance. Collectively, these results suggest that 5αR2 is involved in the establishment of social dominance and its behavioral manifestations. Further studies are warranted to understand how the metabolic actions of 5αR2 on steroid profile may be implicated in social ranking, impulse control, and the modulation of dopamine receptor expression in the nucleus accumbens.
Collapse
Affiliation(s)
- Laura J Mosher
- Dept of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, United States; Dept of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Sean C Godar
- Dept of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, United States
| | - Marc Morissette
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Quebec, Canada
| | - Kenneth M McFarlin
- Dept of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States.
| | - Simona Scheggi
- Dept of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, United States; Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Carla Gambarana
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Stephen C Fowler
- Dept of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Thérèse Di Paolo
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Quebec, Canada; Faculty of Pharmacy, Laval University, Quebec City, Quebec, Canada
| | - Marco Bortolato
- Dept of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
24
|
Tobiansky DJ, Wallin-Miller KG, Floresco SB, Wood RI, Soma KK. Androgen Regulation of the Mesocorticolimbic System and Executive Function. Front Endocrinol (Lausanne) 2018; 9:279. [PMID: 29922228 PMCID: PMC5996102 DOI: 10.3389/fendo.2018.00279] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/11/2018] [Indexed: 12/21/2022] Open
Abstract
Multiple lines of evidence indicate that androgens, such as testosterone, modulate the mesocorticolimbic system and executive function. This review integrates neuroanatomical, molecular biological, neurochemical, and behavioral studies to highlight how endogenous and exogenous androgens alter behaviors, such as behavioral flexibility, decision making, and risk taking. First, we briefly review the neuroanatomy of the mesocorticolimbic system, which mediates executive function, with a focus on the ventral tegmental area (VTA), nucleus accumbens (NAc), medial prefrontal cortex (mPFC), and orbitofrontal cortex (OFC). Second, we present evidence that androgen receptors (AR) and other steroid receptors are expressed in the mesocorticolimbic system. Using sensitive immunohistochemistry and quantitative polymerase chain reaction (qPCR) techniques, ARs are detected in the VTA, NAc, mPFC, and OFC. Third, we describe recent evidence for local androgens ("neuroandrogens") in the mesocorticolimbic system. Steroidogenic enzymes are expressed in mesocorticolimbic regions. Furthermore, following long-term gonadectomy, testosterone is nondetectable in the blood but detectable in the mesocorticolimbic system, using liquid chromatography tandem mass spectrometry. However, the physiological relevance of neuroandrogens remains unknown. Fourth, we review how anabolic-androgenic steroids (AAS) influence the mesocorticolimbic system. Fifth, we describe how androgens modulate the neurochemistry and structure of the mesocorticolimbic system, particularly with regard to dopaminergic signaling. Finally, we discuss evidence that androgens influence executive functions, including the effects of androgen deprivation therapy and AAS. Taken together, the evidence indicates that androgens are critical modulators of executive function. Similar to dopamine signaling, there might be optimal levels of androgen signaling within the mesocorticolimbic system for executive functioning. Future studies should examine the regulation and functions of neurosteroids in the mesocorticolimbic system, as well as the potential deleterious and enduring effects of AAS use.
Collapse
Affiliation(s)
- Daniel J. Tobiansky
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Daniel J. Tobiansky,
| | - Kathryn G. Wallin-Miller
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, United States
| | - Stan B. Floresco
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Ruth I. Wood
- Department of Integrative Anatomical Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Kiran K. Soma
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
25
|
Frau R, Bini V, Soggiu A, Scheggi S, Pardu A, Fanni S, Roncada P, Puligheddu M, Marrosu F, Caruso D, Devoto P, Bortolato M. The Neurosteroidogenic Enzyme 5α-Reductase Mediates Psychotic-Like Complications of Sleep Deprivation. Neuropsychopharmacology 2017; 42:2196-2205. [PMID: 28102229 PMCID: PMC5603808 DOI: 10.1038/npp.2017.13] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/18/2016] [Accepted: 01/05/2017] [Indexed: 01/02/2023]
Abstract
Acute sleep deprivation (SD) can trigger or exacerbate psychosis- and mania-related symptoms; the neurobiological basis of these complications, however, remains elusive. Given the extensive involvement of neuroactive steroids in psychopathology, we hypothesized that the behavioral complications of SD may be contributed by 5α-reductase (5αR), the rate-limiting enzyme in the conversion of progesterone into the neurosteroid allopregnanolone. We first tested whether rats exposed to SD may exhibit brain-regional alterations in 5αR isoenzymes and neuroactive steroid levels; then, we assessed whether the behavioral and neuroendocrine alterations induced by SD may be differentially modulated by the administration of the 5αR inhibitor finasteride, as well as progesterone and allopregnanolone. SD selectively enhanced 5αR expression and activity, as well as AP levels, in the prefrontal cortex; furthermore, finasteride (10-100 mg/kg, IP) dose-dependently ameliorated PPI deficits, hyperactivity, and risk-taking behaviors, in a fashion akin to the antipsychotic haloperidol and the mood stabilizer lithium carbonate. Finally, PPI deficits were exacerbated by allopregnanolone (10 mg/kg, IP) and attenuated by progesterone (30 mg/kg, IP) in SD-subjected, but not control rats. Collectively, these results provide the first-ever evidence that 5αR mediates a number of psychosis- and mania-like complications of SD through imbalances in cortical levels of neuroactive steroids.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences,
Division of Neuroscience and Clinical Pharmacology, Monserrato,
Italy,Tourette Syndrome Center,
Monserrato, Italy,Sleep Medicine Center; University of
Cagliari, Monserrato, Italy
| | - Valentina Bini
- Department of Biomedical Sciences,
Division of Neuroscience and Clinical Pharmacology, Monserrato,
Italy,Tourette Syndrome Center,
Monserrato, Italy
| | - Alessio Soggiu
- ‘L. Spallanzani’ Institute,
Proteomics Section—Department of Veterinary Sciences and Public Health,
University of Milan, Milan, Italy
| | - Simona Scheggi
- Department of Molecular and Developmental
Medicine, University of Siena, Siena, Italy,Department of Pharmacology and
Toxicology, College of Pharmacy, University of Utah, Salt Lake
City, UT, USA
| | - Alessandra Pardu
- Department of Biomedical Sciences,
Division of Neuroscience and Clinical Pharmacology, Monserrato,
Italy
| | - Silvia Fanni
- Department of Biomedical Sciences,
Division of Neuroscience and Clinical Pharmacology, Monserrato,
Italy
| | - Paola Roncada
- ‘L. Spallanzani’ Institute,
Proteomics Section—Department of Veterinary Sciences and Public Health,
University of Milan, Milan, Italy
| | - Monica Puligheddu
- Tourette Syndrome Center,
Monserrato, Italy,Sleep Medicine Center; University of
Cagliari, Monserrato, Italy,Department of Public Health, Section of
Neurology, Monserrato, Italy
| | - Francesco Marrosu
- Tourette Syndrome Center,
Monserrato, Italy,Sleep Medicine Center; University of
Cagliari, Monserrato, Italy,Department of Public Health, Section of
Neurology, Monserrato, Italy
| | - Donatella Caruso
- Department of Pharmacological and
Biomolecular Sciences, Center of Excellence for Neurodegenerative Diseases,
University of Milan, Milan, Italy
| | - Paola Devoto
- Department of Biomedical Sciences,
Division of Neuroscience and Clinical Pharmacology, Monserrato,
Italy,Tourette Syndrome Center,
Monserrato, Italy,Sleep Medicine Center; University of
Cagliari, Monserrato, Italy
| | - Marco Bortolato
- Department of Pharmacology and
Toxicology, College of Pharmacy, University of Utah, Salt Lake
City, UT, USA,Department of Pharmacology and Toxicology, College of
Pharmacy, University of Utah, Skaggs Hall, Room 3916, 30S 2000
E, Salt Lake City
84112, UT, USA, Tel: 801 587 3352, Fax:
801 585 5111, E-mail:
| |
Collapse
|
26
|
Allopregnanolone mediates the exacerbation of Tourette-like responses by acute stress in mouse models. Sci Rep 2017; 7:3348. [PMID: 28611376 PMCID: PMC5469807 DOI: 10.1038/s41598-017-03649-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/02/2017] [Indexed: 12/26/2022] Open
Abstract
Tourette syndrome (TS) is a neuropsychiatric disorder characterized by multiple tics and sensorimotor abnormalities, the severity of which is typically increased by stress. The neurobiological underpinnings of this exacerbation, however, remain elusive. We recently reported that spatial confinement (SC), a moderate environmental stressor, increases tic-like responses and elicits TS-like sensorimotor gating deficits in the D1CT-7 mouse, one of the best-validated models of TS. Here, we hypothesized that these adverse effects may be mediated by neurosteroids, given their well-documented role in stress-response orchestration. Indeed, SC increased the levels of progesterone, as well as its derivatives 5α-dihydroprogesterone and allopregnanolone, in the prefrontal cortex (PFC) of D1CT-7 mice. Among these steroids, however, only allopregnanolone (5-15 mg/kg, IP) dose-dependently exacerbated TS-like manifestations in D1CT-7, but not wild-type littermates; these effects were countered by the benchmark anti-tic therapy haloperidol (0.3 mg/kg, IP). Furthermore, the phenotypic effects of spatial confinement in D1CT-7 mice were suppressed by finasteride (25-50 mg/kg, IP), an inhibitor of the main rate-limiting enzyme in allopregnanolone synthesis. These findings collectively suggest that stress may exacerbate TS symptoms by promoting allopregnanolone synthesis in the PFC, and corroborate previous clinical results pointing to finasteride as a novel therapeutic avenue to curb symptom fluctuations in TS.
Collapse
|
27
|
Frau R, Savoia P, Fanni S, Fiorentini C, Fidalgo C, Tronci E, Stancampiano R, Meloni M, Cannas A, Marrosu F, Bortolato M, Devoto P, Missale C, Carta M. The 5-alpha reductase inhibitor finasteride reduces dyskinesia in a rat model of Parkinson's disease. Exp Neurol 2017; 291:1-7. [DOI: 10.1016/j.expneurol.2017.01.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/13/2017] [Accepted: 01/24/2017] [Indexed: 02/09/2023]
|
28
|
Exploring the neural mechanisms of finasteride: a proteomic analysis in the nucleus accumbens. Psychoneuroendocrinology 2016; 74:387-396. [PMID: 27750143 DOI: 10.1016/j.psyneuen.2016.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 09/07/2016] [Accepted: 10/05/2016] [Indexed: 02/06/2023]
Abstract
The enzyme 5α-reductase (5αR) catalyzes the conversion of progesterone and testosterone into neuroactive steroids implicated in a wide array of behavioral functions. The prototypical 5αR inhibitor, finasteride (FIN), is clinically approved for the treatment of androgenic alopecia and benign prostatic hyperplasia. Recent evidence has shown that FIN, albeit generally well tolerated, can induce untoward psychological effects in a subset of patients; furthermore, this drug may have therapeutic efficacy for a number of different neuropsychiatric conditions, ranging from Tourette syndrome to schizophrenia. In rat models of these conditions, FIN has been shown to block the effects of dopamine receptors in the nucleus accumbens (NAcc), a key terminal of the dopamine mesolimbic system. The biological underpinnings of these effects, however, remain mostly elusive. To elucidate the neurochemical networks that may be responsible for the behavioral effects of FIN, we evaluated the proteomic profile of the NAcc following acute (100mg/kg, IP) and subchronic (7 days; 100mg/kg/day, IP) treatment with this drug, in comparison with vehicle treatment (n=5/group). Two-dimensional electrophoresis (2-DE) analysis coupled to mass spectrometry revealed significant changes in the expression of nine proteins (CRMP2, PSMD1, STX18, KCNC3, CYP255, GABRP, GABT, PRPS1, CYP2B3), which were further analyzed by ontological classification (PANTHER). These results point to a number of novel potential chemical targets of FIN, and may help elucidate the underpinnings of FIN's behavioral effects and therapeutic potential for neuropsychiatric disorders.
Collapse
|
29
|
Nespoli E, Rizzo F, Boeckers TM, Hengerer B, Ludolph AG. Addressing the Complexity of Tourette's Syndrome through the Use of Animal Models. Front Neurosci 2016; 10:133. [PMID: 27092043 PMCID: PMC4824761 DOI: 10.3389/fnins.2016.00133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/16/2016] [Indexed: 01/06/2023] Open
Abstract
Tourette's syndrome (TS) is a neurodevelopmental disorder characterized by fluctuating motor and vocal tics, usually preceded by sensory premonitions, called premonitory urges. Besides tics, the vast majority—up to 90%—of TS patients suffer from psychiatric comorbidities, mainly attention deficit/hyperactivity disorder (ADHD) and obsessive-compulsive disorder (OCD). The etiology of TS remains elusive. Genetics is believed to play an important role, but it is clear that other factors contribute to TS, possibly altering brain functioning and architecture during a sensitive phase of neural development. Clinical brain imaging and genetic studies have contributed to elucidate TS pathophysiology and disease mechanisms; however, TS disease etiology still is poorly understood. Findings from genetic studies led to the development of genetic animal models, but they poorly reflect the pathophysiology of TS. Addressing the role of neurotransmission, brain regions, and brain circuits in TS disease pathomechanisms is another focus area for preclinical TS model development. We are now in an interesting moment in time when numerous innovative animal models are continuously brought to the attention of the public. Due to the diverse and largely unknown etiology of TS, there is no single preclinical model featuring all different aspects of TS symptomatology. TS has been dissected into its key symptomst hat have been investigated separately, in line with the Research Domain Criteria concept. The different rationales used to develop the respective animal models are critically reviewed, to discuss the potential of the contribution of animal models to elucidate TS disease mechanisms.
Collapse
Affiliation(s)
- Ester Nespoli
- Competence in Neuro Spine Department, Boehringer Ingelheim Pharma GmbH & Co. KGBiberach an der Riss, Germany; Department of Child and Adolescence Psychiatry/Psychotherapy, University of UlmUlm, Germany
| | - Francesca Rizzo
- Department of Child and Adolescence Psychiatry/Psychotherapy, University of UlmUlm, Germany; Institute of Anatomy and Cell Biology, University of UlmUlm, Germany
| | - Tobias M Boeckers
- Institute of Anatomy and Cell Biology, University of Ulm Ulm, Germany
| | - Bastian Hengerer
- Competence in Neuro Spine Department, Boehringer Ingelheim Pharma GmbH & Co. KG Biberach an der Riss, Germany
| | - Andrea G Ludolph
- Department of Child and Adolescence Psychiatry/Psychotherapy, University of Ulm Ulm, Germany
| |
Collapse
|
30
|
5α-Reductase Inhibition Prevents the Luteal Phase Increase in Plasma Allopregnanolone Levels and Mitigates Symptoms in Women with Premenstrual Dysphoric Disorder. Neuropsychopharmacology 2016; 41:1093-102. [PMID: 26272051 PMCID: PMC4748434 DOI: 10.1038/npp.2015.246] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 07/07/2015] [Accepted: 07/18/2015] [Indexed: 11/08/2022]
Abstract
Changes in neurosteroid levels during the luteal phase of the menstrual cycle may precipitate affective symptoms. To test this hypothesis, we stabilized neurosteroid levels by administering the 5α-reductase inhibitor dutasteride to block conversion of progesterone to its neurosteroid metabolite allopregnanolone in women with premenstrual dysphoric disorder (PMDD) and in asymptomatic control women. Sixteen women with prospectively confirmed PMDD and 16 control women participated in one of two separate randomized, double-blind, placebo-controlled, cross-over trials, each lasting three menstrual cycles. After one menstrual cycle of single-blind placebo, participants were randomized to receive, for the next two menstrual cycles, either double-blind placebo or dutasteride (low-dose 0.5 mg/day in the first eight PMDD and eight control women or high-dose 2.5 mg/day in the second group of women). All women completed the daily rating form (DRF) and were evaluated in clinic during the follicular and luteal phases of each menstrual cycle. Main outcome measures were the DRF symptoms of irritability, sadness, and anxiety. Analyses were performed with SAS PROC MIXED. In the low-dose group, no significant effect of dutasteride on PMDD symptoms was observed compared with placebo (ie, symptom cyclicity maintained), and plasma allopregnanolone levels increased in women with PMDD from follicular to the luteal phases, suggesting the absence of effect of the low-dose dutasteride on 5α-reductase. In contrast, the high-dose group experienced a statistically significant reduction in several core PMDD symptoms (ie, irritability, sadness, anxiety, food cravings, and bloating) on dutasteride compared with placebo. Dutasteride had no effect on mood in controls. Stabilization of allopregnanolone levels from the follicular to the luteal phase of the menstrual cycle by blocking the conversion of progesterone to its 5α-reduced neurosteroid metabolite mitigates symptoms in PMDD. These data provide preliminary support for the pathophysiologic relevance of neurosteroids in this condition.
Collapse
|
31
|
Frau R, Mosher LJ, Bini V, Pillolla G, Pes R, Saba P, Fanni S, Devoto P, Bortolato M. The neurosteroidogenic enzyme 5α-reductase modulates the role of D1 dopamine receptors in rat sensorimotor gating. Psychoneuroendocrinology 2016; 63:59-67. [PMID: 26415119 PMCID: PMC4695380 DOI: 10.1016/j.psyneuen.2015.09.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 08/21/2015] [Accepted: 09/14/2015] [Indexed: 01/26/2023]
Abstract
Neurosteroids exert diverse modulatory actions on dopamine neurotransmission and signaling. We previously documented that the enzyme 5α-reductase, which catalyzes the main rate-limiting step in neurosteroid synthesis, is required for the behavioral responses of Sprague-Dawley rats to non-selective dopaminergic agonists, such as the D1-D2 receptor agonist apomorphine. Specifically, systemic and intra-accumbal administrations of the 5α-reductase inhibitor finasteride countered apomorphine-induced deficits of sensorimotor gating, as measured by the prepulse inhibition (PPI) of the startle reflex; the classes of dopamine receptors involved in these effects, however, remain unknown. Prior rodent studies have revealed that the contributions of dopamine receptors to PPI regulation vary depending on the genetic background; thus, we analyzed the effect of finasteride on the PPI deficits induced by selective dopamine receptor agonists in Long-Evans (a strain exhibiting PPI deficits in response to both D1 and D2 receptor agonists) and Sprague-Dawley rats (which display PPI reductions following treatment with D2, and D3, but not D1 receptor agonists). In Long-Evans rats, finasteride opposed the PPI deficits induced by activation of D1, but not D2 receptors; conversely, in Sprague-Dawley rats, finasteride prevented the reductions in %PPI and accumbal dopamine extracellular levels caused by selective stimulation of D3, but not D2 receptors; however, the effects on %PPI were not confirmed by analyses on absolute PPI values. Our findings suggest that 5α-reductase modulates the effects of D1, but not D2 receptor agonists on sensorimotor gating. These data may help elucidate the role of neurosteroids in neuropsychiatric disorders featuring PPI deficits, including schizophrenia and Tourette syndrome.
Collapse
MESH Headings
- 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism
- 5-alpha Reductase Inhibitors/pharmacology
- Animals
- Dopamine Agonists/pharmacology
- Finasteride/pharmacology
- Male
- Microdialysis
- Nucleus Accumbens/drug effects
- Nucleus Accumbens/metabolism
- Prepulse Inhibition/drug effects
- Prepulse Inhibition/physiology
- Rats
- Rats, Long-Evans
- Rats, Sprague-Dawley
- Receptors, Dopamine D1/drug effects
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D2/drug effects
- Receptors, Dopamine D2/metabolism
- Receptors, Dopamine D3/drug effects
- Receptors, Dopamine D3/metabolism
- Reflex, Startle/drug effects
- Reflex, Startle/physiology
- Sensory Gating/drug effects
- Sensory Gating/physiology
Collapse
Affiliation(s)
- Roberto Frau
- "Guy Everett" Laboratory, Dept. of Biomedical Sciences, University of Cagliari, Italy; Tourette Syndrome Center, University of Cagliari, Italy
| | - Laura J Mosher
- Dept. of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA; Consortium for Translational Research on Aggression and Drug Abuse (ConTRADA), University of Kansas, Lawrence, KS, USA; Problem Gambling Research Studies (ProGResS) Network, University of Kansas, Lawrence, KS, USA
| | - Valentina Bini
- "Guy Everett" Laboratory, Dept. of Biomedical Sciences, University of Cagliari, Italy; Tourette Syndrome Center, University of Cagliari, Italy
| | - Giuliano Pillolla
- "Guy Everett" Laboratory, Dept. of Biomedical Sciences, University of Cagliari, Italy
| | - Romina Pes
- "Guy Everett" Laboratory, Dept. of Biomedical Sciences, University of Cagliari, Italy; Dept. of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | - Pierluigi Saba
- "Guy Everett" Laboratory, Dept. of Biomedical Sciences, University of Cagliari, Italy
| | - Silvia Fanni
- "Guy Everett" Laboratory, Dept. of Biomedical Sciences, University of Cagliari, Italy
| | - Paola Devoto
- "Guy Everett" Laboratory, Dept. of Biomedical Sciences, University of Cagliari, Italy; Tourette Syndrome Center, University of Cagliari, Italy
| | - Marco Bortolato
- Tourette Syndrome Center, University of Cagliari, Italy; Dept. of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA; Consortium for Translational Research on Aggression and Drug Abuse (ConTRADA), University of Kansas, Lawrence, KS, USA; Problem Gambling Research Studies (ProGResS) Network, University of Kansas, Lawrence, KS, USA.
| |
Collapse
|
32
|
Neuroactive gonadal drugs for neuroprotection in male and female models of Parkinson's disease. Neurosci Biobehav Rev 2015; 67:79-88. [PMID: 26708712 DOI: 10.1016/j.neubiorev.2015.09.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/18/2015] [Accepted: 09/24/2015] [Indexed: 12/19/2022]
Abstract
The existence of sex differences in Parkinson's disease (PD) incidence is well documented with greater prevalence and earlier age at onset in men than in women. These reported sex differences could be related to estrogen exposure. In PD animal models, estrogen is well documented to be neuroprotective against dopaminergic neuron loss induced by neurotoxins. Using the 1-methyl 4-phenyl-1,2,3,6 tetrahydropyridine (MPTP) mouse model, we showed that several compounds are neuroprotective on dopaminergic neurons including estrogen, the selective estrogen receptor modulator raloxifene, progesterone, dehydroepiandrosterone, the estrogen receptor alpha (ERα) agonist PPT as well as the G protein-coupled membrane estrogen receptor (GPER1) specific agonist G1. Accumulating evidence suggests that GPER1 could be implicated in the neuroprotective effects of estrogen, raloxifene and G1 in collaboration with ERα. We recently reported that the 5α-reductase inhibitor Dutasteride is also neuroprotective and could bring an alternative to estrogens for therapy in male. Additional studies are needed to optimize therapies with these gonadal drugs into safe personalized treatments according to sex for treatment of PD.
Collapse
|
33
|
Frau R, Abbiati F, Bini V, Casti A, Caruso D, Devoto P, Bortolato M. Targeting neurosteroid synthesis as a therapy for schizophrenia-related alterations induced by early psychosocial stress. Schizophr Res 2015; 168:640-8. [PMID: 25999042 PMCID: PMC4628592 DOI: 10.1016/j.schres.2015.04.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 04/29/2015] [Accepted: 04/29/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Cogent evidence has shown that schizophrenia vulnerability is enhanced by psychosocial stress in adolescence, yet the underpinnings of this phenomenon remain elusive. One of the animal models that best capture the relationship between juvenile stress and schizophrenia is isolation rearing (IR). This manipulation, which consists in subjecting rats to social isolation from weaning through adulthood, results in neurobehavioral alterations akin to those observed in schizophrenia patients. In particular, IR-subjected rats display a marked reduction of the prepulse inhibition (PPI) of the startle reflex, which are posited to reflect imbalances in dopamine neurotransmission in the nucleus accumbens (NAcc). We recently documented that the key neurosteroidogenic enzyme 5α-reductase (5αR) plays an important role in the dopaminergic regulation of PPI; given that IR leads to a marked down-regulation of this enzyme in the NAcc, the present study was designed to further elucidate the functional role of 5αR in the regulation of PPI of IR-subjected rats. METHODS We studied the impact of the prototypical 5αR inhibitor finasteride (FIN) on the PPI deficits and NAcc steroid profile of IR-subjected male rats, in comparison with socially reared (SR) controls. RESULTS FIN (25-100 mg/kg, i.p.) dose-dependently countered IR-induced PPI reduction, without affecting gating integrity in SR rats. The NAcc and striatum of IR-subjected rats displayed several changes in neuroactive steroid profile, including a reduction in pregnenolone in both SR and IR-subjected groups, as well as a decrease in allopregnanolone content in the latter group; both effects were significantly opposed by FIN. CONCLUSIONS These results show that 5αR inhibition counters the PPI deficits induced by IR, possibly through limbic changes in pregnenolone and/or allopregnanolone concentrations.
Collapse
Affiliation(s)
- Roberto Frau
- “Guy Everett” Laboratory, Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy,Tourette Syndrome Center, University of Cagliari, Cagliari, Italy
| | - Federico Abbiati
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases; University of Milan, Milan, Italy
| | - Valentina Bini
- “Guy Everett” Laboratory, Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy,Tourette Syndrome Center, University of Cagliari, Cagliari, Italy
| | - Alberto Casti
- “Guy Everett” Laboratory, Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases; University of Milan, Milan, Italy
| | - Paola Devoto
- “Guy Everett” Laboratory, Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy,Tourette Syndrome Center, University of Cagliari, Cagliari, Italy
| | - Marco Bortolato
- Tourette Syndrome Center, University of Cagliari, Cagliari, Italy; Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA; Consortium for Translational Research on Aggression and Drug Abuse (ConTRADA), University of Kansas, Lawrence, KS, USA; Problem Gambling Research Studies (ProGResS) Network, University of Kansas, Lawrence, KS, USA.
| |
Collapse
|
34
|
Traish AM, Melcangi RC, Bortolato M, Garcia-Segura LM, Zitzmann M. Adverse effects of 5α-reductase inhibitors: What do we know, don't know, and need to know? Rev Endocr Metab Disord 2015; 16:177-98. [PMID: 26296373 DOI: 10.1007/s11154-015-9319-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Steroids are important physiological orchestrators of endocrine as well as peripheral and central nervous system functions. One of the key processes for regulation of these molecules lies in their enzymatic processing by a family of 5α-reductase (5α-Rs) isozymes. By catalyzing a key rate-limiting step in steroidogenesis, this family of enzymes exerts a crucial role not only in the physiological control but also in pathological events. Indeed, both 5α-R inhibition and supplementation of 5α-reduced metabolites are currently used or have been proposed as therapeutic strategies for a wide array of pathological conditions. In particular, the potent 5α-R inhibitors finasteride and dutasteride are used in the treatments of benign prostatic hyperplasia (BPH), as well as in male pattern hair loss (MPHL) known as androgenetic alopecia (AGA). Recent preclinical and clinical findings indicate that 5α-R inhibitors evoke not only beneficial, but also adverse effects. Future studies should investigate the biochemical and physiological mechanisms that underlie the persistence of the adverse sexual side effects to determine why a subset of patients is afflicted with such persistence or irreversible adverse effects. Also a better focus of clinical research is urgently needed to better define those subjects who are likely to be adversely affected by such agents. Furthermore, research on the non-sexual adverse effects such as diabetes, psychosis, depression, and cognitive function are needed to better understand the broad spectrum of the effects these drugs may elicit during their use in treatment of AGA or BPH. In this review, we will summarize the state of art on this topic, overview the key unresolved questions that have emerged on the pharmacological targeting of these enzymes and their products, and highlight the need for further studies to ascertain the severity and duration of the adverse effects of 5α-R inhibitors, as well as their biological underpinnings.
Collapse
Affiliation(s)
- Abdulmaged M Traish
- Department of Biochemistry and Department of Urology, Boston University School of Medicine, 715 Albany Street, A502, Boston, MA, 02118, USA.
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences- Center of Excellence on Neurodegenerative Diseases, Iniversità degli Studi di Milano, Milan, Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | | | - Michael Zitzmann
- Centre for Reproductive Medicine and Andrology, University Clinics Muenster, Domagkstrasse 11, D-48149, Muenster, Germany
| |
Collapse
|
35
|
Purves-Tyson TD, Boerrigter D, Allen K, Zavitsanou K, Karl T, Djunaidi V, Double KL, Desai R, Handelsman DJ, Weickert CS. Testosterone attenuates and the selective estrogen receptor modulator, raloxifene, potentiates amphetamine-induced locomotion in male rats. Horm Behav 2015; 70:73-84. [PMID: 25747465 DOI: 10.1016/j.yhbeh.2015.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 01/16/2015] [Accepted: 02/28/2015] [Indexed: 11/17/2022]
Abstract
Although sex steroids are known to modulate brain dopamine, it is still unclear how testosterone modifies locomotor behaviour controlled, at least in part, by striatal dopamine in adolescent males. Our previous work suggests that increasing testosterone during adolescence may bias midbrain neurons to synthesise more dopamine. We hypothesised that baseline and amphetamine-induced locomotion would differ in adult males depending on testosterone exposure during adolescence. We hypothesised that concomitant stimulation of estrogen receptor signaling, through a selective estrogen receptor modulator (SERM), raloxifene, can counter testosterone effects on locomotion. Male Sprague-Dawley rats at postnatal day 45 were gonadectomised (G) or sham-operated (S) prior to the typical adolescent testosterone increase. Gonadectomised rats were either given testosterone replacement (T) or blank implants (B) for six weeks and sham-operated (i.e. intact or endogenous testosterone group) were given blank implants. Subgroups of sham-operated, gonadectomised and gonadectomised/testosterone-replaced rats were treated with raloxifene (R, 5mg/kg) or vehicle (V), daily for the final four weeks. There were six groups (SBV, GBV, GTV, SBR, GBR, GTR). Saline and amphetamine-induced (1.25mg/kg) locomotion in the open field was measured at PND85. Gonadectomy increased amphetamine-induced locomotion compared to rats with endogenous or with exogenous testosterone. Raloxifene increased amphetamine-induced locomotion in rats with either endogenous or exogenous testosterone. Amphetamine-induced locomotion was negatively correlated with testosterone and this relationship was abolished by raloxifene. Lack of testosterone during adolescence potentiates and testosterone exposure during adolescence attenuates amphetamine-induced locomotion. Treatment with raloxifene appears to potentiate amphetamine-induced locomotion and to have an opposite effect to that of testosterone in male rats.
Collapse
Affiliation(s)
- Tertia D Purves-Tyson
- Schizophrenia Research Institute, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Sydney, Australia; School of Medical Sciences, Faculty of Medicine, University of New South Wales, Australia.
| | - Danny Boerrigter
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Sydney, Australia
| | - Katherine Allen
- Schizophrenia Research Institute, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Sydney, Australia; School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Katerina Zavitsanou
- Schizophrenia Research Institute, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Sydney, Australia; School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Tim Karl
- Neuroscience Research Australia, Barker Street, Sydney, Australia; School of Medical Sciences, Faculty of Medicine, University of New South Wales, Australia
| | - Vanezha Djunaidi
- Schizophrenia Research Institute, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Sydney, Australia; School of Medical Sciences, Faculty of Medicine, University of New South Wales, Australia
| | - Kay L Double
- Discipline of Biomedical Science, School of Medical Sciences, Sydney Medical School, University of Sydney, Australia
| | - Reena Desai
- ANZAC Research Institute, University of Sydney, Concord Hospital, Australia
| | - David J Handelsman
- ANZAC Research Institute, University of Sydney, Concord Hospital, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Institute, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Sydney, Australia; School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia
| |
Collapse
|
36
|
Godar SC, Mosher LJ, Di Giovanni G, Bortolato M. Animal models of tic disorders: a translational perspective. J Neurosci Methods 2014; 238:54-69. [PMID: 25244952 DOI: 10.1016/j.jneumeth.2014.09.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 09/09/2014] [Accepted: 09/11/2014] [Indexed: 12/30/2022]
Abstract
Tics are repetitive, sudden movements and/or vocalizations, typically enacted as maladaptive responses to intrusive premonitory urges. The most severe tic disorder, Tourette syndrome (TS), is a childhood-onset condition featuring multiple motor and at least one phonic tic for a duration longer than 1 year. The pharmacological treatment of TS is mainly based on antipsychotic agents; while these drugs are often effective in reducing tic severity and frequency, their therapeutic compliance is limited by serious motor and cognitive side effects. The identification of novel therapeutic targets and development of better treatments for tic disorders is conditional on the development of animal models with high translational validity. In addition, these experimental tools can prove extremely useful to test hypotheses on the etiology and neurobiological bases of TS and related conditions. In recent years, the translational value of these animal models has been enhanced, thanks to a significant re-organization of our conceptual framework of neuropsychiatric disorders, with a greater focus on endophenotypes and quantitative indices, rather than qualitative descriptors. Given the complex and multifactorial nature of TS and other tic disorders, the selection of animal models that can appropriately capture specific symptomatic aspects of these conditions can pose significant theoretical and methodological challenges. In this article, we will review the state of the art on the available animal models of tic disorders, based on genetic mutations, environmental interventions as well as pharmacological manipulations. Furthermore, we will outline emerging lines of translational research showing how some of these experimental preparations have led to significant progress in the identification of novel therapeutic targets for tic disorders.
Collapse
Affiliation(s)
- Sean C Godar
- Department of Pharmacology and Toxicology, School of Pharmacy; University of Kansas, Lawrence, KS, USA
| | - Laura J Mosher
- Department of Pharmacology and Toxicology, School of Pharmacy; University of Kansas, Lawrence, KS, USA
| | - Giuseppe Di Giovanni
- Department of Physiology and Biochemistry, University of Malta, Msida, Malta; School of Biosciences, Cardiff University, Cardiff, UK
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, School of Pharmacy; University of Kansas, Lawrence, KS, USA; Consortium for Translational Research on Aggression and Drug Abuse (ConTRADA), University of Kansas, Lawrence, KS, USA.
| |
Collapse
|
37
|
Godar SC, Bortolato M. Gene-sex interactions in schizophrenia: focus on dopamine neurotransmission. Front Behav Neurosci 2014; 8:71. [PMID: 24639636 PMCID: PMC3944784 DOI: 10.3389/fnbeh.2014.00071] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 02/19/2014] [Indexed: 02/02/2023] Open
Abstract
Schizophrenia is a severe mental disorder, with a highly complex and heterogenous clinical presentation. Our current perspectives posit that the pathogenic mechanisms of this illness lie in complex arrays of gene × environment interactions. Furthermore, several findings indicate that males have a higher susceptibility for schizophrenia, with earlier age of onset and overall poorer clinical prognosis. Based on these premises, several authors have recently begun exploring the possibility that the greater schizophrenia vulnerability in males may reflect specific gene × sex (G×S) interactions. Our knowledge on such G×S interactions in schizophrenia is still rudimentary; nevertheless, the bulk of preclinical evidence suggests that the molecular mechanisms for such interactions are likely contributed by the neurobiological effects of sex steroids on dopamine (DA) neurotransmission. Accordingly, several recent studies suggest a gender-specific association of certain DAergic genes with schizophrenia. These G×S interactions have been particularly documented for catechol-O-methyltransferase (COMT) and monoamine oxidase (MAO), the main enzymes catalyzing DA metabolism. In the present review, we will outline the current evidence on the interactions of DA-related genes and sex-related factors, and discuss the potential molecular substrates that may mediate their cooperative actions in schizophrenia pathogenesis.
Collapse
Affiliation(s)
- Sean C Godar
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas Lawrence, KS, USA
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas Lawrence, KS, USA ; Consortium for Translational Research on Aggression and Drug Abuse, University of Kansas Lawrence, KS, USA
| |
Collapse
|
38
|
Inhibition of 17α-hydroxylase/C17,20 lyase reduces gating deficits consequent to dopaminergic activation. Psychoneuroendocrinology 2014; 39:204-213. [PMID: 24140269 PMCID: PMC3940882 DOI: 10.1016/j.psyneuen.2013.09.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 09/01/2013] [Accepted: 09/15/2013] [Indexed: 11/23/2022]
Abstract
Cogent evidence points to the involvement of neurosteroids in the regulation of dopamine (DA) neurotransmission and signaling, yet the neurobiological bases of this link remain poorly understood. We previously showed that inhibition of 5α-reductase (5αR), a key neurosteroidogenic enzyme, attenuates the sensorimotor gating deficits induced by DA receptor activation, as measured by the prepulse inhibition (PPI) of the acoustic startle reflex. To extend these findings, the present study was aimed at the assessment of the role of other key neurosteroidogenic enzymes in PPI, such as 17α-hydroxylase/C17,20 lyase (CYP17A1), 3α- and 3β-hydroxysteroid dehydrogenase (HSD), in Sprague-Dawley rats. The PPI deficits induced by the DAergic non-selective agonist apomorphine (APO, 0.25mg/kg, SC) were dose-dependently attenuated by the selective CYP17A1 inhibitor abiraterone (ABI, 10-50mg/kg, IP) in a fashion akin to that of the 5αR inhibitor finasteride (FIN, 100mg/kg, IP). These systemic effects were reproduced by intracerebroventricular injection of ABI (1 μg/1 μl), suggesting the involvement of brain CYP17A1 in PPI regulation. Conversely, the PPI disruption induced by APO was not significantly affected by the 3α- and 3β-HSD inhibitors indomethacin and trilostane. Given that CYP17A1 catalyzes androgen synthesis, we also tested the impact on PPI of the androgen receptor (AR) antagonist flutamide (10mg/kg, IP). However, this agent failed to reverse APO-induced PPI deficits; furthermore, AR endogenous ligands testosterone and dihydrotestosterone failed to disrupt PPI. Collectively, these data highlight CYP17A1 as a novel target for antipsychotic-like action, and suggest that the DAergic regulation of PPI is modulated by androgenic neurosteroids, through AR-unrelated mechanisms.
Collapse
|
39
|
Bortolato M, Frau R, Godar SC, Mosher LJ, Paba S, Marrosu F, Devoto P. The implication of neuroactive steroids in Tourette's syndrome pathogenesis: A role for 5α-reductase? J Neuroendocrinol 2013; 25:1196-208. [PMID: 23795653 PMCID: PMC3849218 DOI: 10.1111/jne.12066] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Revised: 06/01/2013] [Accepted: 06/18/2013] [Indexed: 01/04/2023]
Abstract
Tourette's syndrome (TS) is a neurodevelopmental disorder characterised by recurring motor and phonic tics. The pathogenesis of TS is considered to reflect dysregulations in the signalling of dopamine (DA) and other neurotransmitters, which lead to excitation/inhibition imbalances in cortico-striato-thalamocortical circuits. The causes of these deficits may reflect complex gene × environment × sex (G × E × S) interactions; indeed, the disorder is markedly predominant in males, with a male-to-female prevalence ratio of approximately 4 : 1. Converging lines of evidence point to neuroactive steroids as being likely molecular candidates to account for G × E × S interactions in TS. Building on these premises, our group has begun examining the possibility that alterations in the steroid biosynthetic process may be directly implicated in TS pathophysiology; in particular, our research has focused on 5α-reductase (5αR), the enzyme catalysing the key rate-limiting step in the synthesis of pregnane and androstane neurosteroids. In clinical and preclinical studies, we found that 5αR inhibitors exerted marked anti-DAergic and tic-suppressing properties, suggesting a central role for this enzyme in TS pathogenesis. Based on these data, we hypothesise that enhancements in 5αR activity in early developmental stages may lead to an inappropriate activation of the 'backdoor' pathway for androgen synthesis from adrenarche until the end of puberty. We predict that the ensuing imbalances in steroid homeostasis may impair the signalling of DA and other neurotransmitters, ultimately resulting in the facilitation of tics and other behavioural abnormalities in TS.
Collapse
Affiliation(s)
- Marco Bortolato
- Dept. of Pharmacology and Toxicology, School of Pharmacy; University of Kansas, Lawrence (KS), USA
| | - Roberto Frau
- Dept. of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato (CA), Italy
| | - Sean C Godar
- Dept. of Pharmacology and Toxicology, School of Pharmacy; University of Kansas, Lawrence (KS), USA
| | - Laura J Mosher
- Dept. of Pharmacology and Toxicology, School of Pharmacy; University of Kansas, Lawrence (KS), USA
| | - Silvia Paba
- Dept. of Public Health, Clinical and Molecular Medicine, Section of Neurology, University of Cagliari, Monserrato (CA), Italy
| | - Francesco Marrosu
- Dept. of Public Health, Clinical and Molecular Medicine, Section of Neurology, University of Cagliari, Monserrato (CA), Italy
| | - Paola Devoto
- Dept. of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato (CA), Italy
| |
Collapse
|
40
|
Wyatt LR, Godar SC, Khoja S, Jakowec MW, Alkana RL, Bortolato M, Davies DL. Sociocommunicative and sensorimotor impairments in male P2X4-deficient mice. Neuropsychopharmacology 2013; 38:1993-2002. [PMID: 23604007 PMCID: PMC3746707 DOI: 10.1038/npp.2013.98] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 04/08/2013] [Accepted: 04/10/2013] [Indexed: 12/23/2022]
Abstract
Purinergic P2X receptors are a family of ligand-gated ion channels gated by extracellular adenosine 5'-triphosphate (ATP). Of the seven P2X subtypes, P2X4 receptors (P2X4Rs) are richly expressed in the brain, yet their role in behavioral organization remains poorly understood. In this study, we examined the behavioral responses of P2X4R heterozygous (HZ) and knockout (KO) mice in a variety of testing paradigms designed to assess complementary aspects of sensory functions, emotional reactivity, and cognitive organization. P2X4R deficiency did not induce significant alterations of locomotor activity and anxiety-related indices in the novel open field and elevated plus-maze tests. Conversely, P2X4R KO mice displayed marked deficits in acoustic startle reflex amplitude, as well as significant sensorimotor gating impairments, as assessed by the prepulse inhibition of the startle. In addition, P2X4R KO mice displayed enhanced tactile sensitivity, as signified by a lower latency in the sticky-tape removal test. Moreover, both P2X4R HZ and KO mice showed significant reductions in social interaction and maternal separation-induced ultrasonic vocalizations in pups. Notably, brain regions of P2X4R KO mice exhibited significant brain-regional alterations in the subunit composition of glutamate ionotropic receptors. These results collectively document that P2X4-deficient mice exhibit a spectrum of phenotypic abnormalities partially akin to those observed in other murine models of autism-spectrum disorder. In conclusion, our findings highlight a putative role of P2X4Rs in the regulation of perceptual and sociocommunicative functions and point to these receptors as putative targets for disturbances associated with neurodevelopmental disorders.
Collapse
Affiliation(s)
- Letisha R Wyatt
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Sean C Godar
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Sheraz Khoja
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Michael W Jakowec
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Ronald L Alkana
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Daryl L Davies
- Titus Family Department of Clinical Pharmacy and Pharmaceutical Economics and Policy, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
41
|
Hopf FW, Seif T, Chung S, Civelli O. MCH and apomorphine in combination enhance action potential firing of nucleus accumbens shell neurons in vitro. PeerJ 2013; 1:e61. [PMID: 23646281 PMCID: PMC3642701 DOI: 10.7717/peerj.61] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 03/12/2013] [Indexed: 11/20/2022] Open
Abstract
The MCH and dopamine receptor systems have been shown to modulate a number of behaviors related to reward processing, addiction, and neuropsychiatric conditions such as schizophrenia and depression. In addition, MCH and dopamine receptors can interact in a positive manner, for example in the expression of cocaine self-administration. A recent report (Chung et al., 2011a) showed that the DA1/DA2 dopamine receptor activator apomorphine suppresses pre-pulse inhibition, a preclinical model for some aspects of schizophrenia. Importantly, MCH can enhance the effects of lower doses of apomorphine, suggesting that co-modulation of dopamine and MCH receptors might alleviate some symptoms of schizophrenia with a lower dose of dopamine receptor modulator and thus fewer potential side effects. Here, we investigated whether MCH and apomorphine could enhance action potential firing in vitro in the nucleus accumbens shell (NAshell), a region which has previously been shown to mediate some behavioral effects of MCH. Using whole-cell patch-clamp electrophysiology, we found that MCH, which has no effect on firing on its own, was able to increase NAshell firing when combined with a subthreshold dose of apomorphine. Further, this MCH/apomorphine increase in firing was prevented by an antagonist of either a DA1 or a DA2 receptor, suggesting that apomorphine acts through both receptor types to enhance NAshell firing. The MCH/apomorphine-mediated firing increase was also prevented by an MCH receptor antagonist or a PKA inhibitor. Taken together, our results suggest that MCH can interact with lower doses of apomorphine to enhance NAshell firing, and thus that MCH and apomorphine might interact in vivo within the NAshell to suppress pre-pulse inhibition.
Collapse
Affiliation(s)
- F Woodward Hopf
- Ernest Gallo Clinic and Research Center, Department of Neurology, University of California , San Francisco, Emeryville, CA , USA
| | | | | | | |
Collapse
|
42
|
Frau R, Pillolla G, Bini V, Tambaro S, Devoto P, Bortolato M. Inhibition of 5α-reductase attenuates behavioral effects of D1-, but not D2-like receptor agonists in C57BL/6 mice. Psychoneuroendocrinology 2013; 38:542-51. [PMID: 22877998 PMCID: PMC3540184 DOI: 10.1016/j.psyneuen.2012.07.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2012] [Revised: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 11/30/2022]
Abstract
Converging lines of evidence point to the involvement of neurosteroids in the regulation of dopamine (DA) neurotransmission and signaling, yet the neurobiological bases of this link remain poorly understood. We previously showed that inhibition of steroid 5α-reductase (5αR), the key rate-limiting enzyme in neurosteroidogenesis, attenuates the behavioral effects of non-selective DA receptor agonists in rats, including stereotyped responses and sensorimotor gating deficits, as measured by the prepulse inhibition (PPI) of the acoustic startle reflex. Since previous findings suggested that the role of DA D(1)- and D(2)-like receptor families in behavioral regulation may exhibit broad interspecies and interstrain variations, we assessed the impact of 5αR blockade on the behavioral effects of DAergic agonists in C57BL/6 mice. The prototypical 5αR inhibitor finasteride (FIN; 25-50 mg/kg, intraperitoneally, IP) dose-dependently countered the PPI deficits and the enhancement of rearing responses induced by the full D(1)-like receptor agonist SKF-82958 (0.3 mg/kg, IP); however, FIN did not significantly affect the hyperlocomotive and startle-attenuating effects of SKF-82958. Whereas the D(2)-like receptor agonist quinpirole (QUIN; 0.5 mg/kg, IP) did not induce significant changes in PPI, the combination of this agent and FIN surprisingly produced marked gating and startle deficits. In contrast with previous data on rats, FIN did not affect the reductions of startle reflex and PPI produced by the non-selective DAergic agonist apomorphine (APO; 0.5 mg/kg, IP). These findings collectively indicate that, in C57BL/6 mice, 5αR differentially modulates the effects of D(1)- and D(2)-like receptor agonists in behavioral regulation.
Collapse
Affiliation(s)
- Roberto Frau
- “Guy Everett” laboratory, Dept. of Biomedical Sciences, University of Cagliari, Italy
- Tourette Syndrome Center, University of Cagliari, Italy
| | - Giuliano Pillolla
- “Guy Everett” laboratory, Dept. of Biomedical Sciences, University of Cagliari, Italy
| | - Valentina Bini
- “Guy Everett” laboratory, Dept. of Biomedical Sciences, University of Cagliari, Italy
| | - Simone Tambaro
- Dept. of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles (CA), USA
| | - Paola Devoto
- “Guy Everett” laboratory, Dept. of Biomedical Sciences, University of Cagliari, Italy
| | - Marco Bortolato
- Tourette Syndrome Center, University of Cagliari, Italy
- Dept. of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles (CA), USA
- Corresponding author: Marco Bortolato, MD PhD, Dept. of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Rm. 527, PSC 1985 Zonal Ave, Los Angeles, CA 90089, Phone: 323-442-3225, Fax: 323-442-3229,
| |
Collapse
|
43
|
Regional distribution of 5α-reductase type 2 in the adult rat brain: an immunohistochemical analysis. Psychoneuroendocrinology 2013; 38:281-93. [PMID: 22776423 PMCID: PMC3762250 DOI: 10.1016/j.psyneuen.2012.06.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 06/18/2012] [Accepted: 06/18/2012] [Indexed: 11/20/2022]
Abstract
The enzyme 5α-reductase (5αR) catalyzes the conversion of testosterone and other Δ(4)-3-ketosteroids into their 5α-reduced metabolites. Of the five members of the 5αR family, the type 2 enzyme (5αR2) plays a key role in androgen metabolism, and is abundantly distributed in the urogenital system. Although 5αR2 has been reported to be highly expressed in the brain during early developmental stages, little is currently known on its anatomical and cellular distribution in the adult brain. Thus, the present study was designed to determine the detailed localization of 5αR2 in the adult rat brain, using a highly specific polyclonal antibody against this isoform. Parasagittal and coronal sections revealed 5αR2 immunoreactivity throughout most brain regions, with strong immunolabeling in the layers III and VI of the prefrontal and somatosensory cortex, olfactory bulb, thalamic nuclei, CA3 field of hippocampus, basolateral amygdala and Purkinje cell layer of cerebellum. Lower 5αR2 levels were detected in the hypothalamus and midbrain. Moreover, double labeling fluorescence with confocal laser scanning microscopy (CLSM) revealed that 5αR2 is localized in neurons, but not in glial cells. Specifically, the enzyme was documented in the pyramidal neurons of the cortex by CLSM analysis of simultaneous Golgi-Cox and immunofluorescent staining. Finally, low levels of 5αR2 expression were identified in GABAergic cells across the cortex, hippocampus and striatum. These findings show that, in the adult brain, 5αR2 is distributed in critical regions for behavioral regulation, suggesting that the functional role of this isoform is present throughout the entire lifespan of the individual.
Collapse
|
44
|
Martino D, Macerollo A, Leckman JF. Neuroendocrine aspects of Tourette syndrome. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2013; 112:239-79. [PMID: 24295624 DOI: 10.1016/b978-0-12-411546-0.00009-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There is sparse evidence suggesting the participation of neuroendocrine mechanisms, mainly involving sex and stress steroid hormones, to the pathophysiology of neurodevelopmental disorders such as Tourette syndrome (TS) and obsessive-compulsive disorder (OCD). Patients with TS exhibit a sex-specific variability in gender distribution (male/female ratio=3-4/1) and in its natural history, with a severity peak in the period around puberty. The administration of exogenous androgens may worsen tics in males with TS, whereas drugs counteracting the action of testosterone might show some antitic efficacy. This suggests a higher susceptibility of patients with TS to androgen steroids. There are insufficient data on the regulation of the hypothalamic-pituitary-gonadal (HPG) axis in TS. However, preliminary evidence suggests that a subgroup of women with TS might be more sensitive to the premenstrual trough of estrogen levels. Patients with TS exhibit differences in a number of behavioral, cognitive, and anatomical traits that appear to be sex related. There is a body of evidence supporting, albeit indirectly, the hypothesis of an increased exposure to androgenic steroids during the very early phases of neural development. Animal models in rodents suggest a complex role of gonadal hormones upon the modulation of anxiety-related and stereotyped behaviors during adult life. Patients with TS exhibit an enhanced reactivity of the hypothalamic-pituitary-adrenal axis to external stressors, despite a preserved diurnal cortisol rhythm and a normal restoration of the baseline activity of the axis following the acute stress response. Preliminary evidence suggests the possible implication of oxytocin (OT) in disorders related to the TS spectrum, especially non-tic-related OCD. The injection of OT in the amygdala of rodents was shown to be able to induce hypergrooming, suggesting the possible involvement of this neuropeptide in the pathophysiology of complex, stereotyped behaviors. In contrast, there is anecdotal clinical evidence that tics improve following periods of affectionate touch and sexual intercourse.
Collapse
Affiliation(s)
- Davide Martino
- Queen Elizabeth Hospital, Woolwich, London, United Kingdom; Centre for Neuroscience and Trauma, Queen Mary University of London, London, United Kingdom; King's College Hospital, London, United Kingdom.
| | | | | |
Collapse
|
45
|
Swerdlow NR. Update: studies of prepulse inhibition of startle, with particular relevance to the pathophysiology or treatment of Tourette Syndrome. Neurosci Biobehav Rev 2012; 37:1150-6. [PMID: 23017868 DOI: 10.1016/j.neubiorev.2012.09.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 08/28/2012] [Accepted: 09/05/2012] [Indexed: 11/19/2022]
Abstract
Prepulse inhibition of the startle reflex (PPI) is an operational measure of sensorimotor gating, in which the motor response to an abrupt, intense stimulus is inhibited by a weak lead stimulus. PPI is reduced in several brain disorders, including Tourette Syndrome (TS); it is regulated by forebrain circuitry, including portions of the basal ganglia implicated in the pathophysiology of TS, and is also heritable and under strong genetic control. PPI has been the focus of numerous translational models, because it is expressed by most mammalian species, with remarkable conservation of response characteristics and underlying neural circuitry between rodents and primates. Several of these models have recently explored causative factors in TS - from genes to specific basal ganglia perturbations - as well as potential TS therapeutics, including novel pharmacological and neurosurgical interventions. With the focus on Comprehensive Behavioral Interventions for Tics (CBIT) in the evolving treatment model for TS, future studies might apply PPI as a predictive measure for CBIT response, or for identifying medications that might augment CBIT efficacy. In the end, a measure based on a simple pontine-based reflex will have limitations in its ability to explicate any complex behavioral phenotype.
Collapse
Affiliation(s)
- Neal R Swerdlow
- Department of Psychiatry, School of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0804, USA.
| |
Collapse
|