1
|
Ilatovskaya DV, Ogola B, Faulkner JL, Mamenko M, Taylor EB, Dent E, Ryan MJ, Sullivan JC. Guidelines for sex-specific considerations to improve rigor in renal research and how we got there. Am J Physiol Renal Physiol 2025; 328:F204-F217. [PMID: 39705719 PMCID: PMC12146879 DOI: 10.1152/ajprenal.00136.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/22/2024] Open
Abstract
Biological sex significantly influences disease presentation, progression, and therapeutic outcomes in chronic kidney disease and acute kidney injury. Sex hormones, including estrogen and testosterone, modulate key renal functions, including renal blood flow, glomerular filtration, and electrolyte transport, thereby affecting disease trajectory in a sex-specific manner. It is critical for researchers to understand why and how to integrate sex as a biological variable in data collection, analysis, and reporting. Integrating a sex-based perspective in kidney research will lead to more personalized and efficacious treatment strategies, optimizing therapeutic interventions for each sex. If addressed properly, the incorporation of sex as a biological variable (SABV) in renal research not only enhances the mechanistic understanding of renal disease, but also paves the way for precision medicine, promising improved clinical outcomes, and tailored treatment protocols for all patients. This paper is designed to serve as a guideline for researchers interested in rigorously incorporating sex as a biological variable in their studies.
Collapse
Affiliation(s)
- Daria V Ilatovskaya
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Benard Ogola
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Jessica L Faulkner
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Mykola Mamenko
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Erin B Taylor
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Elena Dent
- The Graduate School, Augusta University, Augusta, Georgia, United States
| | - Michael J Ryan
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina, United States
- Columbia VA Health Care System, Columbia, South Carolina, United States
| | - Jennifer C Sullivan
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- The Graduate School, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
2
|
Miller CN, Li Y, Beier KT, Aoto J. Acute stress causes sex-dependent changes to ventral subiculum synapses, circuitry, and anxiety-like behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606264. [PMID: 39131353 PMCID: PMC11312572 DOI: 10.1101/2024.08.02.606264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Experiencing a single severe stressor is sufficient to drive sexually dimorphic psychiatric disease development. The ventral subiculum (vSUB) emerges as a site where stress may induce sexually dimorphic adaptations due to its sex-specific organization and pivotal role in stress integration. Using a 1-hr acute restraint stress model, we uncover that stress causes a net decrease in vSUB activity in females that is potent, long-lasting, and driven by adrenergic receptor signaling. By contrast, males exhibit a net increase in vSUB activity that is transient and driven by corticosterone signaling. We further identified sex-dependent changes in vSUB output to the bed nucleus of the stria terminalis and in anxiety-like behavior in response to stress. These findings reveal striking changes in psychiatric disease-relevant brain regions and behavior following stress with sex-, cell-type, and synapse-specificity that contribute to our understanding of sex-dependent adaptations that may shape stress-related psychiatric disease risk.
Collapse
Affiliation(s)
- Carley N Miller
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Yuan Li
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA 92697
| | - Kevin T Beier
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA 92697
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA 92697
- Department of Biomedical Engineering, University of California, Irvine, CA, USA 92697
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA 92697
| | - Jason Aoto
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
3
|
Younes S. The relationship between gender and pharmacology. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2024; 7:100192. [PMID: 39101002 PMCID: PMC11295939 DOI: 10.1016/j.crphar.2024.100192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 08/05/2024] Open
Abstract
The part of sexuality in pharmacology research was not acknowledged, and it was not thought-out to be a determinant that could impact strength and disease. For decades research has mainly contained male, women and animals, leading to a lack of news about syndromes in females. Still, it is critical to guarantee equal likeness so that determine the security, influence, and resistance of healing agents for all individuals. The underrepresentation of female models in preclinical studies over various decades has surpassed to disparities in the understanding, disease, and treatment of ailments 'tween genders. The closeness of sexuality bias has happened recognized as a contributing determinant to the restricted interpretation and replicability of preclinical research. Many demands operation have stressed the significance of including sexuality as a organic changeable, and this view is acquire growing support. Regardless of important progress in incorporating more female models into preclinical studies, differences prevail contemporary. The current review focuses on the part of sexuality and common in biomedical research and, therefore, their potential function in pharmacology and analyze the potential risks guide.
Collapse
Affiliation(s)
- Samer Younes
- Department of Pharmacy, Tartous University, Syria
| |
Collapse
|
4
|
Mabry S, Bradshaw JL, Gardner JJ, Wilson EN, Cunningham RL. Sex-dependent effects of chronic intermittent hypoxia: implication for obstructive sleep apnea. Biol Sex Differ 2024; 15:38. [PMID: 38664845 PMCID: PMC11044342 DOI: 10.1186/s13293-024-00613-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Obstructive sleep apnea (OSA) affects 10-26% of adults in the United States with known sex differences in prevalence and severity. OSA is characterized by elevated inflammation, oxidative stress (OS), and cognitive dysfunction. However, there is a paucity of data regarding the role of sex in the OSA phenotype. Prior findings suggest women exhibit different OSA phenotypes than men, which could result in under-reported OSA prevalence in women. To examine the relationship between OSA and sex, we used chronic intermittent hypoxia (CIH) to model OSA in rats. We hypothesized that CIH would produce sex-dependent phenotypes of inflammation, OS, and cognitive dysfunction, and these sex differences would be dependent on mitochondrial oxidative stress (mtOS). METHODS Adult male and female Sprague Dawley rats were exposed to CIH or normoxia for 14 days to examine the impact of sex on CIH-associated circulating inflammation (IL-1β, IL-6, IL-10, TNF-α), circulating steroid hormones, circulating OS, and behavior (recollective and spatial memory; gross and fine motor function; anxiety-like behaviors; and compulsive behaviors). Rats were implanted with osmotic minipumps containing either a mitochondria-targeting antioxidant (MitoTEMPOL) or saline vehicle 1 week prior to CIH initiation to examine how inhibiting mtOS would affect the CIH phenotype. RESULTS Sex-specific differences in CIH-induced inflammation, OS, motor function, and compulsive behavior were observed. In female rats, CIH increased inflammation (plasma IL-6 and IL-6/IL-10 ratio) and impaired fine motor function. Conversely, CIH elevated circulating OS and compulsivity in males. These sex-dependent effects of CIH were blocked by inhibiting mtOS. Interestingly, CIH impaired recollective memory in both sexes but these effects were not mediated by mtOS. No effects of CIH were observed on spatial memory, gross motor function, or anxiety-like behavior, regardless of sex. CONCLUSIONS Our results indicate that the impact of CIH is dependent on sex, such as an inflammatory response and OS response in females and males, respectively, that are mediated by mtOS. Interestingly, there was no effect of sex or mtOS in CIH-induced impairment of recollective memory. These results indicate that mtOS is involved in the sex differences observed in CIH, but a different mechanism underlies CIH-induced memory impairments.
Collapse
Affiliation(s)
- Steve Mabry
- Department of Pharmaceutical Sciences, System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107-2699, USA
| | - Jessica L Bradshaw
- Department of Pharmaceutical Sciences, System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107-2699, USA
| | - Jennifer J Gardner
- Department of Pharmaceutical Sciences, System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107-2699, USA
| | - E Nicole Wilson
- Department of Pharmaceutical Sciences, System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107-2699, USA
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107-2699, USA.
| |
Collapse
|
5
|
Dalla C, Jaric I, Pavlidi P, Hodes GE, Kokras N, Bespalov A, Kas MJ, Steckler T, Kabbaj M, Würbel H, Marrocco J, Tollkuhn J, Shansky R, Bangasser D, Becker JB, McCarthy M, Ferland-Beckham C. Practical solutions for including sex as a biological variable (SABV) in preclinical neuropsychopharmacological research. J Neurosci Methods 2024; 401:110003. [PMID: 37918446 PMCID: PMC10842858 DOI: 10.1016/j.jneumeth.2023.110003] [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: 08/29/2023] [Revised: 10/13/2023] [Accepted: 10/27/2023] [Indexed: 11/04/2023]
Abstract
Recently, many funding agencies have released guidelines on the importance of considering sex as a biological variable (SABV) as an experimental factor, aiming to address sex differences and avoid possible sex biases to enhance the reproducibility and translational relevance of preclinical research. In neuroscience and pharmacology, the female sex is often omitted from experimental designs, with researchers generalizing male-driven outcomes to both sexes, risking a biased or limited understanding of disease mechanisms and thus potentially ineffective therapeutics. Herein, we describe key methodological aspects that should be considered when sex is factored into in vitro and in vivo experiments and provide practical knowledge for researchers to incorporate SABV into preclinical research. Both age and sex significantly influence biological and behavioral processes due to critical changes at different timepoints of development for males and females and due to hormonal fluctuations across the rodent lifespan. We show that including both sexes does not require larger sample sizes, and even if sex is included as an independent variable in the study design, a moderate increase in sample size is sufficient. Moreover, the importance of tracking hormone levels in both sexes and the differentiation between sex differences and sex-related strategy in behaviors are explained. Finally, the lack of robust data on how biological sex influences the pharmacokinetic (PK), pharmacodynamic (PD), or toxicological effects of various preclinically administered drugs to animals due to the exclusion of female animals is discussed, and methodological strategies to enhance the rigor and translational relevance of preclinical research are proposed.
Collapse
Affiliation(s)
- Christina Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece.
| | - Ivana Jaric
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Pavlina Pavlidi
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Georgia E Hodes
- School of Neuroscience, Virginia Tech, Blacksburg, VA 24060, USA
| | - Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece; First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Greece
| | - Anton Bespalov
- Partnership for Assessment and Accreditation of Scientific Practice (PAASP GmbH), Heidelberg, Germany
| | - Martien J Kas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| | | | - Mohamed Kabbaj
- Department of Biomedical Sciences & Neurosciences, College of Medicine, Florida State University, USA
| | - Hanno Würbel
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Jordan Marrocco
- Department of Biology, Touro University, New York, NY 10027, USA
| | | | - Rebecca Shansky
- Department of Psychology, Northeastern University, Boston, MA 02128, USA
| | - Debra Bangasser
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Jill B Becker
- Department of Psychology and Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Margaret McCarthy
- University of Maryland School of Medicine, Department of Pharmacology, Baltimore MD, USA
| | | |
Collapse
|
6
|
Mabry S, Wilson EN, Bradshaw JL, Gardner JJ, Fadeyibi O, Vera E, Osikoya O, Cushen SC, Karamichos D, Goulopoulou S, Cunningham RL. Sex and age differences in social and cognitive function in offspring exposed to late gestational hypoxia. Biol Sex Differ 2023; 14:81. [PMID: 37951901 PMCID: PMC10640736 DOI: 10.1186/s13293-023-00557-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Gestational sleep apnea is a hypoxic sleep disorder that affects 8-26% of pregnancies and increases the risk for central nervous system dysfunction in offspring. Specifically, there are sex differences in the sensitivity of the fetal hippocampus to hypoxic insults, and hippocampal impairments are associated with social dysfunction, repetitive behaviors, anxiety, and cognitive impairment. Yet, it is unclear whether gestational sleep apnea impacts these hippocampal-associated functions and if sex and age modify these effects. To examine the relationship between gestational sleep apnea and hippocampal-associated behaviors, we used chronic intermittent hypoxia (CIH) to model late gestational sleep apnea in pregnant rats. We hypothesized that late gestational CIH would produce sex- and age-specific social, anxiety-like, repetitive, and cognitive impairments in offspring. METHODS Timed pregnant Long-Evans rats were exposed to CIH or room air normoxia from GD 15-19. Behavioral testing of offspring occurred during either puberty or young adulthood. To examine gestational hypoxia-induced behavioral phenotypes, we quantified hippocampal-associated behaviors (social function, repetitive behaviors, anxiety-like behaviors, and spatial memory and learning), hippocampal neuronal activity (glutamatergic NMDA receptors, dopamine transporter, monoamine oxidase-A, early growth response protein 1, and doublecortin), and circulating hormones in offspring. RESULTS Late gestational CIH induced sex- and age-specific differences in social, repetitive, and memory functions in offspring. In female pubertal offspring, CIH impaired social function, increased repetitive behaviors, and elevated circulating corticosterone levels but did not impact memory. In contrast, CIH transiently induced spatial memory dysfunction in pubertal male offspring but did not impact social or repetitive functions. Long-term effects of gestational CIH on social behaviors were only observed in female offspring, wherein CIH induced social disengagement and suppression of circulating corticosterone levels in young adulthood. No effects of gestational CIH were observed in anxiety-like behaviors, hippocampal neuronal activity, or circulating testosterone and estradiol levels, regardless of sex or age of offspring. CONCLUSIONS Our results indicate that hypoxia-associated pregnancy complications during late gestation can increase the risk for behavioral and physiological outcomes in offspring, such as social dysfunction, repetitive behaviors, and cognitive impairment, that are dependent on sex and age.
Collapse
Affiliation(s)
- Steve Mabry
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - E Nicole Wilson
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Jessica L Bradshaw
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Jennifer J Gardner
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Oluwadarasimi Fadeyibi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Edward Vera
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Oluwatobiloba Osikoya
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Spencer C Cushen
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Dimitrios Karamichos
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
- Department of Pharmacology and Neuroscience, University of North Texas Health Science, Fort Worth, TX, 76107, USA
| | - Styliani Goulopoulou
- Departments of Basic Sciences, Gynecology and Obstetrics, Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA.
| |
Collapse
|
7
|
Unger CA, Aladhami AK, Hope MC, Cotham WE, Nettles KW, Clegg DJ, Velázquez KT, Enos RT. Skeletal Muscle Endogenous Estrogen Production Ameliorates the Metabolic Consequences of a High-Fat Diet in Male Mice. Endocrinology 2023; 164:bqad105. [PMID: 37421340 PMCID: PMC10368313 DOI: 10.1210/endocr/bqad105] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/20/2023] [Accepted: 07/06/2023] [Indexed: 07/10/2023]
Abstract
AIMS The role of skeletal muscle estrogen and its ability to mitigate the negative impact of a high-fat diet (HFD) on obesity-associated metabolic impairments is unknown. To address this, we developed a novel mouse model to determine the role of endogenous 17β-estradiol (E2) production in males in skeletal muscle via inducible, skeletal muscle-specific aromatase overexpression (SkM-Arom↑). METHODS Male SkM-Arom↑ mice and littermate controls were fed a HFD for 14 weeks prior to induction of SkM-Arom↑ for a period of 6.5 weeks. Glucose tolerance, insulin action, adipose tissue inflammation, and body composition were assessed. Indirect calorimetry and behavioral phenotyping experiments were performed using metabolic cages. Liquid chromatography mass spectrometry was used to determine circulating and tissue (skeletal muscle, hepatic, and adipose) E2 and testosterone concentrations. RESULTS SkM-Arom↑ significantly increased E2 in skeletal muscle, circulation, the liver, and adipose tissue. SkM-Arom↑ ameliorated HFD-induced hyperglycemia, hyperinsulinemia, impaired glucose tolerance, adipose tissue inflammation, and reduced hepatic lipid accumulation while eliciting skeletal muscle hypertrophy. CONCLUSION Enhanced skeletal muscle aromatase activity in male mice induces weight loss, improves metabolic and inflammatory outcomes and mitigates the negative effects of a HFD. Additionally, our data demonstrate for the first time skeletal muscle E2 has anabolic effects on the musculoskeletal system.
Collapse
Affiliation(s)
- Christian A Unger
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29209, USA
| | - Ahmed K Aladhami
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29209, USA
- University of Baghdad, Nursing College, Baghdad, Iraq
| | - Marion C Hope
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29209, USA
| | - William E Cotham
- Department of Chemistry and Biochemistry, College of Arts and Science, University of South Carolina, Columbia, SC 29209, USA
| | - Kendall W Nettles
- Department of Integrative Structural and Computational Biology, Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL 33458, USA
| | - Deborah J Clegg
- Department of Internal Medicine, Texas Tech Health Sciences Center, El Paso, TX 79905, USA
| | - Kandy T Velázquez
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29209, USA
| | - Reilly T Enos
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29209, USA
| |
Collapse
|
8
|
Mabry S, Wilson EN, Bradshaw JL, Gardner JJ, Fadeyibi O, Vera E, Osikoya O, Cushen SC, Karamichos D, Goulopoulou S, Cunningham RL. Sex and age differences in social and cognitive function in offspring exposed to late gestational hypoxia. RESEARCH SQUARE 2023:rs.3.rs-2507737. [PMID: 37333114 PMCID: PMC10275064 DOI: 10.21203/rs.3.rs-2507737/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Background Gestational sleep apnea affects 8-26% of pregnancies and can increase the risk for autism spectrum disorder (ASD) in offspring. ASD is a neurodevelopmental disorder associated with social dysfunction, repetitive behaviors, anxiety, and cognitive impairment. To examine the relationship between gestational sleep apnea and ASD-associated behaviors, we used a chronic intermittent hypoxia (CIH) protocol between gestational days (GD) 15-19 in pregnant rats to model late gestational sleep apnea. We hypothesized that late gestational CIH would produce sex- and age-specific social, mood, and cognitive impairments in offspring. Methods Timed pregnant Long-Evans rats were exposed to CIH or room air normoxia from GD 15-19. Behavioral testing of offspring occurred during either puberty or young adulthood. To examine ASD-associated phenotypes, we quantified ASD-associated behaviors (social function, repetitive behaviors, anxiety-like behaviors, and spatial memory and learning), hippocampal activity (glutamatergic NMDA receptors, dopamine transporter, monoamine oxidase-A, EGR-1, and doublecortin), and circulating hormones in offspring. Results Late gestational CIH induced sex- and age-specific differences in social, repetitive and memory functions in offspring. These effects were mostly transient and present during puberty. In female pubertal offspring, CIH impaired social function, increased repetitive behaviors, and increased circulating corticosterone levels, but did not impact memory. In contrast, CIH transiently induced spatial memory dysfunction in pubertal male offspring but did not impact social or repetitive functions. Long-term effects of gestational CIH were only observed in female offspring, wherein CIH induced social disengagement and suppression of circulating corticosterone levels in young adulthood. No effects of gestational CIH were observed on anxiety-like behaviors, hippocampal activity, circulating testosterone levels, or circulating estradiol levels, regardless of sex or age of offspring. Conclusions Our results indicate that hypoxia-associated pregnancy complications during late gestation can increase the risk for ASD-associated behavioral and physiological outcomes, such as pubertal social dysfunction, corticosterone dysregulation, and memory impairments.
Collapse
Affiliation(s)
- Steve Mabry
- UNTHSC: University of North Texas Health Science Center
| | | | | | | | | | - Edward Vera
- UNTHSC: University of North Texas Health Science Center
| | | | | | | | | | | |
Collapse
|
9
|
Allegra S, Chiara F, Di Grazia D, Gaspari M, De Francia S. Evaluation of Sex Differences in Preclinical Pharmacology Research: How Far Is Left to Go? Pharmaceuticals (Basel) 2023; 16:786. [PMID: 37375734 DOI: 10.3390/ph16060786] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/10/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Until the last quarter of the 20th century, sex was not recognized as a variable in health research, nor was it believed to be a factor that could affect health and illness. Researchers preferred studying male models for a variety of reasons, such as simplicity, lower costs, hormone confounding effects, and fear of liability from perinatal exposure in case of pregnancy. Equitable representation is imperative for determining the safety, effectiveness, and tolerance of therapeutic agents for all consumers. Decades of female models' underrepresentation in preclinical studies has resulted in inequality in the understanding, diagnosis, and treatment of disease between the sexes. Sex bias has been highlighted as one of the contributing factors to the poor translation and replicability of preclinical research. There have been multiple calls for action, and the inclusion of sex as a biological variable is increasingly supported. However, although there has been substantial progress in the efforts to include more female models in preclinical studies, disparities today remain. In the present review, we consider the current standard practice of the preclinical research setting, why the sex bias exists, why there is the need to include female models, and what risks may arise from continuing this exclusion from experimental design.
Collapse
Affiliation(s)
- Sarah Allegra
- Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
| | - Francesco Chiara
- Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
| | - Daniela Di Grazia
- Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
| | - Marco Gaspari
- Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
| | - Silvia De Francia
- Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
| |
Collapse
|
10
|
Fang Y, Medina D, Stockwell R, McFadden S, Hascup ER, Hascup KN, Bartke A. Resistance to mild cold stress is greater in both wild-type and long-lived GHR-KO female mice. GeroScience 2023; 45:1081-1093. [PMID: 36527583 PMCID: PMC9886789 DOI: 10.1007/s11357-022-00706-0] [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: 09/08/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Adapting to stress, including cold environmental temperature (eT), is crucial for the survival of mammals, especially small rodents. Long-lived mutant mice have enhanced stress resistance against oxidative and non-oxidative challenges. However, much less is known about the response of those long-lived mice to cold stress. Growth hormone receptor knockout (GHR-KO) mice are long-lived with reduced growth hormone signaling. We wanted to test whether GHR-KO mice have enhanced resistance to cold stress. To examine the response of GHR-KO mice to cold eT, GHR-KO mice were housed at mild cold eT (16 °C) immediately following weaning. Longevity results showed that female GHR-KO and wild-type (WT) mice retained similar lifespan, while both male GHR-KO and WT mice had shortened lifespan compared to the mice housed at 23 °C eT. Female GHR-KO and WT mice housed at 16 °C had upregulated fibroblast growth factor 21 (FGF21), enhanced energy metabolism, reduced plasma triglycerides, and increased mRNA expression of some xenobiotic enzymes compared to females housed at 23 °C and male GHR-KO and WT mice housed under the same condition. In contrast, male GHR-KO and WT mice housed at 16 °C showed deleterious effects in parameters which might be associated with their shortened longevity compared to male GHR-KO and WT mice housed at 23 °C. Together, this study suggests that in response to mild cold stress, sex plays a pivotal role in the regulation of longevity, and female GHR-KO and WT mice are more resistant to this challenge than the males.
Collapse
Affiliation(s)
- Yimin Fang
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA.
| | - David Medina
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Robert Stockwell
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Samuel McFadden
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Erin R Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Kevin N Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| |
Collapse
|
11
|
El Kharraz S, Dubois V, Launonen KM, Helminen L, Palvimo JJ, Libert C, Smeets E, Moris L, Eerlings R, Vanderschueren D, Helsen C, Claessens F. N/C Interactions Are Dispensable for Normal In Vivo Functioning of the Androgen Receptor in Male Mice. Endocrinology 2022; 163:6652495. [PMID: 35908178 PMCID: PMC9756762 DOI: 10.1210/endocr/bqac104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Indexed: 11/19/2022]
Abstract
The androgen receptor (AR) plays a central role in the development and maintenance of the male phenotype. The binding of androgens to the receptor induces interactions between the carboxyterminal ligand-binding domain and the highly conserved 23FQNLF27 motif in the aminoterminal domain. The role of these so-called N/C interactions in AR functioning is debated. In vitro assays show that mutating the AR in the 23FQNLF27 motif (called ARNoC) attenuates the AR transactivation of reporter genes, has no effect on ligand binding, but does affect protein-protein interactions with several AR coregulators. To test the in vivo relevance of the N/C interaction, we analyzed the consequences of the genomic introduction of the ARNoC mutation in mice. Surprisingly, the ARNoC/Y mice show a normal male development, with unaffected male anogenital distance and normal accessory sex glands, male circulating androgen levels, body composition, and fertility. The responsiveness of androgen target genes in kidney, prostate, and testes was also unaffected. We thus conclude that the N/C interactions in the AR are not essential for the development of a male phenotype under normal physiological conditions.
Collapse
Affiliation(s)
- Sarah El Kharraz
- Correspondence: Frank Claessens, PhD, Department of Cellular and Molecular Medicine, Molecular Endocrinology Laboratory, KU Leuven, Leuven, 3000, Belgium. . Reprint requests can be sent to or
| | - Vanessa Dubois
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, 3000, Belgium
- Department of Basic and Applied Medical Sciences, Basic and Translational Endocrinology, Ghent University, Ghent, 9000, Belgium
| | - Kaisa-Mari Launonen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, 70210, Finland
| | - Laura Helminen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, 70210, Finland
| | - Jorma J Palvimo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, 70210, Finland
| | - Claude Libert
- VIB Center for Inflammation Research, VIB, Ghent, 9052, Belgium
- Department for Biomedical Molecular Biology, Ghent University, Ghent, 9052, Belgium
| | - Elien Smeets
- Department of Cellular and Molecular Medicine, Molecular Endocrinology Laboratory, KU Leuven, Leuven, 3000, Belgium
| | - Lisa Moris
- Department of Cellular and Molecular Medicine, Molecular Endocrinology Laboratory, KU Leuven, Leuven, 3000, Belgium
| | - Roy Eerlings
- Department of Cellular and Molecular Medicine, Molecular Endocrinology Laboratory, KU Leuven, Leuven, 3000, Belgium
| | - Dirk Vanderschueren
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, 3000, Belgium
| | - Christine Helsen
- Department of Cellular and Molecular Medicine, Molecular Endocrinology Laboratory, KU Leuven, Leuven, 3000, Belgium
| | - Frank Claessens
- Correspondence: Frank Claessens, PhD, Department of Cellular and Molecular Medicine, Molecular Endocrinology Laboratory, KU Leuven, Leuven, 3000, Belgium. . Reprint requests can be sent to or
| |
Collapse
|
12
|
Lulic-Kuryllo T, Greig Inglis J. Sex differences in motor unit behaviour: A review. J Electromyogr Kinesiol 2022; 66:102689. [PMID: 36095969 DOI: 10.1016/j.jelekin.2022.102689] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 10/15/2022] Open
|
13
|
Chu L, Liu W, Deng J, Wu Y, Yang H, Wang W, Hussain A, Li N, Zhou D, Deng H. Age-related changes in endogenous glucocorticoids, gonadal steroids, endocannabinoids and their ratios in plasma and hair from the male C57BL/6 mice. Gen Comp Endocrinol 2021; 301:113651. [PMID: 33122035 DOI: 10.1016/j.ygcen.2020.113651] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/27/2020] [Accepted: 10/19/2020] [Indexed: 01/08/2023]
Abstract
Age-related level changes of hormones, endocannabinoids and their ratios are of pathophysiological significance for understanding functions, activities and interactions of the endocrine systems, including the hypothalamic-pituitaryadrenal (HPA), hypothalamic-pituitary-gonadal (HPG) axes and endogenous cannabinoid system (ECS). The present study aimed to investigate the age-dependent fluctuations of glucocorticoids, gonadal steroids, endocannabinoids and their ratios from 21 days to 10 months in both plasma and hair from the male C57BL/6 mice. A novel framework based on the liquid chromatography-tandem mass spectrometry was developed to simultaneously determine ten hormones and two endocannabinoids in plasma and hair. Results showed that glucocorticoids, corticosterone (CORT), aldosterone (ALD), 11-dehydrocorticosterone (11-DHC), gonadal steroids, progesterone (P), dehydroepiandrosterone (DHEA), testosterone (T) and dihydrotestosterone (DHT) in plasma were unimodally fluctuated (ps < 0.001) along age with the maximum value at 2.7-month-old. In contrast, the other two gonadal steroids, estrone (E1) and estradiol (E2) were declined with age (ps < 0.001). Differently, endocannabinoids, N-arachidonoyl-ethanolamine (AEA) and 1-arachydonoyl glycerol (1-AG) showed nadir and zenith values at 2.7-month-old and 3.4-month-old, respectively (ps < 0.001). Additionally, the ratios of CORT to 11-DHC and ALD in plasma were dropped similarly with age (ps < 0.001). The ratios of 1-AG to AEA, and of T to A4 and DHT, and of DHEA to A4 were unimodally changed (ps < 0.001) along age with maximum value at 2.7- or 3.4-month-old. In contrast, the ratios of E2 to T and E1 to A4 were decreased with age (ps < 0.05). The rest six ratios that reflected the interactions among the three endocrine systems, were similar age-dependent and showed nadir and zenith values at 2.7-month-old and 3.4-month-old, respectively (ps < 0.05). Most importantly, these findings in light of age-related changing patterns in plasma were repeated in hair, suggesting that the fi41-ndings in the two matrices were mutually validated. However, it was worth noting that their magnitude of levels in the two bio-matrices were markedly different. The current findings could provide reliable hormone and endocannabinoid signatures with age on neuroendocrine profiles as well as their ratios for the male C57BL/6 mice.
Collapse
Affiliation(s)
- Liuxi Chu
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210096, PR China; Key Laboratory of Child Development and Learning Science (Southeast University), Ministry of Education, Nanjing 210096, PR China; Institute of Child Development and Education, Research Center for Learning Science, Southeast University, Nanjing 210096, PR China
| | - Wenhua Liu
- Institute of Life Sciences, Southeast University, Nanjing 210096, PR China
| | - Jia Deng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yan Wu
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210096, PR China; Key Laboratory of Child Development and Learning Science (Southeast University), Ministry of Education, Nanjing 210096, PR China; Institute of Child Development and Education, Research Center for Learning Science, Southeast University, Nanjing 210096, PR China
| | - Haoran Yang
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210096, PR China; Key Laboratory of Child Development and Learning Science (Southeast University), Ministry of Education, Nanjing 210096, PR China; Institute of Child Development and Education, Research Center for Learning Science, Southeast University, Nanjing 210096, PR China
| | - Wei Wang
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210096, PR China; Key Laboratory of Child Development and Learning Science (Southeast University), Ministry of Education, Nanjing 210096, PR China; Institute of Child Development and Education, Research Center for Learning Science, Southeast University, Nanjing 210096, PR China
| | - Ahad Hussain
- Key Laboratory of Child Development and Learning Science (Southeast University), Ministry of Education, Nanjing 210096, PR China; School of Public Health, Southeast University, Nanjing 210096, PR China
| | - Na Li
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210096, PR China; Key Laboratory of Child Development and Learning Science (Southeast University), Ministry of Education, Nanjing 210096, PR China; Institute of Child Development and Education, Research Center for Learning Science, Southeast University, Nanjing 210096, PR China
| | - Dongrui Zhou
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210096, PR China; Key Laboratory of Child Development and Learning Science (Southeast University), Ministry of Education, Nanjing 210096, PR China; Institute of Child Development and Education, Research Center for Learning Science, Southeast University, Nanjing 210096, PR China
| | - Huihua Deng
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210096, PR China; Key Laboratory of Child Development and Learning Science (Southeast University), Ministry of Education, Nanjing 210096, PR China; Institute of Child Development and Education, Research Center for Learning Science, Southeast University, Nanjing 210096, PR China.
| |
Collapse
|
14
|
Zhang KJ, Ramdev RA, Tuta NJ, Spritzer MD. Dose-dependent effects of testosterone on spatial learning strategies and brain-derived neurotrophic factor in male rats. Psychoneuroendocrinology 2020; 121:104850. [PMID: 32892065 PMCID: PMC7572628 DOI: 10.1016/j.psyneuen.2020.104850] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/15/2022]
Abstract
Studies suggest that males outperform females on some spatial tasks. This may be due to the effects of sex steroids on spatial strategy preferences. Past experiments with male rats have demonstrated that low doses of testosterone bias them toward a response strategy, whereas high doses of testosterone bias them toward a place strategy. We investigated the effect of different testosterone doses on the ability of male rats to effectively employ these two spatial learning strategies. Furthermore, we quantified concentrations of brain-derived neurotrophic factor (pro-, mature-, and total BDNF) in the prefrontal cortex, hippocampus, and striatum. All rats were bilaterally castrated and assigned to one of three daily injection doses of testosterone propionate (0.125, 0.250, or 0.500 mg/rat) or a control injection of the drug vehicle. Using a plus-maze protocol, we found that a lower testosterone dose (0.125 mg) significantly improved rats' performance on a response task, whereas a higher testosterone dose (0.500 mg) significantly improved rats' performance on a place task. In addition, we found that a low dose of testosterone (0.125 mg) increased total BDNF in the striatum, while a high dose (0.500 mg) increased total BDNF in the hippocampus. Taken altogether, these results suggest that high and low levels of testosterone enhance performance on place and response spatial tasks, respectively, and this effect is associated with changes in BDNF levels within relevant brain regions.
Collapse
Affiliation(s)
- Kevin J. Zhang
- Department of Biology, Middlebury College, Middlebury, VT 05753, U.S.A
| | - Rajan A. Ramdev
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, U.S.A
| | - Nicholas J. Tuta
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, U.S.A
| | - Mark D. Spritzer
- Department of Biology, Middlebury College, Middlebury, VT 05753, U.S.A.,Program in Neuroscience, Middlebury College, Middlebury, VT 05753, U.S.A.,Corresponding author: Mark Spritzer, Department of Biology, McCardell Bicentennial Hall, Middlebury College, Middlebury, VT 05753, USA, phone: 802-443-5676, FAX: 802-443-2072,
| |
Collapse
|
15
|
Bonacina E, Negri G, Mattiello S, Gabai G, Groppetti D. Deslorelin subcutaneous implants in Oryx dammah males for reproductive control. Theriogenology 2020; 149:72-78. [PMID: 32247215 DOI: 10.1016/j.theriogenology.2020.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 02/13/2020] [Accepted: 03/09/2020] [Indexed: 11/26/2022]
Abstract
The aim of this study was to assess the effects of the deslorelin subcutaneous implant as a temporary contraceptive method in the Oryx dammah male. For this purpose, deslorelin at different doses, i.e. 14.1 mg and 9.4 mg, was subcutaneously implanted in three males (Phase 1) and one male (Phase 2) adult Oryx dammah, respectively. Quantitative behavior evaluation and androgen concentrations in feces and plasma were assessed before and after implant application. Fecal androgen concentrations observed in treated males were compared with those measured in one orchiectomized male and two females. Fecal androgen concentrations increased up to 15 days after the implant application, then progressively decreased, reaching the basal level at day150 in Phase 1. In Phase 2, levels remained high until day 60 and returned to basal level on day 120. Plasma testosterone concentration was higher on the day of implant application than three months later, but with variable ranges among males. A general increase of activity levels and hierarchical changes were observed after treatment, in accordance with hormonal variations. Despite males cohabiting with two fertile females during the observation period, no births were recorded. However, between the end of Phase 1 and the beginning of Phase 2, i.e. about 10-11 months after the first deslorelin implant, a fertile mating occurred leading to the birth of a calf. Therefore, we can hypothesize a contraceptive effect up to 10 months after the implant. Testicular histology performed on one male at the end of the Phase 2 showed no spermatogenetic activity. Our results suggest that deslorelin implant can be used to temporarily control reproduction in the Oryx dammah male. Behavior and fecal androgen measurements were useful and repeatable, non-invasive methods to monitor response.
Collapse
Affiliation(s)
| | - Gabriela Negri
- Department of Veterinary Medicine, Università Degli Studi di Milano, Via Celoria 10, 20133, Milan, Italy
| | - Silvana Mattiello
- Department of Agricultural and Environmental Sciences - Production, Landscape, Agroenergy, Università Degli Studi di Milano, Via Celoria 2, 20133, Milan, Italy
| | - Gianfranco Gabai
- Department of Comparative Biomedicine and Food Science (BCA), University of Padova, Via Dell'Università 16, 35020, Legnaro, PD, Italy
| | - Debora Groppetti
- Department of Veterinary Medicine, Università Degli Studi di Milano, Via Celoria 10, 20133, Milan, Italy.
| |
Collapse
|
16
|
Gale TJ, Garratt M, Brooks RC. Female mice seek refuge from castrated males, but not intact or vasectomized males, mitigating a socially-induced glucocorticoid response. Physiol Behav 2019; 211:112678. [PMID: 31505190 DOI: 10.1016/j.physbeh.2019.112678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 08/07/2019] [Accepted: 09/05/2019] [Indexed: 10/26/2022]
Abstract
Sexual conflict may be manifested during social interactions, shaping the costs of reproduction in sexually reproducing species. This conflict, and the physical necessity of intromission, can intensify the already costly nature of reproduction for female mammals. To identify and partition the costs that males inflict on females during mating and reproduction, we paired female mice with either other females or castrated, vasectomised, or intact (sham-vasectomised) males, thus manipulating exposure to social mating behavior and costs arising from fertilization. We also provided females with refuges where males could not enter, to test whether females show avoidance or attraction to males of different gonadal status expected to exhibit different levels of social behavior. We found that females paired with vasectomised and castrated males spent the most time in their refuge. Females housed with castrated males also had increased glucocorticoid levels, an effect that was mitigated when females could retreat from these males to a refuge. This suggests that females actively refuge from castrated males, and that housing with such males is sufficient to generate an increased glucocorticoid response. Our results show that females choose to refuge from males depending on the partner's gonadal status, choices that are linked to social induced stress responses but not exposure to male mating behaviour.
Collapse
Affiliation(s)
- Teagan J Gale
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences (BEES), the University of New South Wales, High Street, Kensington, NSW 2052, Australia.
| | - Michael Garratt
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences (BEES), the University of New South Wales, High Street, Kensington, NSW 2052, Australia; Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Robert C Brooks
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences (BEES), the University of New South Wales, High Street, Kensington, NSW 2052, Australia
| |
Collapse
|
17
|
Bedi Y, Chang RC, Gibbs R, Clement TM, Golding MC. Alterations in sperm-inherited noncoding RNAs associate with late-term fetal growth restriction induced by preconception paternal alcohol use. Reprod Toxicol 2019; 87:11-20. [PMID: 31051257 DOI: 10.1016/j.reprotox.2019.04.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/03/2019] [Accepted: 04/25/2019] [Indexed: 12/27/2022]
Abstract
Using a mouse model, our group recently described an association between chronic paternal alcohol use prior to conception and deficits in offspring growth. Here, we sought to determine the impact of alcohol exposure on male reproductive physiology and the association of sperm-inherited noncoding RNAs with the transmission of the observed growth defects. Alcohol exposure did not appreciably alter male reproductive physiology or fertility. However, chronic alcohol use reproducibly induced late-term fetal growth restriction in the offspring, which correlated with a shift in the proportional ratio of transfer RNA-derived small RNAs to Piwi-interacting RNAs, as well as altered enrichment of microRNAs miR21, miR30, and miR142 in alcohol-exposed sperm. Although our dataset share similarities to prior works examining the impact of paternal stress on offspring phenotype, we were unable to identify any changes in plasma corticosterone, indicating alcohol may alter sperm-inherited noncoding RNAs through distinct mechanisms.
Collapse
Affiliation(s)
- Yudhishtar Bedi
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University College Station, Texas, 77843, USA
| | - Richard C Chang
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University College Station, Texas, 77843, USA
| | - Rachel Gibbs
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University College Station, Texas, 77843, USA
| | - Tracy M Clement
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University College Station, Texas, 77843, USA
| | - Michael C Golding
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University College Station, Texas, 77843, USA.
| |
Collapse
|
18
|
Wagner BA, Braddick VC, Batson CG, Cullen BH, Miller LE, Spritzer MD. Effects of testosterone dose on spatial memory among castrated adult male rats. Psychoneuroendocrinology 2018; 89:120-130. [PMID: 29414025 PMCID: PMC5878712 DOI: 10.1016/j.psyneuen.2017.12.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 12/22/2017] [Accepted: 12/28/2017] [Indexed: 12/20/2022]
Abstract
Previous research on the activational effects of testosterone on spatial memory has produced mixed results, possibly because such effects are dose-dependent. We tested a wide range of testosterone doses using two spatial memory tasks: a working-reference memory version of the radial-arm maze (RAM) and an object location memory task (OLMT). Adult male Sprague-Dawley rats were castrated or sham-castrated and given daily injections of drug vehicle (Oil Sham and Oil GDX) or one of four doses of testosterone propionate (0.125, 0.250, 0.500, and 1.000 mg T) beginning seven days before the first day of behavioral tests and continuing throughout testing. For the RAM, four arms of the maze were consistently baited on each day of testing. Testosterone had a significant effect on working memory on the RAM, with the Oil Sham, 0.125 mg T, and 0.500 mg T groups performing better than the Oil GDX group. In contrast, there was no significant effect of testosterone on spatial reference memory on the RAM. For the OLMT, we tested long-term memory using a 2 h inter-trial interval between first exposure to two identical objects and re-exposure after one object had been moved. Only the 0.125 and 0.500 mg T groups showed a significant increase in exploration of the moved object during the testing trials, indicating better memory than all other groups. Testosterone replacement restored spatial memory among castrated male rats on both behavioral tasks, but there was a complex dose-response relationship; therefore, the therapeutic value of testosterone is likely sensitive to dose.
Collapse
Affiliation(s)
- Benjamin A. Wagner
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, U.S.A
| | | | | | - Brendan H. Cullen
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, U.S.A
| | - L. Erin Miller
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, U.S.A
| | - Mark D. Spritzer
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, U.S.A,Department of Biology, Middlebury College, Middlebury, VT 05753, U.S.A,Corresponding author: Mark Spritzer, Department of Biology, McCardell Bicentennial Hall, Middlebury College, Middlebury, VT 05753, USA, phone: 802-443-5676, FAX: 802-443-2072
| |
Collapse
|
19
|
Langhammer M, Michaelis M, Hartmann MF, Wudy SA, Sobczak A, Nürnberg G, Reinsch N, Schön J, Weitzel JM. Reproductive performance primarily depends on the female genotype in a two-factorial breeding experiment using high-fertility mouse lines. Reproduction 2017; 153:361-368. [PMID: 28096494 DOI: 10.1530/rep-16-0434] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 11/22/2016] [Accepted: 12/19/2016] [Indexed: 11/08/2022]
Abstract
Mouse models showing an improved fertility phenotype are barely described in the literature. In the present study, we further characterized two outbred mouse models that have been selected for the phenotype 'high fertility' for more than 177 generations (fertility lines (FL) 1 and 2). In order to delineate the impact of males and females on fertility parameters, we performed a two-factorial breeding experiment by mating males and females of the three different genotypes (FL1, FL2, unselected control (Ctrl)) in all 9 possible combinations. Reproductive performance, such as number of offspring per litter or total birth weight of the entire pup, mainly depends on the female genotype. Although the reproductive performance of FL1 and FL2 is very similar, their phenotypes differ. FL2 animals of both genders are larger compared to FL1 and control animals. Females of the control line delivered offspring earlier compared to FL1 and FL2 dams. Males of FL1 are the lightest and the only ones who gained weight during the two weeks mating period. To address whether this effect is correlated with differing serum androgen levels, we measured the concentrations of testosterone, dehydroepiandrosterone, 4-androstenedione, androstanediol and dihydrotestosterone in males of all three lines by GC-MS. We measured serum testosterone between 5.0 and 6.4 ng/mL, whereas the concentrations of the other androgens were at least one order of magnitude lower, with no significant differences between the lines. Our data indicate that reproductive outcome largely depends on the genotype of the female in a two-factorial breeding experiment and supports previous findings that the phenotype 'high fertility' is warranted by using different physiological strategies.
Collapse
Affiliation(s)
- Martina Langhammer
- Institute of Genetics and BiometryLeibniz Institute for Farm Animal Biology, FBN, Dummerstorf, Mecklenburg-Vorpommern, Germany
| | - Marten Michaelis
- Institute of Reproductive BiologyLeibniz Institute for Farm Animal Biology, FBN, Dummerstorf, Mecklenburg-Vorpommern, Germany
| | - Michaela F Hartmann
- Steroid Research & Mass Spectrometry UnitLaboratory for Translational Hormone Analytics, Paediatric Endocrinology & Diabetology, Center for Child and Adolescent Medicine, Justus Liebig University, Giessen, Hessen, Germany
| | - Stefan A Wudy
- Steroid Research & Mass Spectrometry UnitLaboratory for Translational Hormone Analytics, Paediatric Endocrinology & Diabetology, Center for Child and Adolescent Medicine, Justus Liebig University, Giessen, Hessen, Germany
| | - Alexander Sobczak
- Institute of Reproductive BiologyLeibniz Institute for Farm Animal Biology, FBN, Dummerstorf, Mecklenburg-Vorpommern, Germany
| | - Gerd Nürnberg
- Institute of Genetics and BiometryLeibniz Institute for Farm Animal Biology, FBN, Dummerstorf, Mecklenburg-Vorpommern, Germany
| | - Norbert Reinsch
- Institute of Genetics and BiometryLeibniz Institute for Farm Animal Biology, FBN, Dummerstorf, Mecklenburg-Vorpommern, Germany
| | - Jennifer Schön
- Institute of Reproductive BiologyLeibniz Institute for Farm Animal Biology, FBN, Dummerstorf, Mecklenburg-Vorpommern, Germany
| | - Joachim M Weitzel
- Institute of Reproductive BiologyLeibniz Institute for Farm Animal Biology, FBN, Dummerstorf, Mecklenburg-Vorpommern, Germany
| |
Collapse
|
20
|
Gonadal hormones affect alcohol drinking, but not cue+yohimbine-induced alcohol seeking, in male and female rats. Physiol Behav 2017; 203:70-80. [PMID: 29106989 DOI: 10.1016/j.physbeh.2017.10.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/18/2017] [Accepted: 10/23/2017] [Indexed: 11/22/2022]
Abstract
Alcohol use disorder (AUD) is a chronic, relapsing disease characterized by maladaptive patterns of alcohol drinking and seeking. Though sex differences exist in the etiology of AUD, much remains to be elucidated concerning the mechanisms underlying sex-related vulnerability to developing excessive alcohol-motivated behavior. While a large body of evidence points to an important role of circulating gonadal hormones in mediating cocaine reinforcement, findings are less consistent with respect to ethanol. Critically, the effects of gonadal hormones on the reinstatement of ethanol seeking, a model of "craving"-like behavior that reveals pronounced sex differences, has not yet been examined. Thus, the goal of the present experiment was to directly compare manipulations of gonadal hormones in male and female rats on ethanol-motivated behavior. Rats received sham or gonadectomy surgery with or without hormone replacement prior to and throughout three weeks of operant ethanol self-administration to determine the effects of chronically high or low gonadal hormone levels on ethanol drinking. Hormone treatment ceased during extinction training, and the effects of an acute injection of either testosterone (in males) or estradiol (in females) on cue+yohimbine-induced reinstatement of ethanol seeking was determined. Separate groups of gonadally-intact female rats went through similar training, but the effects of either the antiestrogen, fulvestrant, the selective estrogen receptor modulator, clomiphene, or the estrogen receptor β antagonist, PHTPP, on the reinstatement of ethanol seeking were determined. Chronic estradiol replacement produced significant increases in ethanol drinking in female rats, while chronic testosterone significantly decreased ethanol drinking in male rats. Gonadectomy alone only produced modest shifts in drinking towards the opposite-sex pattern, and did not eliminate the robust sex differences that persisted regardless of hormone manipulations. Neither prior chronic nor acute hormone manipulations altered cue+yohimbine-induced reinstatement of ethanol seeking, though blockade of estrogen receptors tended to reduce reinstatement in gonadally-intact females. Overall, our findings indicate that gonadal hormones at least partially mediate, but do not totally account for the sex differences evident in ethanol self-administration, and circulating gonadal hormones have little effect on the reinstatement of ethanol seeking. These results provide a foundation for future studies examining the neuronal mechanisms underlying sex differences in ethanol drinking and seeking.
Collapse
|
21
|
Jardí F, Laurent MR, Dubois V, Khalil R, Deboel L, Schollaert D, Van Den Bosch L, Decallonne B, Carmeliet G, Claessens F, Vanderschueren D. A shortened tamoxifen induction scheme to induce CreER recombinase without side effects on the male mouse skeleton. Mol Cell Endocrinol 2017; 452:57-63. [PMID: 28504114 DOI: 10.1016/j.mce.2017.05.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 05/03/2017] [Accepted: 05/09/2017] [Indexed: 12/30/2022]
Abstract
The selective estrogen receptor modulator tamoxifen exerts estrogen agonistic or antagonistic actions on several tissues, including bone. The off-target effects of tamoxifen are one of the most widely recognized pitfalls of tamoxifen-inducible Cre recombinases (CreERs), potentially confounding the phenotypic findings. Still, the validation of tamoxifen induction schemes that minimize the side effects of the drug has not been addressed. Here, we compared the side effects on the skeleton and other androgen-responsive targets of a shortened tamoxifen regimen (2 doses of 190 mg/kg body weight by oral gavage) to a standard protocol (4 doses) and determined their efficiency in inducing CreER-mediated gene deletion. In addition, both a vehicle- and a 10-dose group, which served as a positive control for tamoxifen side effects, were also included. For this purpose, we generated male mice with a floxed androgen receptor (AR) and a neuron-specifically expressed CreER. Treatment with two doses of tamoxifen was the only regimen that did not diminish androgenic bioactivity, as assessed by both seminal vesicles and levator ani/bulbocavernosus muscle weights and serum testosterone concentrations. Similarly, trabecular and cortical femoral bone structure were dramatically altered by both the standard and high-dose protocols but not by the shortened version. Serum osteocalcin and bone-gene expression analyses confirmed the absence of effects on bone by 2 doses of tamoxifen. This protocol decreased AR mRNA levels efficiently and specifically in the nervous system. Thus, we optimized a protocol for tamoxifen-induced CreER gene deletion in mice without off-target effects on bone and male reproductive organs.
Collapse
Affiliation(s)
- Ferran Jardí
- Clinical and Experimental Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 PO Box 902, 3000 Leuven, Belgium
| | - Michaël R Laurent
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 PO Box 901, 3000 Leuven, Belgium; Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, KU Leuven, Herestraat 49 PO Box 7003, Leuven, Belgium
| | - Vanessa Dubois
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 PO Box 901, 3000 Leuven, Belgium; INSERM UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Rougin Khalil
- Clinical and Experimental Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 PO Box 902, 3000 Leuven, Belgium
| | - Ludo Deboel
- Clinical and Experimental Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 PO Box 902, 3000 Leuven, Belgium
| | - Dieter Schollaert
- Clinical and Experimental Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 PO Box 902, 3000 Leuven, Belgium
| | - Ludo Van Den Bosch
- Laboratory of Neurobiology, VIB Center for Brain and Disease Research and KU Leuven, 3000 Leuven, Belgium
| | - Brigitte Decallonne
- Clinical and Experimental Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 PO Box 902, 3000 Leuven, Belgium
| | - Geert Carmeliet
- Clinical and Experimental Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 PO Box 902, 3000 Leuven, Belgium
| | - Frank Claessens
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 PO Box 901, 3000 Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 PO Box 902, 3000 Leuven, Belgium.
| |
Collapse
|
22
|
Progressive effects of silver nanoparticles on hormonal regulation of reproduction in male rats. Toxicol Appl Pharmacol 2016; 313:35-46. [DOI: 10.1016/j.taap.2016.10.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 10/06/2016] [Accepted: 10/11/2016] [Indexed: 02/05/2023]
|
23
|
Rocha JS, Bonkowski MS, França LR, Bartke A. Mild Calorie Restriction Does Not Affect Testosterone Levels and Testicular Gene Expression in Mutant Mice. Exp Biol Med (Maywood) 2016; 232:1050-63. [PMID: 17720951 DOI: 10.3181/0703-rm-52] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The hypothalamic-pituitary-gonadal (HPG) axis and the somatotropic axis are influenced by nutritional factors. Calorie restriction (CR) extends lifespan but suppresses both the HPG and the somatotropic axes. Since most CR studies use a fairly severe (40%–60%) reduction of calorie intake, we hypothesized that a milder CR (20%) might not be deleterious to reproduction in male mice. To test this hypothesis, we evaluated the effects of 20% CR on testicular testosterone content and on testicular expression of genes that are relevant to testicular function and reproductive competence, including insulin-like growth factor-I, cytochrome P450 aromatase (Cyp19a1), androgen receptor, luteinizing hormone receptor, follicle-stimulating hormone receptor, cytochrome P450c17 and 3-β-hydroxysteroid dehydrogenase/isomerase. To relate CR effects to the activity of the somatotropic axis, we have used growth hormone–resistant GHR knockout mice as well as transgenic mice overexpressing GH. Mild CR did not affect testosterone levels in testis homogenates and had little effect on expression of the examined genes in the reproductive organs. Altered activity of the GH/insulin-like growth factor–1 axis had a major impact on the parameters analyzed. The results also suggest that expression of several key genes involved in the control of testicular function is preserved under conditions of mild CR and encourage speculation that mild regimens of CR can produce longevity benefits without impairing reproduction.
Collapse
Affiliation(s)
- Juliana S Rocha
- Laboratory of Cellular Biology, Avenue Antonio Carlos 6627, Pampulha Belo Horizonte, MG 31270-901, Brazil.
| | | | | | | |
Collapse
|
24
|
Barda S, Yogev L, Paz G, Yavetz H, Hauser R, Breitbart H, Kleiman SE. New insights into the role of the Brdt protein in the regulation of development and spermatogenesis in the mouse. Gene Expr Patterns 2016; 20:130-7. [DOI: 10.1016/j.gep.2016.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/11/2016] [Accepted: 03/14/2016] [Indexed: 12/01/2022]
|
25
|
Chapman JC, Chapman FM, Michael SD. The production of alpha/beta and gamma/delta double negative (DN) T-cells and their role in the maintenance of pregnancy. Reprod Biol Endocrinol 2015; 13:73. [PMID: 26164866 PMCID: PMC4499209 DOI: 10.1186/s12958-015-0073-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 07/08/2015] [Indexed: 01/19/2023] Open
Abstract
The ability of the thymus gland to convert bone marrow-derived progenitor cells into single positive (SP) T-cells is well known. In this review we present evidence that the thymus, in addition to producing SP T-cells, also has a pathway for the production of double negative (DN) T-cells. The existence of this pathway was noted during our examination of relevant literature to determine the cause of sex steroid-induced thymocyte loss. In conducting this search our objective was to answer the question of whether thymocyte loss is the end product of a typical interaction between the reproductive and immune systems, or evidence that the two systems are incompatible. We can now report that "thymocyte loss" is a normal process that occurs during the production of DN T-cells. The DN T-cell pathway is unique in that it is mediated by thymic mast cells, and becomes functional following puberty. Sex steroids initiate the development of the pathway by binding to an estrogen receptor alpha located in the outer membrane of the mast cells, causing their activation. This results in their uptake of extracellular calcium, and the production and subsequent release of histamine and serotonin. Lymphatic vessels, located in the subcapsular region of the thymus, respond to the two vasodilators by undergoing a substantial and preferential uptake of gamma/delta and alpha/beta DN T- cells. These T- cells exit the thymus via efferent lymphatic vessels and enter the lymphatic system.The DN pathway is responsible for the production of three subsets of gamma/delta DN T-cells and one subset of alpha/beta DN T-cells. In postpubertal animals approximately 35 % of total thymocytes exit the thymus as DN T-cells, regardless of sex. In pregnant females, their levels undergo a dramatic increase. Gamma/delta DN T-cells produce cytokines that are essential for the maintenance of pregnancy.
Collapse
Affiliation(s)
- John C Chapman
- Department of Biological Sciences, Binghamton University, Binghamton, New York, 13902-6000, USA.
| | - Fae M Chapman
- Department of Biological Sciences, Binghamton University, Binghamton, New York, 13902-6000, USA.
| | - Sandra D Michael
- Department of Biological Sciences, Binghamton University, Binghamton, New York, 13902-6000, USA.
| |
Collapse
|
26
|
Abstract
INTRODUCTION The mouse is an important, though imperfect, organism with which to model human disease and to discover and test novel drugs in a preclinical setting. Many experimental strategies have been used to discover new biological and molecular targets in the mouse, with the hopes of translating these discoveries into novel drugs to treat prostate cancer in humans. Modeling prostate cancer in the mouse, however, has been challenging, and often drugs that work in mice have failed in human trials. AREAS COVERED The authors discuss the similarities and differences between mice and men; the types of mouse models that exist to model prostate cancer; practical questions one must ask when using a mouse as a model; and potential reasons that drugs do not often translate to humans. They also discuss the current value in using mouse models for drug discovery to treat prostate cancer and what needs are still unmet in field. EXPERT OPINION With proper planning and following practical guidelines by the researcher, the mouse is a powerful experimental tool. The field lacks genetically engineered metastatic models, and xenograft models do not allow for the study of the immune system during the metastatic process. There remain several important limitations to discovering and testing novel drugs in mice for eventual human use, but these can often be overcome. Overall, mouse modeling is an essential part of prostate cancer research and drug discovery. Emerging technologies and better and ever-increasing forms of communication are moving the field in a hopeful direction.
Collapse
Affiliation(s)
- Kenneth C Valkenburg
- The Johns Hopkins University, The James Buchanan Brady Urological Institute, Department of Urology , 600 North Wolfe Street, Baltimore, MD 21287 , USA
| | | |
Collapse
|
27
|
La Sala G, Marazziti D, Di Pietro C, Golini E, Matteoni R, Tocchini-Valentini GP. Modulation of Dhh signaling and altered Sertoli cell function in mice lacking the GPR37-prosaposin receptor. FASEB J 2015; 29:2059-69. [PMID: 25609427 DOI: 10.1096/fj.14-269209] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 12/22/2014] [Indexed: 01/10/2023]
Abstract
The mammalian G-protein-coupled receptor 37 (GPR37) is expressed in brain, in adult testis, and during the early phase of gonad differentiation. Somatic Sertoli cells (SCs) are located within the seminiferous tubules where they support the germinal epithelium. An adequate number of SCs is required for the complete prepubertal differentiation of germ cells and adult fertility. This study shows that Gpr37 and its ligand prosaposin are both postnatally expressed by SCs, whose proliferation and maturation are affected in Gpr37-null mutant mice during postnatal testicular development. Mutant pups show a delayed timing in sperm cell development, with a partial arrest of spermatocytes at the meiotic pachytene (e.g., 1.5-fold increase in Gpr37(-/-) P21 pups) and their increased apoptosis (e.g., 1.8-fold and 3.5-fold increase in Gpr37(-/-) P21 and adult mice, respectively). Mutant adults have reduced testis weight (wild type, 299 ± 5 mg; knockout, 258 ± 16 mg; P < 0.05) and epididymal sperm count and motility (e.g., 1.5-fold and 1.45-fold decrease in Gpr37(-/-) mice, respectively). Lack of Gpr37 results in the reduction in androgen receptor levels during prepubertal testis development, alongside the altered expression of SC maturation markers. It also affects the prepubertal testis expression of desert hedgehog (Dhh) mitogenic cascade components (Dhh, 1.3-fold increase in Gpr37(-/-) P10 and P21 pups; Gli2, 1.4-fold and 1.6-fold increase in Gpr37(-/-) P10 and P21 pups, respectively) including patched homolog 1 (1.3-fold increase in Gpr37(-/-) P10 and P21 pups), which is found localized in prepubertal SCs and is associated with Gpr37 in cultured primary SC samples. These results indicate that Gpr37 is a specific modulator of murine testis Dhh mitogenic signaling and SC proliferation and maturation.
Collapse
Affiliation(s)
- Gina La Sala
- Consiglio Nazionale delle Ricerche, Emma-Infrafrontier-Impc, "A. Buzzati-Traverso" Campus, Istituto di Biologia Cellulare e Neurobiologia, Monterotondo Scalo, Rome, Italy
| | - Daniela Marazziti
- Consiglio Nazionale delle Ricerche, Emma-Infrafrontier-Impc, "A. Buzzati-Traverso" Campus, Istituto di Biologia Cellulare e Neurobiologia, Monterotondo Scalo, Rome, Italy
| | - Chiara Di Pietro
- Consiglio Nazionale delle Ricerche, Emma-Infrafrontier-Impc, "A. Buzzati-Traverso" Campus, Istituto di Biologia Cellulare e Neurobiologia, Monterotondo Scalo, Rome, Italy
| | - Elisabetta Golini
- Consiglio Nazionale delle Ricerche, Emma-Infrafrontier-Impc, "A. Buzzati-Traverso" Campus, Istituto di Biologia Cellulare e Neurobiologia, Monterotondo Scalo, Rome, Italy
| | - Rafaele Matteoni
- Consiglio Nazionale delle Ricerche, Emma-Infrafrontier-Impc, "A. Buzzati-Traverso" Campus, Istituto di Biologia Cellulare e Neurobiologia, Monterotondo Scalo, Rome, Italy
| | - Glauco P Tocchini-Valentini
- Consiglio Nazionale delle Ricerche, Emma-Infrafrontier-Impc, "A. Buzzati-Traverso" Campus, Istituto di Biologia Cellulare e Neurobiologia, Monterotondo Scalo, Rome, Italy
| |
Collapse
|
28
|
Effects of crude kerosene on testosterone levels, aggression and toxicity in rat. Toxicol Rep 2014; 2:175-183. [PMID: 28962349 PMCID: PMC5598145 DOI: 10.1016/j.toxrep.2014.11.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 11/20/2014] [Accepted: 11/24/2014] [Indexed: 02/07/2023] Open
Abstract
Kerosene supplementation increases serum testosterone levels in rat. Increased testosterone levels were associated with increased aggression. Kerosene supplementation had varied effects on blood parameters, notably, increased WBC counts. Supplementation resulted in active/chronic gastritis in the stomach of our rat model.
The use of crude kerosene as a dietary supplement in boarding schools has been a common practice in east Africa and other countries for many years, with the belief of it reducing the sex drive (libido) at the pubertal stage. There is however no scientific basis for this belief. The present study aimed at using a rat animal model to investigate the effects of crude kerosene on serum testosterone levels, aggression and its possible toxic effects. Fifteen male albino rats of approximately similar age and average weights were put into three groups of five animals each; the control group (placebo), low kerosene dose (10 μl/day) group and high kerosene dose (300 μl/day) group. ELISA was used to determine the serum testosterone levels. During treatment, changes in aggression were observed and noted. Liver toxicity was determined using enzyme assays, total protein and albumin while renal toxicity was monitored using serum creatinine levels. A full hemogram was conducted to determine hematological effects. Various tissue biopsies were obtained and examined using histopathological techniques for evidence of toxicity. Contrary to the common belief, our findings showed an overall increase of serum testosterone levels of up to 66% in the low dose and 75% in the high dose groups, with an increasing trend by the end of the study. The high dose group showed significantly increased levels of white blood cells (WBC) (p = 0.036), red blood cells (RBC) (p = 0.025), hematocrit (HCT) (p = 0.03), red cell distribution width (p = 0.028) and platelets (p = 0.017). The histological results of the stomach indicated chronic gastritis.
Collapse
Key Words
- ALP, alkaline phosphatase
- ALT, alanine transaminase
- AST, aspartate transaminase
- Aggression
- Bromocresol green (PubChem CID: 6451)
- Creatinine (PubChem CID: 588)
- Crude kerosene
- EDTA, ethylenediaminetetraacetate
- ELISA, enzyme linked immunosorbent assay
- Eosin (PubChem CID: 11048)
- Ethylenediaminetetraacetic acid (PubChem CID: 6049)
- Formaldehyde (PubChem CID: 712)
- Gastritis
- HCT, hematocrit concentration
- Hematoxylin (PubChem CID: 442514)
- LFT, liver function tests
- Picrate (PubChem CID: 62496)
- RBC, red blood cells
- RDW, red cell distribution width
- RFT, renal function tests
- Sex drive
- T, testosterone
- Testosterone
- Testosterone (PubChem CID: 6013)
- Toxicity
- WBC, white blood cell
Collapse
|
29
|
Das RK, Banerjee S, Shapiro BH. Irreversible perinatal imprinting of adult expression of the principal sex-dependent drug-metabolizing enzyme CYP2C11. FASEB J 2014; 28:4111-22. [PMID: 24942648 DOI: 10.1096/fj.13-248864] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 06/02/2014] [Indexed: 01/17/2023]
Abstract
We proposed to determine whether, like other sexual dimorphisms, drug metabolism is permanently imprinted by perinatal hormones, resulting in its irreversible sex-dependent expression. We treated newborn male rats with monosodium glutamate (MSG), a total growth hormone (GH) blocker, and, using cultured hepatocytes, examined expression of adult CYP2C11, the predominant cytochrome-P450 expressed only in males, as well as the signal transduction pathway by which episodic GH solely regulates the isoform's expression. In addition, adolescent hypophysectomized (hypox) male rats served as controls in which GH was eliminated after the critical imprinting period. Whereas renaturalization of the masculine episodic GH profile restored normal male-like levels of CYP2C11, as well as CYP2C12, in hepatocytes from hypox rats, the cells derived from the MSG-treated rats were completely unresponsive. Moreover, GH exposure of hepatocytes from hypox rats resulted in normal induction, activation, nuclear translocation, and binding to the CYP2C11 promoter of the signal transducers mediating GH regulation of CYP2C11 expression, which dramatically contrasted with the complete unresponsiveness of the MSG-derived hepatocytes, also associated with hypermethylation of GH-response elements in the CYP2C11 promoter. Lastly, neonatal MSG treatment had no adverse effect on postnatal and adult testosterone levels. The results demonstrate that the sexually dimorphic expression of CYP2C11 is irreversibly imprinted shortly after birth by a hormone other than the customary testosterone, but likely by GH.
Collapse
Affiliation(s)
- Rajat Kumar Das
- Laboratories of Biochemistry, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Sarmistha Banerjee
- Laboratories of Biochemistry, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Bernard H Shapiro
- Laboratories of Biochemistry, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
30
|
Yurkovetskiy L, Burrows M, Khan AA, Graham L, Volchkov P, Becker L, Antonopoulos D, Umesaki Y, Chervonsky AV. Gender bias in autoimmunity is influenced by microbiota. Immunity 2013; 39:400-12. [PMID: 23973225 DOI: 10.1016/j.immuni.2013.08.013] [Citation(s) in RCA: 684] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 05/24/2013] [Indexed: 12/14/2022]
Abstract
Gender bias and the role of sex hormones in autoimmune diseases are well established. In specific pathogen-free nonobese diabetic (NOD) mice, females have 1.3-4.4 times higher incidence of type 1 diabetes (T1D). Germ-free (GF) mice lost the gender bias (female-to-male ratio 1.1-1.2). Gut microbiota differed in males and females, a trend reversed by male castration, confirming that androgens influence gut microbiota. Colonization of GF NOD mice with defined microbiota revealed that some, but not all, lineages overrepresented in male mice supported a gender bias in T1D. Although protection of males did not correlate with blood androgen concentration, hormone-supported expansion of selected microbial lineages may work as a positive-feedback mechanism contributing to the sexual dimorphism of autoimmune diseases. Gene-expression analysis suggested pathways involved in protection of males from T1D by microbiota. Our results favor a two-signal model of gender bias, in which hormones and microbes together trigger protective pathways.
Collapse
Affiliation(s)
- Leonid Yurkovetskiy
- Committee on Microbiology, The University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
|
32
|
Sedelaar JM, Dalrymple SS, Isaacs JT. Of mice and men--warning: intact versus castrated adult male mice as xenograft hosts are equivalent to hypogonadal versus abiraterone treated aging human males, respectively. Prostate 2013; 73:1316-25. [PMID: 23775398 PMCID: PMC4009979 DOI: 10.1002/pros.22677] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 03/26/2013] [Indexed: 11/07/2022]
Abstract
BACKGROUND Immune deficient male mice bearing human prostate cancer xenografts are used to evaluate therapeutic response to novel androgen ablation approaches and the results compared to surgical castration based upon assumption that testosterone microenvironment in intact and castrated adult male mice mimics eugonadal and castrated aging adult human males. METHODS To test these assumptions, serum total testosterone (TT) and free testosterone (FT) were determined longitudinally in groups (n > 20) of intact versus castrated adult male nude, NOG, and immune competent C57BL/6 mice. RESULTS In adult male mice, TT and FT varies by 30- to 100-fold within the same animal providing a microenvironment that is only equivalent to hypogonadal, not eugonadal, adult human males (TT is 1.7 ± 1.2 ng/ml [5.8 ± 4.1 nM] in nude and 2.5 ± 1.3 ng/ml [8.7 ± 4.4 nM] in NOG mice versus >4.2 ng/ml [14.7 nM] in eugonadal humans). This was confirmed based upon enhanced growth of androgen dependent human prostate cancer xenografts inoculated into mice supplemented with exogenous testosterone to elevate and chronically maintain serum TT at a level (5 ng/ml [18 nM]) equivalent to a 50-year-old eugonadal human male. In castrated mice, TT and FT range from 2 to 20 pg/ml (7-70 pM) and <0.8 pg/ml (<2.6 pM), respectively, which is equivalent to castrate resistant prostate cancer (CRPC) patients treated with abiraterone. This was confirmed based upon the inability of another CYP17A1 inhibitor, ketoconazole, to inhibit the growth of CRPC xenografts in castrated mice. CONCLUSIONS Adult male mice supplemented with testosterone mimic eugonadal human males, while unsupplemented animals mimic standard androgen ablation and castrated animals mimic abiraterone treated patients. These studies confirm what is claimed in Robert Burns' poem "To a Mouse" that "The best laid schemes of mice and men/often go awry.".
Collapse
Affiliation(s)
- J.P. Michiel Sedelaar
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Susan S. Dalrymple
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
- Correspondence to: Sidney Kimmel Comprehensive Cancer Center, Bunting and Blaustein Building (CRB-1), 1650 Orleans Street, Baltimore, MD 21287.
| | - John T. Isaacs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
- Department of Urology, James Buchanan Brady Urological Institute, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
33
|
Spritzer MD, Fox EC, Larsen GD, Batson CG, Wagner BA, Maher J. Testosterone influences spatial strategy preferences among adult male rats. Horm Behav 2013; 63:800-12. [PMID: 23597827 PMCID: PMC3759970 DOI: 10.1016/j.yhbeh.2013.03.018] [Citation(s) in RCA: 37] [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: 09/25/2012] [Revised: 03/26/2013] [Accepted: 03/31/2013] [Indexed: 01/09/2023]
Abstract
Males outperform females on some spatial tasks, and this may be partially due to the effects of sex steroids on spatial strategy preferences. Previous work with rodents indicates that low estradiol levels bias females toward a striatum-dependent response strategy, whereas high estradiol levels bias them toward a hippocampus-dependent place strategy. We tested whether testosterone influenced the strategy preferences in male rats. All subjects were castrated and assigned to one of three daily injection doses of testosterone (0.125, 0.250, or 0.500 mg/rat) or a control group that received daily injections of the drug vehicle. Three different maze protocols were used to determine rats' strategy preferences. A low dose of testosterone (0.125 mg) biased males toward a motor-response strategy on a T-maze task. In a water maze task in which the platform itself could be used intermittently as a visual cue, a low testosterone dose (0.125 mg) caused a significant increase in the use of a cued-response strategy relative to control males. Results from this second experiment also indicated that males receiving a high dose of testosterone (0.500 mg) were biased toward a place strategy. A third experiment indicated that testosterone dose did not have a strong influence on the ability of rats to use a nearby visual cue (floating ball) in the water maze. For this experiment, all groups seemed to use a combination of place and cued-response strategies. Overall, the results indicate that the effects of testosterone on spatial strategy preference are dose dependent and task dependent.
Collapse
Affiliation(s)
- Mark D Spritzer
- Department of Biology, Middlebury College, Middlebury, VT 05753, USA.
| | | | | | | | | | | |
Collapse
|
34
|
FRICK J, DANNER CH, JOOS H, KUNIT G, LUUKKAINEN T. Spermatogenesis in Men Treated with Subcutaneous Application of Levonorgestrel and Estrone Rods. ACTA ACUST UNITED AC 2013. [DOI: 10.1002/j.1939-4640.1981.tb00638.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
35
|
|
36
|
BEX FREDERICKJ, CORBIN ALAN. In VivoandIn VitroInvestigation of the Extrapituitary Antireproductive Effects of a Potent LHRH Agonist in Immature and Adult Male Rats. ACTA ACUST UNITED AC 2013. [DOI: 10.1002/j.1939-4640.1981.tb00610.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
37
|
DALTERIO S, BARTKE A, SWEENEY C. Interactive Effects of Ethanol and Δ9-Tetrahydrocannabinol on Endocrine Functions in Male Mice. ACTA ACUST UNITED AC 2013. [DOI: 10.1002/j.1939-4640.1981.tb00600.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
38
|
TORGERSEN H, STEINER M, ROVAN E, ADAM H, FRICK J. The Use of BCG (Bacille Calmette Guérin) as an Antispermatogenic Agent: Structural and Hormonal Changes. ACTA ACUST UNITED AC 2013. [DOI: 10.1002/j.1939-4640.1981.tb00629.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
39
|
GERRITY M, FREUND M, PETERSON RN, FALVO RE. The Physiologic Effects of Testosterone in Hydrogenated Soybean Oil Vehicle as Compared to Free Testosterone, Testosterone Propionate, and Testosterone Enanthate in a Conventional Oil Vehicle. ACTA ACUST UNITED AC 2013. [DOI: 10.1002/j.1939-4640.1982.tb00673.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
40
|
SCHANBACHER BD, D'OCCHIO MJ. Validation of a Direct Radioimmunoassay for Testosterone in Unextracted Serum from Five Species: Application to Study of the Hypothalamic-Pituitary-Gonadal Axis in Males. ACTA ACUST UNITED AC 2013. [DOI: 10.1002/j.1939-4640.1982.tb00644.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
41
|
Xu X, Coats JK, Yang CF, Wang A, Ahmed OM, Alvarado M, Izumi T, Shah NM. Modular genetic control of sexually dimorphic behaviors. Cell 2012; 148:596-607. [PMID: 22304924 DOI: 10.1016/j.cell.2011.12.018] [Citation(s) in RCA: 199] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Revised: 09/22/2011] [Accepted: 12/16/2011] [Indexed: 12/18/2022]
Abstract
Sex hormones such as estrogen and testosterone are essential for sexually dimorphic behaviors in vertebrates. However, the hormone-activated molecular mechanisms that control the development and function of the underlying neural circuits remain poorly defined. We have identified numerous sexually dimorphic gene expression patterns in the adult mouse hypothalamus and amygdala. We find that adult sex hormones regulate these expression patterns in a sex-specific, regionally restricted manner, suggesting that these genes regulate sex typical behaviors. Indeed, we find that mice with targeted disruptions of each of four of these genes (Brs3, Cckar, Irs4, Sytl4) exhibit extremely specific deficits in sex specific behaviors, with single genes controlling the pattern or extent of male sexual behavior, male aggression, maternal behavior, or female sexual behavior. Taken together, our findings demonstrate that various components of sexually dimorphic behaviors are governed by separable genetic programs.
Collapse
Affiliation(s)
- Xiaohong Xu
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Ernest SR, Wade MG, Lalancette C, Ma YQ, Berger RG, Robaire B, Hales BF. Effects of chronic exposure to an environmentally relevant mixture of brominated flame retardants on the reproductive and thyroid system in adult male rats. Toxicol Sci 2012; 127:496-507. [PMID: 22387749 PMCID: PMC3355309 DOI: 10.1093/toxsci/kfs098] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Brominated flame retardants (BFRs) are incorporated into a wide variety of consumer products, are readily released into home and work environments, and are present in house dust. Studies using animal models have revealed that exposure to polybrominated diphenyl ethers (PBDEs) may impair adult male reproductive function and thyroid hormone physiology. Such studies have generally characterized the outcome of acute or chronic exposure to a single BFR technical mixture or congener but not the impact of environmentally relevant BFR mixtures. We tested whether exposure to the BFRs found in house dust would have an adverse impact on the adult male rat reproductive system and thyroid function. Adult male Sprague Dawley rats were exposed to a complex BFR mixture composed of three commercial brominated diphenyl ethers (52.1% DE-71, 0.4% DE-79, and 44.2% decaBDE-209) and hexabromocyclododecane (3.3%), formulated to mimic the relative congener levels in house dust. BFRs were delivered in the diet at target doses of 0, 0.02, 0.2, 2, or 20 mg/kg/day for 70 days. Compared with controls, males exposed to the highest dose of BFRs displayed a significant increase in the weights of the kidneys and liver, which was accompanied by induction of CYP1A and CYP2B P450 hepatic drug–metabolizing enzymes. BFR exposure did not affect reproductive organ weights, serum testosterone levels, testicular function, or sperm DNA integrity. The highest dose caused thyroid toxicity as indicated by decreased serum thyroxine (T4) and hypertrophy of the thyroid gland epithelium. At lower doses, the thickness of the thyroid gland epithelium was reduced, but no changes in hormone levels (T4 and thyroid-stimulating hormone) were observed. Thus, exposure to BFRs affected liver and thyroid physiology but not male reproductive parameters.
Collapse
Affiliation(s)
- Sheila R Ernest
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada H3G 1Y6
| | | | | | | | | | | | | |
Collapse
|
43
|
Neese SL, Schantz SL. Testosterone impairs the acquisition of an operant delayed alternation task in male rats. Horm Behav 2012; 61:57-66. [PMID: 22047777 PMCID: PMC3308684 DOI: 10.1016/j.yhbeh.2011.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 10/11/2011] [Accepted: 10/18/2011] [Indexed: 10/15/2022]
Abstract
The current study examined the effects of gonadectomy (GDX) and subsequent testosterone treatment of male Long-Evans rats on an operant variable delay spatial alternation task (DSA). Gonadally-intact rats (intact-B), GDX rats receiving implants that delivered a physiological level of testosterone (GDX-T), and GDX rats receiving blank implants (GDX-B) were tested for 25 sessions on a DSA task with variable inter-trial delays ranging from 0 to 18 s. Acquisition of the DSA task was found to be enhanced following GDX in a time and delay dependent manner. Both the GDX-T and the intact-B rats had lower performance accuracies across delays initially, relative to GDX-B rats, and this deficit persisted into subsequent testing sessions at longer delays. The GDX-T and intact-B rats also had a tendency to commit more perseverative errors during the early testing sessions, with both groups persisting in pressing a lever which had not been associated with reinforcement for at least two consecutive trials. However, both the GDX-T and intact-B groups were able to achieve performance accuracy similar to that of the GDX-B rats by the final sessions of testing. Overall, these results suggest that castration of adult male rats enhances their acquisition of an operant DSA task.
Collapse
Affiliation(s)
- Steven L Neese
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign. 2001 S. Lincoln Avenue, Urbana, IL 61802, USA.
| | | |
Collapse
|
44
|
Churchman ML, Roig I, Jasin M, Keeney S, Sherr CJ. Expression of arf tumor suppressor in spermatogonia facilitates meiotic progression in male germ cells. PLoS Genet 2011; 7:e1002157. [PMID: 21811412 PMCID: PMC3141002 DOI: 10.1371/journal.pgen.1002157] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 05/11/2011] [Indexed: 11/18/2022] Open
Abstract
The mammalian Cdkn2a (Ink4a-Arf) locus encodes two tumor suppressor proteins (p16Ink4a and p19Arf) that respectively enforce the anti-proliferative functions of the retinoblastoma protein (Rb) and the p53 transcription factor in response to oncogenic stress. Although p19Arf is not normally detected in tissues of young adult mice, a notable exception occurs in the male germ line, where Arf is expressed in spermatogonia, but not in meiotic spermatocytes arising from them. Unlike other contexts in which the induction of Arf potently inhibits cell proliferation, expression of p19Arf in spermatogonia does not interfere with mitotic cell division. Instead, inactivation of Arf triggers germ cell–autonomous, p53-dependent apoptosis of primary spermatocytes in late meiotic prophase, resulting in reduced sperm production. Arf deficiency also causes premature, elevated, and persistent accumulation of the phosphorylated histone variant H2AX, reduces numbers of chromosome-associated complexes of Rad51 and Dmc1 recombinases during meiotic prophase, and yields incompletely synapsed autosomes during pachynema. Inactivation of Ink4a increases the fraction of spermatogonia in S-phase and restores sperm numbers in Ink4a-Arf doubly deficient mice but does not abrogate γ-H2AX accumulation in spermatocytes or p53-dependent apoptosis resulting from Arf inactivation. Thus, as opposed to its canonical role as a tumor suppressor in inducing p53-dependent senescence or apoptosis, Arf expression in spermatogonia instead initiates a salutary feed-forward program that prevents p53-dependent apoptosis, contributing to the survival of meiotic male germ cells. The intimately linked Arf and Ink4a genes, encoded in part by overlapping reading frames within the Cdkn2a locus, are induced by oncogenic stress, activating the p53 and Rb tumor suppressors, respectively, to inhibit proliferation of incipient cancer cells. As such, expression of the p19Arf and p16Ink4a proteins is undetected in most normal mouse tissues. However, p19Arf is physiologically expressed in mitotically dividing spermatogonia, the progenitor cells that differentiate to form meiotic spermatocytes in which Arf expression is extinguished. We show that, instead of provoking cell cycle arrest or death, Arf expression in spermatogonia facilitates survival of their meiotic progeny, ensuring production of normal numbers of mature sperm. When Arf is ablated, meiotic defects ensue, along with p53-dependent cell death of spermatocytes, indicating an unexpected role of p53 in monitoring meiotic progression. Surprisingly, it is the absence of p19Arf rather than its induction that enforces p53 expression in this setting. Co-inactivation of Ink4a compensates for Arf loss by fueling proliferation of spermatogonial progenitors, but does not correct meiotic defects triggered by Arf loss. Although the Arf and Ink4a tumor suppressors are expected to restrain cellular self-renewal, Arf plays an unexpected role in male germ cells by facilitating their proper meiotic progression.
Collapse
Affiliation(s)
- Michelle L. Churchman
- Howard Hughes Medical Institute, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
- Department of Genetics and Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Ignasi Roig
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Cytology and Histology Unit, Department of Cell Biology, Physiology, and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Maria Jasin
- Developmental Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Scott Keeney
- Howard Hughes Medical Institute, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Charles J. Sherr
- Howard Hughes Medical Institute, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
- Department of Genetics and Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
45
|
Spritzer MD, Daviau ED, Coneeny MK, Engelman SM, Prince WT, Rodriguez-Wisdom KN. Effects of testosterone on spatial learning and memory in adult male rats. Horm Behav 2011; 59:484-96. [PMID: 21295035 PMCID: PMC3081396 DOI: 10.1016/j.yhbeh.2011.01.009] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2010] [Revised: 01/20/2011] [Accepted: 01/26/2011] [Indexed: 10/18/2022]
Abstract
A male advantage over females for spatial tasks has been well documented in both humans and rodents, but it remains unclear how the activational effects of testosterone influence spatial ability in males. In a series of experiments, we tested how injections of testosterone influenced the spatial working and reference memory of castrated male rats. In the eight-arm radial maze, testosterone injections (0.500 mg/rat) reduced the number of working memory errors during the early blocks of testing but had no effect on the number of reference memory errors relative to the castrated control group. In a reference memory version of the Morris water maze, injections of a wide range of testosterone doses (0.0625-1.000 mg/rat) reduced path lengths to the hidden platform, indicative of improved spatial learning. This improved learning was independent of testosterone dose, with all treatment groups showing better performance than the castrated control males. Furthermore, this effect was only observed when rats were given testosterone injections starting 7 days prior to water maze testing and not when injections were given only on the testing days. We also observed that certain doses of testosterone (0.250 and 1.000 mg/rat) increased perseverative behavior in a reversal-learning task. Finally, testosterone did not have a clear effect on spatial working memory in the Morris water maze, although intermediate doses seemed to optimize performance. Overall, the results indicate that testosterone can have positive activational effects on spatial learning and memory, but the duration of testosterone replacement and the nature of the spatial task modify these effects.
Collapse
Affiliation(s)
- Mark D Spritzer
- Department of Biology, Middlebury College, Middlebury, VT 05753, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Chang SI, Kim SH, Park JD, Ryu DY. Decreased Levels of Plasma Testosterone/LH Ratio in Male Mice Exposed to Sodium Arsenite. Biomol Ther (Seoul) 2010. [DOI: 10.4062/biomolther.2010.18.3.257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
47
|
Wu D, Gore AC. Changes in androgen receptor, estrogen receptor alpha, and sexual behavior with aging and testosterone in male rats. Horm Behav 2010; 58:306-16. [PMID: 20223236 PMCID: PMC2879440 DOI: 10.1016/j.yhbeh.2010.03.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 02/26/2010] [Accepted: 03/01/2010] [Indexed: 11/16/2022]
Abstract
Reproductive aging in males is characterized by a diminution in sexual behavior beginning in middle age. We investigated the relationships among testosterone, androgen receptor (AR) and estrogen receptor alpha (ERalpha) cell numbers in the hypothalamus, and their relationship to sexual performance in male rats. Young (3months) and middle-aged (12months) rats were given sexual behavior tests, then castrated and implanted with vehicle or testosterone capsules. Rats were tested again for sexual behavior. Numbers of AR and ERalpha immunoreactive cells were counted in the anteroventral periventricular nucleus and the medial preoptic nucleus, and serum hormones were measured. Middle-aged intact rats had significant impairments of all sexual behavior measures compared to young males. After castration and testosterone implantation, sexual behaviors in middle-aged males were largely comparable to those in the young males. In the hypothalamus, AR cell density was significantly (5-fold) higher, and ERalpha cell density significantly (6-fold) lower, in testosterone- than vehicle-treated males, with no age differences. Thus, restoration of serum testosterone to comparable levels in young and middle-aged rats resulted in similar preoptic AR and ERalpha cell density concomitant with a reinstatement of most behaviors. These data suggest that age-related differences in sexual behavior cannot be due to absolute levels of testosterone, and further, the middle-aged brain retains the capacity to respond to exogenous testosterone with changes in hypothalamic AR and ERalpha expression. Our finding that testosterone replacement in aging males has profound effects on hypothalamic receptors and behavior has potential medical implications for the treatment of age-related hypogonadism in men.
Collapse
Affiliation(s)
- Di Wu
- Division of Pharmacology & Toxicology, College of Pharmacy; The University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrea C. Gore
- Division of Pharmacology & Toxicology, College of Pharmacy; The University of Texas at Austin, Austin, TX, 78712, USA
- Institute for Neuroscience; The University of Texas at Austin, Austin, TX, 78712, USA
- Institute for Cellular & Molecular Biology; The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
48
|
Penatti CAA, Davis MC, Porter DM, Henderson LP. Altered GABAA receptor-mediated synaptic transmission disrupts the firing of gonadotropin-releasing hormone neurons in male mice under conditions that mimic steroid abuse. J Neurosci 2010; 30:6497-506. [PMID: 20463213 PMCID: PMC2881304 DOI: 10.1523/jneurosci.5383-09.2010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Revised: 12/22/2009] [Accepted: 03/22/2010] [Indexed: 11/21/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) neurons are the central regulators of reproduction. GABAergic transmission plays a critical role in pubertal activation of pulsatile GnRH secretion. Self-administration of excessive doses of anabolic androgenic steroids (AAS) disrupts reproductive function and may have critical repercussions for pubertal onset in adolescent users. Here, we demonstrate that chronic treatment of adolescent male mice with the AAS 17alpha-methyltestosterone significantly decreased action potential frequency in GnRH neurons, reduced the serum gonadotropin levels, and decreased testes mass. AAS treatment did not induce significant changes in GABAA receptor subunit mRNA levels or alter the amplitude or decay kinetics of GABAA receptor-mediated spontaneous postsynaptic currents (sPSCs) or tonic currents in GnRH neurons. However, AAS treatment significantly increased action potential frequency in neighboring medial preoptic area (mPOA) neurons and GABAA receptor-mediated sPSC frequency in GnRH neurons. In addition, physical isolation of the more lateral aspects of the mPOA from the medially localized GnRH neurons abrogated the AAS-induced increase in GABAA receptor-mediated sPSC frequency and the decrease in action potential firing in the GnRH cells. Our results indicate that AAS act predominantly on steroid-sensitive presynaptic neurons within the mPOA to impart significant increases in GABAA receptor-mediated inhibitory tone onto downstream GnRH neurons, resulting in diminished activity of these pivotal mediators of reproductive function. These AAS-induced changes in central GABAergic circuits of the forebrain may significantly contribute to the disruptive actions of these drugs on pubertal maturation and the development of reproductive competence in male steroid abusers.
Collapse
Affiliation(s)
- Carlos A. A. Penatti
- Department of Physiology, Dartmouth Medical School, Hanover, New Hampshire 03755
| | - Matthew C. Davis
- Department of Physiology, Dartmouth Medical School, Hanover, New Hampshire 03755
| | - Donna M. Porter
- Department of Physiology, Dartmouth Medical School, Hanover, New Hampshire 03755
| | - Leslie P. Henderson
- Department of Physiology, Dartmouth Medical School, Hanover, New Hampshire 03755
| |
Collapse
|
49
|
Hannes RP. Androgen and Corticoid Levels in Blood and Body Extracts of High- and Low-ranking Swordtail Males (Xiphophorus helleri) before and after Social Isolation. ACTA ACUST UNITED AC 2010. [DOI: 10.1111/j.1439-0310.1984.tb01356.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
50
|
Pröve E. Quantitative Untersuchungen zu Wechselbeziehungen zwischen Balzaktivität und Testosterontitern bei männlichen Zebrafinken. ACTA ACUST UNITED AC 2010. [DOI: 10.1111/j.1439-0310.1978.tb00247.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|