1
|
Silva-Cardoso GK, N'Gouemo P. Activation of anoctamin-1 calcium-activated chloride channels reduces voluntary alcohol consumption in rats. Neuropharmacology 2025; 275:110498. [PMID: 40324648 DOI: 10.1016/j.neuropharm.2025.110498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/18/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Repeated episodes of binge drinking can lead to an alcohol use disorder, yet the underlying pharmacological mechanisms are still not fully understood. Nevertheless, emerging evidence indicates that Ca2+-dependent signaling effectively reduces alcohol consumption without affecting water intake. Therefore, activating anoctamin1 (ANO1), a Ca2+-activated chloride channel and a component of Ca2+-dependent signaling, can similarly decrease alcohol drinking while maintaining normal water intake. This study investigates how activation of ANO1 channels with EACT affects voluntary alcohol consumption in male and female Sprague-Dawley rats using the intermittent alcohol access method in a two-bottle choice paradigm. Rats were trained to drink 7.5 % ethanol or water for four weeks before administering either EACT (2.5, 5, and 10 mg/kg). Afterward, their alcohol intake, preference, and water intake were systematically recorded 2 and 24 h after exposure to water and 7.5 % ethanol solution. The results indicated that female rats consumed more alcohol than males. Furthermore, activating ANO1 channels with EACT significantly decreased alcohol intake and preference in males, only at the 5 mg/kg dose; in females, this effect was observed as a linear response at both the 5 and 10 mg/kg doses, highlighting distinct sex-related differences. Additionally, the inhibitory effect of EACT on alcohol consumption was associated with increased water intake in females, suggesting a potential influence of EACT on thirst homeostasis. Collectively, these findings highlight the differential effects of EACT on alcohol intake, preference, and water intake based on sex, and underscore the complexity of consummatory behavior mechanisms.
Collapse
Affiliation(s)
- Gleice Kelli Silva-Cardoso
- Howard University College of Medicine, Department of Physiology and Biophysics, Washington, DC, 20059, United States of America
| | - Prosper N'Gouemo
- Howard University College of Medicine, Department of Physiology and Biophysics, Washington, DC, 20059, United States of America.
| |
Collapse
|
2
|
Bashir SM, Ali SI, Rather MA, Sheikh WM, Singh H, Nabi SU, Ganie MA, Shafi M, Ul Haq Shah MZ, Bhat JI, Wani IA, Hassan S. Evaluating spironolactone monotherapy against combined treatment with metformin in rat PCOS model. Eur J Pharmacol 2025; 998:177516. [PMID: 40090535 DOI: 10.1016/j.ejphar.2025.177516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 03/18/2025]
Abstract
Polycystic ovarian syndrome (PCOS) is a common gynecological disorder with multifactorial pathogenic risk factors. Combination therapy with metformin and thiazolidinedione derivatives is frequently used, but its synergistic effects have not been thoroughly evaluated. This study aims to compare the therapeutic efficacy of low-dose spironolactone (LDS) at 0.25 mg/kg for 28 days, metformin at 500 mg/kg for 28 days, and a combination of LDS and metformin, against a letrozole (1 mg/kg/day) and 0.5 % carboxymethylcellulose (CMC)-induced PCOS rat model. The study involved five groups of laboratory animals: Group I (Healthy control), Group IIa (Disease control), Group IIb (Metformin), Group IIc (LDS), and Group IId (Metformin + LDS). Therapeutic efficacy was evaluated based on phenotypic, hormonal, and genotypic determinants. Letrozole successfully induced PCOS in the animals, evidenced by elevated levels of Sex Hormone Binding Globulin (SHBG), Follicle Stimulating Hormone (FSH), and progesterone, as well as the presence of multiple ovarian cysts. Hierarchical Cluster Analysis indicated that LDS was superior to metformin and the combination therapy in ameliorating PCOS symptoms. The findings suggest that there is little to no benefit in adding metformin to LDS for the clinical management of PCOS. Although these results are from preclinical studies, further case-controlled, randomized placebo studies on a larger patient sample are necessary to confirm these findings in clinical settings.
Collapse
Affiliation(s)
- Showkeen Muzamil Bashir
- Biochemistry and Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190006, Jammu and Kashmir, India.
| | - Sofi Imtiyaz Ali
- Biochemistry and Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190006, Jammu and Kashmir, India
| | - Muzafar Ahmad Rather
- Biochemistry and Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190006, Jammu and Kashmir, India
| | - Wajid Mohammad Sheikh
- Biochemistry and Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190006, Jammu and Kashmir, India
| | - Hemant Singh
- Department of Biological Sciences, Khalifa University, Abu Dhabi, P.O 127788, United Arab Emirates; Center for Biotechnology, Khalifa University, Abu Dhabi, P.O 127788, United Arab Emirates
| | - Showkat Ul Nabi
- Division of Veterinary Clinical Medicine, Ethics and Jurisprudence, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190006, Jammu and Kashmir, India
| | - Mohd Ashraf Ganie
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, Jammu and Kashmir, 190001, India
| | - Majid Shafi
- Division of Veterinary Pathology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190006, Jammu and Kashmir, India
| | - Mohd Zahoor Ul Haq Shah
- Laboratory of Endocrinology, Department of Bioscience, Barakatullah University Bhopal-462026, India
| | - Javeed Iqbal Bhat
- Biochemistry and Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190006, Jammu and Kashmir, India
| | - Imtiyaz Ahmad Wani
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, Jammu and Kashmir, 190001, India
| | - Shabir Hassan
- Department of Biological Sciences, Khalifa University, Abu Dhabi, P.O 127788, United Arab Emirates; Center for Biotechnology, Khalifa University, Abu Dhabi, P.O 127788, United Arab Emirates.
| |
Collapse
|
3
|
Aggarwal Y, Dixit AB, Siraj F, Tripathi M, Chandra PS, Banerjee J. Differential regulation of GABA A receptor-mediated hyperexcitability at different stages of brain development in focal cortical dysplasia (FCD). Exp Neurol 2025; 389:115265. [PMID: 40246010 DOI: 10.1016/j.expneurol.2025.115265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Focal cortical dysplasia (FCD) is a developmental abnormality of cortex commonly linked with drug-resistant seizures. Altered GABAergic activity is a key contributor to interictal discharges in FCD. In FCD, GABAA receptor associated epileptogenicity is dependent upon the age at seizure onset, as differential epileptogenic networks are observed in early and late onset FCD patients. But the contribution of GABAA receptor alteration to epileptogenic networks during development is unclear. We hypothesize that GABAergic signaling in FCD undergoes age-dependent molecular alterations, contributing to the development of distinct epileptogenic networks. In this study, we investigated age-dependent changes in GABA neurotransmitter levels, GABAA receptor α subunit expression, and GABAA receptor-mediated synaptic activity using the BCNU-rat model of FCD. GABA levels, mRNA, and protein expression of GABAA receptor α subunits were determined by HPLC, qPCR and western blot and spontaneous GABAergic activity from pyramidal neurons was recorded using whole cell patch-clamp technique. At postnatal days (P) 12 and 21, reduced expression of α1, 2 and 4 subunits were observed in FCD rats compared to control. Consistent with this, decreased amplitude and frequency of GABAergic events were observed in FCD rats. In contrast, at P30 and P65, decreased GABA levels, without changes in receptor expression, were observed in FCD rats. Consistently, reduction in the frequency of GABAergic events was observed in FCD rats compared to the control. Furthermore, treatment with tetrodotoxin (TTX) revealed that the observed alterations in GABAergic activity were predominantly action potential (AP)-dependent. Our findings indicate that distinct epileptogenic networks exist in FCD during early and late developmental stages. These networks are driven primarily by altered GABAergic activity, with early age changes linked to aberrant GABAA receptor configurations and late age changes associated with abnormal GABA levels.
Collapse
Affiliation(s)
- Yogesh Aggarwal
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Aparna Banerjee Dixit
- Dr. B. R. Ambedkar Centre for Biomedical Research, University of Delhi, New Delhi, India.
| | - Fouzia Siraj
- National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Manjari Tripathi
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - P Sarat Chandra
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Jyotirmoy Banerjee
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
4
|
Idil E, Yuksel B, Sen Z, Unal G. Estrogen receptor alpha (ERα) partially modulates ketamine's sustained anxiolytic effects without altering its antidepressant properties in female rats. Psychoneuroendocrinology 2025; 177:107455. [PMID: 40179595 DOI: 10.1016/j.psyneuen.2025.107455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 02/26/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Ketamine is a rapid-acting antidepressant with sexually dimorphic effects. Female animals exhibit a higher sensitivity to its antidepressant properties, which has been associated with their ovarian hormone levels. One factor contributing to this sex difference is the faster rate of ketamine metabolism observed in females, potentially regulated by estrogen receptor alpha (ERα) through modulation of enzymatic activity. In this study, we explored the role of ERα in mediating the therapeutic effects of ketamine in adult female Wistar rats. To inhibit ERα, we administered its antagonist, methyl-piperidino-pyrazole (MPP; 1 mg/kg, IP), 24 h and 1 h prior to a single antidepressant dose of ketamine (10 mg/kg, IP) or saline (vehicle). We tested the animals in the forced swim test (FST), open field test (OFT), elevated plus maze (EPM), and auditory fear conditioning. Ketamine administration ameliorated behavioral despair observed in the vehicle group, and ERα antagonism did not affect this outcome. An interaction between MPP and ketamine was observed in anxiety-like behaviors assessed in the OFT and EPM; however, this effect did not reach significance in post-hoc analyses. Neither MPP nor ketamine affected fear memory, as measured in cued fear conditioning. These findings suggest that the sexually dimorphic antidepressant effects of ketamine occur independently of ERα activity, although ERα may influence neural circuits related to anxiety.
Collapse
Affiliation(s)
- Ece Idil
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, Istanbul 34342, Turkey
| | - Bahar Yuksel
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, Istanbul 34342, Turkey
| | - Zeynep Sen
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, Istanbul 34342, Turkey
| | - Gunes Unal
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, Istanbul 34342, Turkey.
| |
Collapse
|
5
|
Lawrence LA, Williams ME, Vidal P, Varughese RS, Li ZRT, Chen TD, Roy MA, Tuske SC, Lowen AC, Scharer CD, Shafer WM, Swaims-Kohlmeier A. Murine modeling of menstruation identifies immune correlates of protection during Chlamydia muridarum challenge. PLoS Pathog 2025; 21:e1012276. [PMID: 40478914 DOI: 10.1371/journal.ppat.1012276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 05/06/2025] [Indexed: 06/11/2025] Open
Abstract
The menstrual cycle influences the risk of acquiring sexually transmitted infections (STIs), including those caused by the pathogen Chlamydia trachomatis (C. trachomatis). However, the underlying immune contributions are poorly defined. A mouse model simulating the repetitive immune-mediated process of menstruation could provide valuable insights into tissue-specific determinants of protection against chlamydial infection within the cervicovaginal and uterine mucosae of the female reproductive tract (FRT). Here, we used the pseudopregnancy approach for inducing menstruation in naïve C57Bl/6 mice and performed vaginal challenge with Chlamydia muridarum (C. muridarum) over the course of decidualization, endometrial tissue remodeling, and menstruation. This strategy identified that a time point over pseudopregnancy corresponding to the late luteal phase of the menstrual cycle correlated with reduced bacterial burden. By evaluating the early infection site following challenge at this time point, we found that a greater abundance of NK cell populations and proinflammatory signaling, including IFNγ, were strongly correlated with protection. FRT immune profiling in uninfected mice over pseudopregnancy or in pig-tailed macaques over the menstrual cycle identified periodic NK cell infiltration into the cervicovaginal tissues and luminal surface occurring over a similar time frame. Notably, these cell populations were transcriptionally distinct and enriched for programs associated with NK cell effector functions. Depletion of FRT NK cells during the late luteal phase resulted in a loss of protection, enabling productive infection following C. muridarum challenge. This study shows that the pseudopregnancy murine menstruation model recapitulates dynamic changes occurring in mucosal immune states throughout the FRT as a result of endometrial remodeling and identifies NK cell localization at the FRT barrier site of pathogen exposure as essential for immune protection against primary C. muridarum infection.
Collapse
Affiliation(s)
- Laurel A Lawrence
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - M Elliott Williams
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Paola Vidal
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Richa S Varughese
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Zheng-Rong Tiger Li
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Thien Duy Chen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Melissa A Roy
- Division of Pathology Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Steven C Tuske
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Anice C Lowen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - William M Shafer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Laboratories of Bacterial Pathogenesis, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States of America
| | - Alison Swaims-Kohlmeier
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of GYNOB, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Division of HIV Prevention Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| |
Collapse
|
6
|
Potapova S, Stratilov V, Vetrovoy O, Safarova D, Tyulkova E. Oestrous cycle synchronization protocols in rats using cloprostenol and progesterone injections. Lab Anim 2025; 59:369-373. [PMID: 39947897 DOI: 10.1177/00236772241309754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2025]
Abstract
This study investigated the effectiveness of combining cloprostenol (Cl) and progesterone (Pg) injections for oestrous synchronization in female rats. A comprehensive series of experiments was conducted to explore the impact of hormonal injections on subsequent reproductive behaviour. The study involved dividing rats into distinct groups, with each group subjected to specific injections of either Cl, Pg, or their combinations. We observed a 100% conception efficiency within the first day after the last Cl injection in the Cl + Pg + Cl group. This finding underscores the remarkable effectiveness of the employed protocol, resulting in a rapid initiation of pregnancy in a substantial number of female rats on the same day.
Collapse
Affiliation(s)
- Sophia Potapova
- Laboratory of Regulation of Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, Russia
- Department of Biochemistry, Faculty of Biology, Saint Petersburg State University, Russia
| | - Viktor Stratilov
- Laboratory of Regulation of Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Oleg Vetrovoy
- Laboratory of Regulation of Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, Russia
- Department of Biochemistry, Faculty of Biology, Saint Petersburg State University, Russia
| | - Diana Safarova
- Department of Biochemistry, Faculty of Biology, Saint Petersburg State University, Russia
| | - Ekaterina Tyulkova
- Laboratory of Regulation of Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, Russia
| |
Collapse
|
7
|
Bharti N, Yadav P, Choudhary SK, Dhaked RK, Mali PC. Cassia siamea-Derived Silver Nanoparticles: Synthesis and Their Impact on Male Fertility Through Biochemical and Histopathologixcal Insights in Rats. Biol Trace Elem Res 2025:10.1007/s12011-025-04658-2. [PMID: 40448812 DOI: 10.1007/s12011-025-04658-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 05/03/2025] [Indexed: 06/02/2025]
Abstract
The present study was designed to assess the effects of Cassia siamea leaf extract-mediated silver nanoparticles treatment on the fertility potential of male albino Wistar rats, to check their contraceptive efficacy. This plant is being traditionally used for various therapeutic purposes, including treating fever, skin diseases, hypertension, insomnia, diabetes, and asthma, as well as other pharmacological activities such as antimicrobial, antifertility, antioxidant, and analgesic properties. For this purpose, nanoparticles of the plant leaves were synthesized and characterized by different techniques as UV-Vis spectroscopic analysis, EDS, and DLS analysis. The animals were divided equally into five treatment groups, drug of 10, 20, and 30 mg/kg body weight per day was administered orally for 60 days, including group-I as control (vehicle treated) and group-V was kept at 30-day recovery period. A significant change in the reproductive organ weights, such as testes, epididymides, ventral prostate, seminal vesicles, and vas deferens, was observed at different dose levels compared to the control. Changes in sperm dynamics (motility and density) were also observed in the treated groups. Tissue biochemistry results revealed a substantial dose-dependent decrease in various parameters including protein, glycogen, cholesterol, fructose, and sialic acid in different reproductive organs, which might be silver-induced toxicity, whereas recovery groups showed a resurgence of these parameters once the doses were ceased for the following 30 days. In hormone analysis, a decrease in testosterone levels was also observed in a dose-response manner. Testosterone exhibited a correlation with the histopathological examination of the testis, revealing degenerative changes in seminiferous tubules and decreased spermatogenesis. These results support the notion that C. siamea leaf extract AgNPs possess antifertility potential and may be employed as a contraceptive.
Collapse
Affiliation(s)
- Neha Bharti
- Reproductive Biomedicine and Natural Products Lab, Department of Zoology, Centre for Advanced Studies, University of Rajasthan, Jaipur, 302004, India
| | - Prity Yadav
- Reproductive Biomedicine and Natural Products Lab, Department of Zoology, Centre for Advanced Studies, University of Rajasthan, Jaipur, 302004, India
| | - Sandeep Kumar Choudhary
- SMS (Plant Protection), Krishi Vigyan Kendra, Banasthali Vidyapith, Tonk, 304022, Rajasthan, India
| | - Rajeev Kumar Dhaked
- RUSA 2.0. Project No.5, Department of Zoology, Centre for Advanced Studies, University of Rajasthan, Jaipur, 302004, India
| | - Pratap Chand Mali
- Reproductive Biomedicine and Natural Products Lab, Department of Zoology, Centre for Advanced Studies, University of Rajasthan, Jaipur, 302004, India.
| |
Collapse
|
8
|
Neitzke EV, Dos Santos FG, Zanini BM, Cavalcante MB, Mason JB, Masternak MM, de Souza ICC, Schneider A. The influence of ovarian activity and menopause on mental health: Evidence from animal models and women. Physiol Behav 2025; 294:114886. [PMID: 40118132 DOI: 10.1016/j.physbeh.2025.114886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/28/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Hormonal variations occurring throughout the female reproductive cycle have a significant impact on physical and mental health, particularly due to the influence of estradiol (E2) and progesterone (P4). These changes are directly related to alterations in neurological systems, being associated with conditions such as premenstrual syndrome (PMS), premenstrual dysphoric disorder (PMDD), and mood disorders during hormonal transition phases, such as perimenopause and menopause. Studies conducted in humans and animal models indicate that these fluctuations affect neurotransmitters, neural plasticity, and patterns of brain activity, ultimately influencing quality of life and mental health. Despite extensive research on the topic, the interactions between sex hormones, mental health, and reproductive aging still require further investigation, emphasizing approaches that simultaneously address experimental and behavioral aspects. Thus, this review aims to sumarize findings about the influence of hormonal fluctuations throughout the female reproductive lifespan, including transitions such as perimenopause and menopause, on mental health. A comparative analysis of data from studies in animal models and humans was conducted, highlighting neuroendocrine, behavioral, and emotional mechanisms associated with hormonal changes and their impacts on female mental health.
Collapse
Affiliation(s)
- Ediana V Neitzke
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas RS, Brazil
| | | | - Bianka M Zanini
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas RS, Brazil
| | - Marcelo B Cavalcante
- Postgraduate Program in Medical Sciences, University of Fortaleza (UNIFOR), Fortaleza, CE, Brazil
| | - Jeffrey B Mason
- College of Veterinary Medicine, Department of Veterinary Clinical and Life Sciences, Center for Integrated BioSystems, Utah State University, Logan, UT, USA
| | - Michal M Masternak
- University of Central Florida, College of Medicine, Burnett School of Biomedical Sciences, Orlando, Florida, USA; Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Izabel C C de Souza
- Instituto de Biologia, Departamento de Morfologia, Universidade Federal de Pelotas, Pelotas RS, Brazil
| | - Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas RS, Brazil.
| |
Collapse
|
9
|
Luyckx L, Myllykangas M, Saarela U, Virtanen N, Hurskainen E, Savolainen A, Ollikainen N, Norlén AK, Ohlsson C, Poutanen M, Velde GV, Arffman RK, Prunskaite-Hyyryläinen R, Vriens J, Piltonen TT. Prenatally androgenized PCOS mice have ovary-independent uterine dysfunction and placental inflammation aggravated by high-fat diet. SCIENCE ADVANCES 2025; 11:eadu3699. [PMID: 40344073 PMCID: PMC12063661 DOI: 10.1126/sciadv.adu3699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/03/2025] [Indexed: 05/11/2025]
Abstract
Polycystic ovary syndrome (PCOS) is a common hyperandrogenic and metabolic condition in women. The syndrome is linked to subfertility and pregnancy complications, yet the independent effects of exposure to hyperandrogenism and obesity on endometrial function remain unclear. Here, PCOS-like mice were generated using prenatal androgenization (PNA) with dihydrotestosterone, followed by a prepubertal high-fat (HF) or standard diet. In ovariectomized mice, PNA impaired uterine closure during the implantation window, disrupted decidualization, and altered extracellular matrix- and inflammation-related gene expression. The effects were aggravated by the HF diet. In naturally mated, ovary-intact mice, PNA and HF diet affected decidual and placental gene expression, suggestive of placental dysfunction and inflammation, and induced fetal growth restriction. This study underlines the role of the uterus in adverse pregnancy outcomes in PCOS and identifies possible underlying mechanisms for future studies. Prepregnancy interventions targeting metabolic health and hyperandrogenism should be the next steps to optimize PCOS pregnancy outcomes.
Collapse
Affiliation(s)
- Lena Luyckx
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
- Research Group for Implantation, Placentation and Pregnancy, Department of Development and Regeneration, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Milena Myllykangas
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Ulla Saarela
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Nikke Virtanen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Elisa Hurskainen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Audrey Savolainen
- Protein and Structural Biology Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland
| | - Nadja Ollikainen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Anna-Karin Norlén
- Department of Clinical Chemistry, Faculty of Medicine, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, Gothenburg University, 41345 Gothenburg, Sweden
| | - Matti Poutanen
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, Gothenburg University, 41345 Gothenburg, Sweden
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, Faculty of Medicine, University of Turku, 20014 Turku, Finland
| | - Greetje Vande Velde
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Riikka K. Arffman
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Renata Prunskaite-Hyyryläinen
- Protein and Structural Biology Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland
| | - Joris Vriens
- Research Group for Implantation, Placentation and Pregnancy, Department of Development and Regeneration, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Terhi T. Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| |
Collapse
|
10
|
Huang W, Wei L, Tang J, Nong L, Tang N, Wen Q, Qin Z, Xu L, Li J, Fan L. Impact of relative estradiol changes during ovarian stimulation on blastocyst formation and live birth in assisted reproductive technology. Sci Rep 2025; 15:15617. [PMID: 40320433 PMCID: PMC12050273 DOI: 10.1038/s41598-025-00200-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/25/2025] [Indexed: 05/08/2025] Open
Abstract
This study aimed to evaluate the predictive value of relative change in E2 levels during controlled ovarian stimulation (COS) on embryo development and pregnancy outcomes in assisted reproductive technology (ART). We retrospectively analyzed 9,376 patients who underwent their first fresh ART cycle from January 1, 2020, to December 31, 2022. Patients were classified into four groups based on relative change in E2 levels: low response group, moderate response group, moderate-high response group, and high response group. The primary outcomes were blastocyst formation rate, clinical pregnancy rate, and live birth rate, while secondary outcomes included miscarriage rate and ectopic pregnancy rate. Most cycles (96.5%) demonstrated an increase in E2 levels during COS. The blastocyst formation rate significantly increased across the groups (low response group: 0.13, moderate response group: 0.21, moderate-high response group: 0.28, high response group: 0.34; P < 0.001). Multivariable logistic regression showed significantly higher blastocyst formation rates in the moderate response group (adjusted OR = 2.012, 95% CI: 1.687-2.399), moderate-high response group (adjusted OR = 4.613, 95% CI: 3.853-5.523), and high response group (adjusted OR = 11.295, 95% CI: 9.192-13.880) compared to the low response group. Both clinical pregnancy rate and live birth rate were significantly higher in the moderate-high response group and high response group compared to the low response group (clinical pregnancy rate: 54.5% and 61.5% vs. 35.5%, adjusted RR = 1.21 [95% CI: 1.03-1.42] and 1.27 [95% CI: 1.08-1.51]; live birth rate: 44.9% and 52.0% vs. 25.7%, adjusted RR = 1.27 [95% CI: 1.06-1.52] and 1.35 [95% CI: 1.11-1.64]). However, no significant differences were observed in either clinical pregnancy rate or live birth rate between the moderate response group and low response group (clinical pregnancy rate: adjusted RR = 1.07 [95% CI: 0.91-1.25]; live birth rate: adjusted RR = 1.11 [95% CI: 0.92-1.33]). No significant differences in miscarriage rate or ectopic pregnancy rate were observed across the groups. Higher E2 responses were associated with improved embryo development and better pregnancy outcomes.
Collapse
Affiliation(s)
- Wenjie Huang
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi, China.
- Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China.
| | - Liuyan Wei
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi, China
| | - Juan Tang
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi, China
| | - Liuying Nong
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi, China
| | - Ni Tang
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi, China
| | - Qiuyue Wen
- Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Zuxing Qin
- Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Lixiang Xu
- Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Jingjing Li
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi, China.
- Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China.
- Guangxi Clinical Research Center for Obstetrics and Gynecology, Liuzhou, Guangxi, China.
- Liuzhou Key Laboratory of Gynecologic Tumor, Liuzhou, Guangxi, China.
| | - Li Fan
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi, China.
- Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China.
- Guangxi Clinical Research Center for Obstetrics and Gynecology, Liuzhou, Guangxi, China.
- Liuzhou Key Laboratory of Gynecologic Tumor, Liuzhou, Guangxi, China.
| |
Collapse
|
11
|
Caulfield ME, Vander Werp MJ, Stancati JA, Collier TJ, Sortwell CE, Sandoval IM, Kordower JH, Manfredsson FP, Steece-Collier K. Advancing age and sex modulate antidyskinetic efficacy of striatal Ca V1.3 gene therapy in a rat model of Parkinson's disease. Neurobiol Aging 2025; 149:54-66. [PMID: 40010015 PMCID: PMC12007665 DOI: 10.1016/j.neurobiolaging.2025.02.003] [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: 09/05/2024] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025]
Abstract
We previously demonstrated that viral vector-mediated striatal CaV1.3 calcium channel downregulation in young adult (3mo) male parkinsonian rats provides uniform, robust protection against levodopa-induced dyskinesias (LID). Acknowledging the association of PD with aging and incidence in male and female sexes, we have expanded our studies to include rats of advancing age of both sexes. The current study directly contrasts age and sex, determining their impact on efficacy of intrastriatal AAV-CaV1.3-shRNA to prevent LID induction, removing the variable of levodopa-priming. Considering both sexes together, late-middle-aged ('aged'; 15mo) parkinsonian rats receiving AAV-CaV1.3-shRNA developed significantly less severe LID compared control AAV-scramble(SCR)-shRNA rats, however therapeutic benefit was significantly less robust than observed in young males. When considered separately, females showed significantly less therapeutic benefit than males. Furthermore, aged non-cycling/proestrous-negative female rats were refractory to LID induction, regardless of vector. This study provides novel insight into the impact of age and sex on the variable antidyskinetic responses of CaV1.3-targeted gene therapy, highlighting the importance of including clinically relevant age and sex populations in PD studies.
Collapse
Affiliation(s)
- Margaret E Caulfield
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Molly J Vander Werp
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Jennifer A Stancati
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Timothy J Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Ivette M Sandoval
- Parkinson's Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Jeffrey H Kordower
- ASU-Banner Neurodegenerative Disease Research Center (NDRC), College of Liberal Arts and Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Fredric P Manfredsson
- Parkinson's Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA.
| |
Collapse
|
12
|
Bentall L, Parr‐Brownlie L. Sexual Dimorphism in Levodopa-Induced Dyskinesia Following Parkinson's Disease: Uncharted Territory. Eur J Neurosci 2025; 61:e70144. [PMID: 40360439 PMCID: PMC12075048 DOI: 10.1111/ejn.70144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 04/12/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025]
Abstract
Sexual dimorphism is well-documented in Parkinson's disease (PD); however, when it comes to levodopa-induced dyskinesia (LID), epidemiological and clinical findings are scarce. This is an oversight because recent studies show significant correlations between LID risk and female sex. Estrogen strongly impacts neuronal function, affecting cognitive tasks such as movement, object recognition, and reward. In movement pathways, estrogen increases dopamine synthesis, transmission, and regulation, resulting in neuroprotection for PD in women. However, following menopause, PD prevalence, symptom severity, and LID risk increase for women. Consequently, early to mid-life estrogen state is neuroprotective, but later in life becomes a risk factor for PD and LID. This review explores estrogen's action in the brain, specifically within the dopamine system. Sexual dimorphism is described for the prevalence and onset of PD and LID. We examine the cellular basis of estrogen's role in sexual dimorphism and integrate these ideas to hypothesize why the risk for LID is higher for women, than men, with PD. Lastly, this review proposes that women with PD need their symptoms to be considered and managed differently to males. Treatment of women with PD should be based on their menopausal stage, as estrogen may be masking, exacerbating, or complicating symptoms. Importantly, we present these concepts to stimulate discussion among clinical and bench scientists so that key experiments can be conducted to examine the mechanisms underlying LID, so they can be prevented to improve the quality of life for women and men living with PD in the future.
Collapse
|
13
|
Cramoisy S, Cabeza L, Ramadan B, Houdayer C, Haffen E, Belin D, Peterschmitt Y, Bourasset F. Cumulative effect of stress on decisional exploration-to-exploitation switch assessed through a gambling task in female mice. Brain Res 2025; 1854:149546. [PMID: 40043786 DOI: 10.1016/j.brainres.2025.149546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/24/2025]
Abstract
Survival and well-being hinge on an organism's ability to evaluate options, weighing costs and benefits to make adaptive decisions. It has long been shown that stress influences cognition and reward-related behaviour, the nature of which depends on the stressor's type and duration as well as gene x environment interactions. However, how stress influence decision-making in females has not been completely elucidated. Here, we have developed a new mouse gambling task (mGT) adapted to assess decision-making under uncertainty and risk. Adult female C57BL/6JRj mice administered with corticosterone (CORT) for 5 or 8 weeks reached similar final performance in the mGT as vehicle-treated controls. All groups tended to learn to maximize gain as the task progressed. Our results revealed that individual choice kinetics is impacted by chronic exposure to CORT, showing an accentuated sensitivity to penalties in female mice. These results confirm the suitability of our new mGT to assess decision-making under uncertainty and risk and are in line with previous reports of the effect of chronic CORT treatment on decision-making in male mice. Thereby this study provides new insights into the influence of sex and stress on decision-making.
Collapse
Affiliation(s)
- Stéphanie Cramoisy
- Université Marie et Louis Pasteur, UMR INSERM 1322 LINC, F-25000 Besançon, France.
| | - Lidia Cabeza
- Université Marie et Louis Pasteur, UMR INSERM 1322 LINC, F-25000 Besançon, France
| | - Bahrie Ramadan
- Université Marie et Louis Pasteur, UMR INSERM 1322 LINC, F-25000 Besançon, France
| | - Christophe Houdayer
- Université Marie et Louis Pasteur, UMR INSERM 1322 LINC, F-25000 Besançon, France
| | - Emmanuel Haffen
- Université Marie et Louis Pasteur, UMR INSERM 1322 LINC, service de psychiatrie de l'adulte, CIC-1431 INSERM, CHU de Besançon F-25030, France
| | - David Belin
- Department of Psychology, University of Cambridge, Cambridge, United Kingdom
| | - Yvan Peterschmitt
- Université Marie et Louis Pasteur, UMR INSERM 1322 LINC, F-25000 Besançon, France
| | - Fanchon Bourasset
- Université Marie et Louis Pasteur, UMR INSERM 1322 LINC, F-25000 Besançon, France.
| |
Collapse
|
14
|
Gilfarb RA, Ranade S, Smail M, Wangler L, Stewart M, Rajesh A, Lenz KM, Leuner B. Hormonal contraceptives during adolescence impact the female brain and behavior in a rat model. Horm Behav 2025; 171:105725. [PMID: 40188588 DOI: 10.1016/j.yhbeh.2025.105725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/08/2025]
Abstract
Millions of people take hormonal contraceptives (HCs), often starting during adolescence when ovarian hormones influence brain and behavioral maturation. However, there is a fundamental lack of information about the neurobehavioral consequences of hormonal alterations via adolescent HC use. To begin addressing this gap, we validated a rodent model of adolescent HC administration and characterized its impact on endocrine, transcriptional, and behavioral endpoints. Cohorts of intact post-pubertal female Sprague-Dawley rats received daily subcutaneous injections of either vehicle or HC [10 μg ethinyl estradiol (EE) + 20 μg levonorgestrel (LNG)] for the duration of adolescence from postnatal day (PND) 35 to PND56. Blood and brain tissue was collected at PND57. Other cohorts received daily injections of vehicle or HC from PND35 until behavioral assays were completed on PND57-64. HC treatment was effective, as vaginal lavage indicated disrupted estrous cycling and ELISA indicated suppressed serum luteinizing hormone in HC-treated rats. Liquid chromatography-mass spectrometry analysis showed EE and LNG in serum and brain as well as diminished serum and brain levels of allopregnanolone and testosterone in HC-treated rats. NanoString nCounter analysis indicated that adolescent HC administration impacted expression of genes related to synapses, white matter, neuroimmune, monoamine, and hormone signaling in the hypothalamus and medial prefrontal cortex. While no effects of HCs were seen on sociability in the social preference test or stress coping behavior in the forced swim test, adolescent HC administration diminished risk-assessment behaviors in the novelty-induced hypophagia paradigm and altered anxiety-like behavior in the open field test and elevated plus maze. Overall, these data suggest that exposure to contraceptive hormones during the critical developmental period of adolescence may shape the brain and behavior.
Collapse
Affiliation(s)
- Rachel A Gilfarb
- Neuroscience Graduate Program, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA
| | - Sanjana Ranade
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Marissa Smail
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Lynde Wangler
- Neuroscience Graduate Program, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA
| | - Meredith Stewart
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Abhishek Rajesh
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Kathryn M Lenz
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, 370 W. 9th Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Benedetta Leuner
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, 370 W. 9th Avenue, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
15
|
Hao J, Zhao Y, Liu X, Xu H, Liu L, Wang H, Lan Y, Ji L. Whole-transcriptome sequencing reveals the effects of acupuncture on early embryos post-IVF-ET in poor ovarian response. J Ovarian Res 2025; 18:91. [PMID: 40307905 PMCID: PMC12044900 DOI: 10.1186/s13048-025-01682-7] [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: 10/03/2024] [Accepted: 04/23/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Poor ovarian response (POR) refers to a pathological condition where the ovaries respond inadequately to gonadotropin stimulation during ovulation induction. The decline in both the quantity and quality of available oocytes makes achieving pregnancy through IVF-ET more challenging. Several studies have demonstrated that acupuncture can improve embryo quality, increase the number of high-quality embryos, and enhance pregnancy outcomes in IVF-ET patients, although the underlying mechanisms remain unclear. Therefore, this study aimed to investigate the molecular mechanisms by which acupuncture influences early embryo development in POR mice after IVF-ET through whole-transcriptome sequencing. METHODS We established POR mice model and performed acupuncture treatment. Fresh denuded oocytes were retrieved and transplanted via IVF-ET into new donor female mice to obtain early-stage embryo tissues. Vaginal smear tests were conducted to monitor estrous cycle changes, while oocyte retrieval, ovarian wet weight, and ovarian index were assessed. Serum concentrations of anti-Müllerian hormone (AMH), follicle-stimulating hormone (FSH), estradiol (E2), and luteinizing hormone (LH) were measured via ELISA. Histopathological changes and apoptosis in the ovaries were evaluated using HE and TUNEL staining. Whole-transcriptome sequencing was employed to establish expression profiles of differentially expressed mRNAs (DEmRNAs), DEmiRNAs, DElncRNAs, and DEcircRNAs. The circ/lncRNA-miRNA-mRNA network was constructed to analyze the biological functions and potential mechanisms of acupuncture on early embryo development post-IVF-ET in POR mice. RESULTS Our results demonstrated that acupuncture improved ovarian function in POR mice, corrected serum hormone imbalances, and alleviated abnormal apoptosis in ovarian granulosa cells. Through whole-transcriptome sequencing, we identified 685 DEmRNAs, 13 DEmiRNAs, 325 DElncRNAs, and 4 DEcircRNAs exhibiting regulatory trends. By constructing the circ/lncRNA-miRNA-mRNA network, we found that miR-291a-3p, miR-294-3p, and miR-295-3p may serve as key targets influencing early embryo development via multiple pathways, including the Toll-like receptor signaling pathway and p53 signaling pathway. CONCLUSION Overall, our study is the first to reveal the molecular mechanisms by which acupuncture improves early embryo development under POR conditions, providing new evidence for the use of acupuncture in assisted reproduction.
Collapse
Affiliation(s)
- Jianheng Hao
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
- The Second Clinical College, Shanxi University of Chinese Medicine, Jinzhong, 030619, Shanxi, China
| | - Yuemeng Zhao
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Xuan Liu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Huichao Xu
- The Second Clinical College, Shanxi University of Chinese Medicine, Jinzhong, 030619, Shanxi, China
| | - Liying Liu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Haijun Wang
- The Second Clinical College, Shanxi University of Chinese Medicine, Jinzhong, 030619, Shanxi, China
| | - Ying Lan
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China.
- Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, 610075, Sichuan, China.
| | - Laixi Ji
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China.
- The Second Clinical College, Shanxi University of Chinese Medicine, Jinzhong, 030619, Shanxi, China.
| |
Collapse
|
16
|
Tsai YR, Liao YN, Tsai CJ, Lee YA, Hsia SM, Lan KC, Kang HY. Differential Effects of Canonical Androgens and 11-Ketotestosterone on Reproductive Phenotypes and Folliculogenesis in Mouse Model of PCOS. Biomedicines 2025; 13:1077. [PMID: 40426904 PMCID: PMC12109480 DOI: 10.3390/biomedicines13051077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/26/2025] [Accepted: 04/27/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Polycystic ovary syndrome (PCOS) is a common female endocrine disorder characterized by hyperandrogenism, ovulatory dysfunction, and polycystic ovarian morphology. While canonical androgens like testosterone (T) and dihydrotestosterone (DHT) are well studied in PCOS pathophysiology, the role of 11-ketotestosterone (11KT) remains unclear. This study investigates the differential effects of these androgens on folliculogenesis, ovulation, and steroidogenesis using in vivo and in vitro models. Methods: Four-week-old female C57BL/6 mice received T, DHT, or 11KT for six weeks. The assessments included body weight, estrous cyclicity, serum hormone profiles, ovarian histology, and follicle classification. In parallel, large preantral follicles were cultured with each androgen to evaluate follicle growth, antrum formation, and ovulation capacity. Androgen receptor (AR) signaling and steroidogenic function were analyzed using western blotting, RT-qPCR, and luciferase reporter assays. Results: The DHT-treated mice exhibited increased weight gain, whereas 11KT-treated mice showed reduced weight gain. T and DHT disrupted the estrous cycle, while 11KT prolonged diestrus. All androgen treatments led to ovarian morphological changes, including follicular arrest and cystic features. In vitro, all androgens enhanced follicle growth, but only T and DHT inhibited ovulation. The AR expression was elevated across all androgen-treated groups, but only DHT significantly activated AR and CYP19A1 promoters. Conclusions: 11KT induces a distinct and milder PCOS-like phenotype compared to classical androgens, promoting follicle growth with minimal impact on ovulation or steroidogenic disruption. These findings underscore the heterogeneity of PCOS and suggest that different androgen profiles may drive diverse clinical phenotypes. By elucidating the distinct roles of different androgens, this may lead to better stratification of PCOS phenotypes based on predominant androgen types for more precise diagnosis and individualized management.
Collapse
Affiliation(s)
- Yi-Ru Tsai
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- An-Ten Obstetrics and Gynecology Clinic, Kaohsiung 802, Taiwan
| | - Yen-Nung Liao
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Department of Chinese Medicine, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan
| | - Cheng-Ju Tsai
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Yu-Ang Lee
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Shih-Min Hsia
- Graduate Institute of Metabolism and Obesity Sciences, School of Food Safety, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Kuo-Chung Lan
- Center for Hormone and Reproductive Medicine Research, Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan
| | - Hong-Yo Kang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Center for Hormone and Reproductive Medicine Research, Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| |
Collapse
|
17
|
Islam J, Rahman MT, Ali M, Kc E, Lee HJ, Hyun SH, Park YS. CaMKIIα-NpHR-Mediated Optogenetic Inhibition of DRG Glutamatergic Neurons by Flexible Optic Fiber Alleviates Chronic Neuropathic Pain. Neuromolecular Med 2025; 27:26. [PMID: 40227491 DOI: 10.1007/s12017-025-08848-y] [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: 10/15/2024] [Accepted: 03/22/2025] [Indexed: 04/15/2025]
Abstract
Glutamatergic neurons of the dorsal root ganglion (DRGg) exert a significant effect on peripheral nociceptive signal transmission. However, assessing the explicit modulatory effect of DRGg during chronic neuropathic pain (CNP) with neuromodulation techniques remains largely unexplored. Therefore, we inhibited DRGg by optogenetic stimulation and examined whether it could alleviate CNP and associated anxiety-related behaviors in a chronic compressed DRG (CCD) rat model. The CCD pain model was established by inserting an L-shaped rod into the lumbar 5 (L5) intervertebral foramen, and either AAV2-CaMKIIα-eNpHR3.0-mCherry or AAV2-CaMKIIα-mCherry was injected into the L5 DRG. Flexible optic fibers were implanted to direct yellow light into the L5 DRG. Pain and anxiety-related behavioral responses were assessed using mechanical threshold, mechanical latency, thermal latency, and open field tests. In vivo single-unit extracellular recording from the DRG and ventral posterolateral (VPL) thalamus was performed. CNP and anxiety-related behavioral responses along with increased neural firing activity of the DRG and VPL thalamus were observed in CCD animals. Enhanced expression of nociception-influencing molecules was found in the DRG and spinal dorsal horn (SDH). In contrast during optogenetic stimulation, specific DRGg inhibition markedly alleviated the CNP responses and reduced the DRG and VPL thalamic neural hyperactivity in CCD animals. Inhibition of DRGg also reduced the active expression of nociceptive signal mediators in the DRG and SDH. Taken together, our findings suggest that CaMKIIα-NpHR-mediated optogenetic inhibition of DRGg can produce antinociceptive effects in CCD rats during peripheral nerve injury-induced CNP condition by altering peripheral nociceptive signal input in the spinothalamic tract.
Collapse
Affiliation(s)
- Jaisan Islam
- Department of Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Md Taufiqur Rahman
- Department of Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Muhammad Ali
- Department of Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Elina Kc
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Hyun Jik Lee
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea
| | - Sang Hwan Hyun
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea
| | - Young Seok Park
- Department of Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Korea.
- Department of Neurosurgery, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, 776, 1 Sunhwanro, Seowon-gu, Cheongju, 28644, Chungbuk, Korea.
| |
Collapse
|
18
|
Jha N, Sarsaiya P, Tomar AK, Pardhiya S, Nirala JP, Chaturvedi PK, Gupta S, Rajamani P. Effects of 700 MHz radiofrequency radiation (5 G lower band) on the reproductive parameters of female Wistar rats. Reprod Toxicol 2025; 135:108910. [PMID: 40220970 DOI: 10.1016/j.reprotox.2025.108910] [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: 01/30/2025] [Revised: 03/31/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
The advent of 5 G technology has raised concerns about its potential biological effects, particularly reproductive health being one key area of focus. This study investigated the impact of 700 MHz, a lower 5 G frequency band, on the reproductive health of female Wistar rats. The experiment analyzed the effects of short-term and long-term exposure to 700 MHz mobile radiation on female Wistar rats. Rats were divided into three groups (control, sham-exposed, and exposed), with sample sizes of n = 6 for short-term and n = 8 for long-term exposure. For short-term exposure, rats were subjected to 6 hrs of radiation daily for 10 days, while for long-term exposure, rats exposed 4 hrs daily for 60 days. Physiological parameters, including estrous cyclicity, were monitored, and histopathological and biochemical analyses were conducted on harvested ovaries. Comet assay was performed to assess DNA damage. The results indicated no changes in estrous cycles or comet assay parameters in either exposure group. Serological hormone levels, including estradiol and progesterone, remained within normal ranges, but a slight yet significant increase in testosterone levels was observed in exposed groups. Oxidative stress markers revealed elevated malondialdehyde (MDA) levels and significant decreases in superoxide dismutase (SOD), total sulfhydryl content, and ferric reducing antioxidant power (FRAP) in exposed ovaries. Histopathological analysis showed no significant changes in ovarian morphology in short-term exposure but revealed alterations, including cystic follicles and abnormal vasculature, in long-term exposure. These findings suggest that 700 MHz radiation may induce oxidative stress and tissue changes in ovarian samples over prolonged exposure.
Collapse
Affiliation(s)
- Neha Jha
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Priyanka Sarsaiya
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Anuj Kumar Tomar
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Sonali Pardhiya
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Jay Prakash Nirala
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, India
| | - P K Chaturvedi
- Department of Reproductive Biology, AIIMS New Delhi, India
| | - Surabhi Gupta
- Department of Reproductive Biology, AIIMS New Delhi, India
| | - Paulraj Rajamani
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
19
|
Khodir SA, Sweed E, Motawea SM, Al-Gholam MA, Elnaidany SS, Dayer MZS, Ameen O. Diacerein and myo-inositol alleviate letrozole-induced PCOS via modulation of HMGB1, SIRT1, and NF-kB: A comparative study. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4179-4197. [PMID: 39432066 PMCID: PMC11978706 DOI: 10.1007/s00210-024-03497-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 09/26/2024] [Indexed: 10/22/2024]
Abstract
Polycystic ovary syndrome (PCOS) is the most prevalent cause of anovulatory infertility in women. Myo-inositol supplementation has displayed effectiveness in curing PCOS patients. Diacerein, an anti-inflammatory medication, has not been extensively studied in the context of reproductive disorders. This study aimed to compare the role of myo-inositol and diacerein in PCOS and the probable mechanisms mediating their actions. Forty adult female rats were divided equally into the following: control, PCOS, PCOS+Myo-inositol, and PCOS+Diacerein groups. Rats were subjected to arterial blood pressure (ABP), electromyography (EMG), and uterine reactivity measurements. Blood samples were collected for measuring hormonal assays, glycemic state, lipid profile, oxidative stress, and inflammatory markers. Ovaries and uteri were extracted for histological examination, including hematoxylin and eosin staining, Masson's trichrome staining, immunohistochemistry, and rt-PCR analysis of ovarian tissues. PCOS was associated with significant increases in ABP, uterine frequency and amplitude of contraction, luteinizing hormone, testosterone, lipid, glycemic and inflammatory markers, malondialdehyde, high-mobility group box 1 (HMGB1), nuclear factor kappa (NF-kB), ovarian fibrosis, and endometrial thickening. In contrast, there was a significant reduction in follicular stimulating hormone, reduced glutathione, and Sirtuin 1 (SIRT1) when compared with control group. Both myo-inositol and diacerein counteract PCOS changes; but diacerein's effects were superior to myo-inositol's for all parameters, except for lipid and glycemic markers. Diacerein possessed anti-inflammatory properties and showed significant efficacy in mitigating the endocrinal, metabolic, and ovarian structural alterations linked to PCOS. Its beneficial actions likely stem from reducing oxidative stress, dyslipidemia, and hyperglycemia, potentially through the modulation of HMGB1, SIRT1, and NF-kB pathways.
Collapse
Affiliation(s)
- Suzan A Khodir
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Menoufia, 32511, Egypt
- Medical Physiology, Menoufia National University, Menoufia, Egypt
| | - Eman Sweed
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Menoufia, 32511, Egypt.
- Quality Assurance Unit, Menoufia National University, Menoufia, Egypt.
| | - Shaimaa Mohamed Motawea
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Menoufia, 32511, Egypt
| | - Marwa A Al-Gholam
- Human Anatomy and Embryology Department, Faculty of Medicine, Menoufia University, Menoufia, 32511, Egypt
| | - Sherin Sobhy Elnaidany
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Menoufia University, Menoufia, 32511, Egypt
| | | | - Omnia Ameen
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Menoufia, 32511, Egypt
| |
Collapse
|
20
|
Lee BH, Eid RS, Hodges TE, Barth C, Galea LAM. Leveraging research into sex differences and steroid hormones to improve brain health. Nat Rev Endocrinol 2025; 21:214-229. [PMID: 39587332 DOI: 10.1038/s41574-024-01061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 11/27/2024]
Abstract
Sex differences, driven in part by steroid hormones, shape the structure and function of the brain throughout the lifespan and manifest across brain health and disease. The influence of steroid hormones on neuroplasticity, particularly in the adult hippocampus, differs between the sexes, which has important implications for disorders and diseases that compromise hippocampus integrity, such as depression and Alzheimer disease. This Review outlines the intricate relationship between steroid hormones and hippocampal neuroplasticity across the adult lifespan and explores how the unique physiology of male and female individuals can affect health and disease. Despite calls to include sex and gender in research, only 5% of neuroscience studies published in 2019 directly investigated the influence of sex. Drawing on insights from depression, Alzheimer disease and relevant hippocampal plasticity, this Review underscores the importance of considering sex and steroid hormones to achieve a comprehensive understanding of disease susceptibility and mechanisms. Such consideration will enable the discovery of personalized treatments, ultimately leading to improved health outcomes for all.
Collapse
Affiliation(s)
- Bonnie H Lee
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Rand S Eid
- Department of Psychology, McGill University, Montreal, Quebec, Canada
| | - Travis E Hodges
- Department of Psychology and Education, Mount Holyoke College, South Hadley, MA, USA
| | - Claudia Barth
- Division for Mental Health and Substance Abuse, Diakonhjemmet Hospital, Oslo, Norway
| | - Liisa A M Galea
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
21
|
Zhang Y, Zou F, Liang L, Liu J, Li H, Liang S, Xu S, Guo Y, Lai J, Hu J, Tan B, Cao H. Suoquan Wan mitigates bladder overactivity via modulation of neuroimmune homeostasis in perimenopausal rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156450. [PMID: 39922151 DOI: 10.1016/j.phymed.2025.156450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 01/03/2025] [Accepted: 01/29/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND Overactive bladder (OAB) is a common disorder in perimenopausal women, involving detrusor regulation by bladder neuro-immune interactions. While Suoquan Wan (SQW) has shown efficacy in alleviating OAB symptoms, its underlying mechanisms remain unclear. PURPOSE The aim of this study was to investigate the therapeutic effects of SQW on perimenopausal overactive bladder (OAB) and elucidate its underlying mechanisms in restoring bladder neuro-immune homeostasis. METHODS Initially, vaginal smears were performed to identify perimenopausal rats. The effects of SQW on the structure and function of the perimenopausal bladder were assessed using urodynamic studies, organ bath assays, and histological analyses, including hematoxylin and eosin (HE) and Masson staining. Subsequently, single-nucleus RNA sequencing (snRNA-seq) was conducted to investigate the mechanisms by which SQW modulates neuron-macrophage interactions. To validate the sequencing data, PCR and immunofluorescence (IF) assays were utilized to evaluate neural homeostasis. Further analyses involved IF, flow cytometry, and fluorescence bead assays to determine the function and proportion of bladder macrophages. The co-localization of neurons and macrophages was visualized using IF. Finally, transwell co-culture experiments were carried out to explore the regulatory effects of SQW on bladder neuro-immune homeostasis. RESULTS SQW significantly attenuates the frequency of non-micturition contractions and reduces residual urine volume, alleviates detrusor hyperactivity in OAB rats in response to external stimuli such as EFS, CCh, and KCl, and markedly improves urinary efficiency in perimenopausal OAB rats. SnRNA-seq data indicate that SQW modulates bladder neuro-immune homeostasis in these rats. Furthermore, SQW obviously decreases the proportion of ChAT-positive nerve fibers while enhancing the abundance of nNOS and MAP-positive nerve fibers in the bladder. SQW also reduces the proportion of CD45+ CD11b/C+MHCⅡ+ macrophages in the bladder muscle layer. Transwell co-culture assays reveal that the effects of SQW on MAP-2 and ChAT are mediated through the neuron-macrophage interaction mechanism. CONCLUSIONS SQW alleviated perimenopausal OAB by specifically modulating the neuron-macrophage interaction and enhancing bladder neuro-immune homeostasis.
Collapse
Affiliation(s)
- Yao Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Feng Zou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Lang Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Jiaye Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Hongliang Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Shaochan Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Siyuan Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Yixin Guo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Junming Lai
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Jingyi Hu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Bo Tan
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Hongying Cao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
22
|
Colcimen N, Keskin S. Evaluation of the Protective Effects of Alpha Lipoic Acid on Bleomycin-Induced Ovarian Toxicity. J Biochem Mol Toxicol 2025; 39:e70230. [PMID: 40117335 DOI: 10.1002/jbt.70230] [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: 11/19/2024] [Revised: 02/10/2025] [Accepted: 03/07/2025] [Indexed: 03/23/2025]
Abstract
Chemotherapeutic drugs administered during cancer therapy, may lead to the depletion of ovarian follicles, and subsequent infertility in fertile patients. We aimed to determine toxic effects of bleomycin (BLM) on rat ovary, and to evaluate protective effects of alpha lipoic acid (ALA) on BLM toxicity. The total of 30 adult female rats were split into 4 groups. First, an intramuscular injection (i.m) of BLM (30 mg/m2) was administered to BLM and BLM + ALA groups except the control and ALA groups on the 1st, 8th and 15th days. The control group received 0.1 mL (i.m) saline on those days. BLM + ALA group received ALA (50 mg/kg) subcutaneously (s.c) for 2 weeks at the same time with BLM injections, and ALA group received ALA s.c for the same period. Ovarian tissues were evaluated by histopathological, stereological, immunohistochemical (StAR, VDAC2, Caspase-3, Bcl-2, and expression levels) and ELISA (AMH serum levels) methods. The vascular areas and collagen density increased in the medulla, and the volumes of medulla, cortex, and total ovary increased in BLM group, whereas these changes decreased in BLM + ALA group. On the other hand, VDAC2 and Caspase-3 expressions decreased, StAR and Bcl-2 expressions increased in BLM group, whereas VDAC2 and Caspase-3 expressions increased, and StAR and Bcl-2 expression levels significantly decreased in BLM + ALA group. Besides, follicle number and AMH levels decreased in the BLM group, but remarkably increased in the BLM + ALA group. We established that ALA may have ameliorative effects on the harmful effects of BLM on ovary.
Collapse
Affiliation(s)
- Nese Colcimen
- Department of Histology and Embryology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Seda Keskin
- Department of Histology and Embryology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| |
Collapse
|
23
|
Almeida-Souza T, Campos ACP, Rabelo TK, Emtyazi D, McCann E, Brandão-Lima P, Diwan M, Lipsman N, Hamani C. Sex differences in long-term fear and anxiety-like responses to deep brain stimulation in a preclinical model of PTSD. J Psychiatr Res 2025; 184:198-209. [PMID: 40056639 DOI: 10.1016/j.jpsychires.2025.02.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 01/22/2025] [Accepted: 02/20/2025] [Indexed: 03/10/2025]
Abstract
Deep brain stimulation (DBS) is currently being investigated in patients and preclinical models of posttraumatic stress disorder (PTSD), but differences in behaviour according to sex remain elusive. We exposed female and male rats to fear conditioning and extinction. Thereafter, animals were treated with ventromedial prefrontal cortex DBS, followed by a battery of tests to measure fear and anxiety-like behaviour. As in our prior work, animals with high freezing scores during extinction (weak extinction; WE) were segregated from those with lower freezing scores (non-weak extinction; nWE), since the former population was previously shown to develop prolonged fear and anxiety-like responses. Vaginal lavages were collected after fear extinction to study the estrous cycle. After the experiments, brains were processed for the measurement of estrogen (ER) and progesterone receptors (PR) in the hypothalamus and hippocampus. We found that DBS-treated males had a more pronounced reduction in freezing than females during all recall sessions. In females, DBS induced an anxiolytic-like effect in the open field, while a reduction in the latency to feed during novelty suppressed feeding was noticed in both sexes. Noteworthy, a reduction in freezing during recall and anxiolytic-like responses following DBS were observed in males of all phenotypes, but only in nWE females. While no effect of the estrous cycle was noticed on fear memory, DBS-treated females in metestrus/diestrus during extinction had a more prominent response in the elevated plus maze. A similar expression of ERα, ERβ and PRβ in the hypothalamus and hippocampus was found in DBS-treated females and controls.
Collapse
Affiliation(s)
| | | | | | - Delara Emtyazi
- . Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Eliza McCann
- . Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | | | - Mustansir Diwan
- . Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Nir Lipsman
- . Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada; . Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada; . Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada
| | - Clement Hamani
- . Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada; . Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada; . Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada.
| |
Collapse
|
24
|
Reichard TM, Miller CH, Yang J, Sheehan MJ. Seasonality of the estrus cycle in laboratory mice under constant conditions. Lab Anim 2025:236772251318772. [PMID: 40159114 DOI: 10.1177/00236772251318772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Seasonality governs every aspect of life in the natural environment. Controlled laboratory settings are intended to keep animals under a constant set of environmental cues with no seasonality. However, prior research suggests that seasonal variation may exist despite aseasonal lab environments. Here, we examined whether the length of each phase of the estrus cycle varied seasonally in addition to seasonal changes in the overall estrus cycle length in a laboratory mouse strain (C57BL/6J) under standard laboratory housing conditions. We found that female C57BL/6J mice exhibited reproductive seasonality mirroring the outside environment, in a controlled "simulated summer" environment. In the winter and spring, females have longer ovulating phases (proestrus and estrus), compared to the fall. Females similarly experience lengthier quiescent phases (metestrus and diestrus) in the summer, compared to fall and winter. Interestingly, females showed no significant variation in overall estrus cycle length across seasons. Notably, females spent more time in ovulating phases across seasons than previously reported. Laboratory mice are sensitive to external seasonal changes, even when housed in standard laboratory environments designed to control light, temperature, and humidity. Humidity is indicated by some analyses as a potential seasonal cue, however, we cannot rule out other unidentified external cues that may provide information about external seasonal changes. These findings represent just one example of how seasonality may impact mouse physiology in laboratory settings, emphasizing the need to account for such influences in biomedical research and improve environmental control in mouse holding facilities.
Collapse
Affiliation(s)
- Tess M Reichard
- Laboratory for Animal Social Evolution and Recognition, Department of Neurobiology and Behavior, Cornell University, Ithaca, USA
| | - Caitlin H Miller
- Laboratory for Animal Social Evolution and Recognition, Department of Neurobiology and Behavior, Cornell University, Ithaca, USA
- Department of Psychology, University of California-San Diego, La Jolla, CA 92093, USA
| | - Jay Yang
- Laboratory for Animal Social Evolution and Recognition, Department of Neurobiology and Behavior, Cornell University, Ithaca, USA
| | - Michael J Sheehan
- Laboratory for Animal Social Evolution and Recognition, Department of Neurobiology and Behavior, Cornell University, Ithaca, USA
| |
Collapse
|
25
|
Salinas P, Ponce N, del Sol M, Vásquez B. Impact of PM2.5 Exposure from Wood Combustion on Reproductive Health: Implications for Fertility, Ovarian Function, and Fetal Development. TOXICS 2025; 13:238. [PMID: 40278554 PMCID: PMC12031264 DOI: 10.3390/toxics13040238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/28/2025] [Accepted: 03/12/2025] [Indexed: 04/26/2025]
Abstract
This study evaluates the impact of PM2.5 exposure from wood combustion on reproductive health and fetal development using an experimental model in Sprague Dawley rats. The study was conducted in Temuco, Chile, where high levels of air pollution are primarily attributed to residential wood burning. A multigenerational exposure model was implemented using controlled exposure chambers with filtered (FA) and unfiltered (NFA) air. Second-generation (G2) female rats (n = 48) were exposed pregestationally (60 days) and gestationally (23 days) under four conditions: FA/FA, FA/NFA, NFA/FA, and NFA/NFA. PM2.5 concentration and composition were monitored using beta-ray attenuation and X-ray fluorescence spectrometry. Reproductive parameters, ovarian follicle counts, and hormonal levels were assessed via vaginal cytology, histological analysis, and chemiluminescence immunoassays. PM2.5 exposure disrupted estrous cyclicity (p = 0.0001), reduced antral and growing follicles (p = 0.0020; p = 0.0317), and increased post-implantation losses (p = 0.0149). Serum progesterone and estradiol levels were significantly altered (p < 0.05). Despite ovarian disruptions, fertility rates remained unchanged. These findings suggest that chronic exposure to wood smoke-derived PM2.5 adversely affects ovarian function and fetal growth without significantly impairing overall reproductive capacity. This study highlights the need for public health policies to mitigate wood smoke pollution.
Collapse
Affiliation(s)
- Paulo Salinas
- Laboratory of Animal & Experimental Morphology, Institute of Biology, Faculty of Sciences, Pontificia Universidad Católica de Valparaíso, Valparaíso 2374631, Chile
| | - Nikol Ponce
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile; (N.P.); (M.d.S.)
- Center of Excellence in Morphological and Surgical Studies, Universidad de La Frontera, Temuco 4811230, Chile;
| | - Mariano del Sol
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile; (N.P.); (M.d.S.)
- Center of Excellence in Morphological and Surgical Studies, Universidad de La Frontera, Temuco 4811230, Chile;
| | - Bélgica Vásquez
- Center of Excellence in Morphological and Surgical Studies, Universidad de La Frontera, Temuco 4811230, Chile;
- Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
| |
Collapse
|
26
|
Lord RA, Inglis MA, Juengel JL, Anderson GM. A Leptin Receptor Mutation Which Impairs Fertility in Ewes Causes Delayed Puberty in Male and Female Mice. Endocrinology 2025; 166:bqaf058. [PMID: 40130278 PMCID: PMC11979093 DOI: 10.1210/endocr/bqaf058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/19/2025] [Accepted: 03/23/2025] [Indexed: 03/26/2025]
Abstract
Reproductive function is tightly linked to nutritional status due to its high energetic demands. Leptin, a key adipose tissue-derived hormone signalling energy reserves to the brain, integrates metabolic status with the hypothalamic-pituitary-gonadal axis to ensure reproductive function is maintained or suppressed appropriately. Mutations in leptin or its receptor (LepR) are known to cause infertility and obesity in mice. In Davisdale ewes, 2 naturally occurring LepR mutations (R62C and P1019S) were associated with delayed puberty and subfertility, but their effects in males or in other species remain to be determined. This study examined the impact of analogous LepR mutations (A63C and P1018S) in mice using CRISPR-Cas9 gene editing. Puberty onset, adult fertility, and metabolic phenotypes were assessed in wild-type, heterozygous, and homozygous mutant mice. The A63C mutation, located in the extracellular domain of the receptor, resulted in increased body weight and adiposity in females, along with delays in puberty onset in both sexes. Despite these delays, adult reproductive function was maintained. Immunohistochemical analysis revealed no detectable reductions in leptin-induced pSTAT3, pERK1/2, or pmTOR signalling in the hypothalamic arcuate nucleus in either mutant line, indicating these pathways remain largely intact. These findings demonstrate the conserved importance of this region of the leptin receptor for puberty onset and adiposity across species, but also the resilience of leptin signalling in preserving reproductive function despite genetic variation.
Collapse
Affiliation(s)
- Rebecca A Lord
- Centre for Neuroendocrinology, and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin 9016, New Zealand
| | - Megan A Inglis
- Centre for Neuroendocrinology, and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin 9016, New Zealand
| | - Jennifer L Juengel
- Agricultural Systems and Reproduction, AgResearch Ltd, Invermay Agricultural Centre, Mosgiel 9092, New Zealand
| | - Greg M Anderson
- Centre for Neuroendocrinology, and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin 9016, New Zealand
| |
Collapse
|
27
|
Bindels J, Squatrito M, Bernet L, Nisolle M, Munaut C. Ovarian cryopreservation with rapamycin improves fertility restoration in a murine orthotopic transplantation model. Sci Rep 2025; 15:9441. [PMID: 40108278 PMCID: PMC11923166 DOI: 10.1038/s41598-025-94588-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 03/14/2025] [Indexed: 03/22/2025] Open
Abstract
Currently, the only fertility preservation option of prepubertal patients is ovarian tissue cryopreservation followed by autotransplantation (OTCTP). Once in remission and patients desire to conceive, autotransplantation of frozen/thawed tissue is performed. A major issue of this technique is follicular loss directly after transplantation, mainly due to follicle activation. Our previous research showed that adding rapamycin to the freezing medium counteracted follicle proliferation and activation induced by OTCTP in heterotopic autotransplantation of ovaries in mice. Our current study aimed to test the potential of this approach to improve fertility restoration in mice. Forty 4-week-old female C57BL/6 mice underwent unilateral oophorectomy followed by slow-freezing of ovaries with or without rapamycin. After chemically disabling the remaining ovary, orthotopic autotransplantation was performed. After recovery, estrous cycle analysis was conducted using daily vaginal smears. The mice were mated with males for 4 months, and pregnancy outcomes were recorded. After mating, half the females were super-ovulated for oocyte quantification and ovarian analysis, while the others had their ovaries collected for analysis of remaining primordial follicles using immunohistochemistry. Female mice whose ovaries were cryopreserved with rapamycin prior to chemically disabling the remaining ovary and orthotopic autotransplantation, gave birth to more pups (102 rapamycin, 48 control). The live birth rate was also higher (P = 0.0025) when ovaries were cryopreserved in rapamycin compared to control medium. Additionally, more mice in the rapamycin group gave birth (13 rapamycin, 8 control) with a higher average litter size (P = 0.0837). More mice had primordial follicles left at the end of the experiment in the rapamycin group (P = 0.0397). Superovulation showed a similar number of oocytes collected (P = 0.4462). While rapamycin did not influence cyst formation after autotransplantation, mice that developed ovarian cysts gave birth to fewer pups per dam (P = 0.0119) with a lower live birth rate compared to mice without ovarian cysts (P = 0.0032). The use of rapamycin improved fertility restoration in mice. Using rapamycin during OTCTP in humans could potentially resolve the massive follicular loss directly after grafting, and thus eventually lead to better opportunities for women to become pregnant.
Collapse
Affiliation(s)
- Jules Bindels
- Laboratory of Biology of Tumor and Development, GIGA-Cancer, Université de Liège, 4000, Liège, Belgium
| | - Marlyne Squatrito
- Laboratory of Biology of Tumor and Development, GIGA-Cancer, Université de Liège, 4000, Liège, Belgium
| | - Laëtitia Bernet
- Laboratory of Biology of Tumor and Development, GIGA-Cancer, Université de Liège, 4000, Liège, Belgium
| | - Michelle Nisolle
- Department of Obstetrics and Gynecology, Hôpital de la Citadelle, Université de Liège, 4000, Liège, Belgium
| | - Carine Munaut
- Laboratory of Biology of Tumor and Development, GIGA-Cancer, Université de Liège, 4000, Liège, Belgium.
| |
Collapse
|
28
|
Abd-Elkareem M, Alnasser SM, Meshal A, Kotob MH, Amer AS, Abdullah RI, Ali AU. The effect of norethisterone acetate on the uterine telocytes, immune cells and progesterone receptors in albino rats. Sci Rep 2025; 15:8997. [PMID: 40089502 PMCID: PMC11910565 DOI: 10.1038/s41598-025-92354-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/27/2025] [Indexed: 03/17/2025] Open
Abstract
This study is the first attempt to examine the effects of NETA on immune cells and telocytes. The results of this study form an important knowledge base for the development of new information on the mechanism of contraceptive action of NETA in the uterus. Norethisterone acetate (NETA) is a synthetic progestogen medication commonly utilized in birth control pills, menopausal hormone therapy, and for curing abnormal uterine bleeding and endometriosis. Furthermore NETA has many beneficial uses in veterinary medicine as control and synchronization of estrous cycle. The impact of NETA on the endometrial stromal cells (ESCs), telocytes, and uterine immune cells is not well understood. Therefore, this study focuses on assessing changes in uterine immune cells, ESCs, and telocytes following exposure to NETA in albino rats. To achieve this objective, fourteen adult female albino rats were randomly divided into two groups: a control group and an NETA-treated group. Rats in the control group received daily pelleted food, water, and were oral administered of 2 ml distilled water. In contrast, rats in the NETA-treated group received daily pelleted food, water, and were orally administered 20 µg of NETA dissolved in 2 ml distilled water. The experiment spanned three weeks. The findings of this study revealed that NETA usage increases the infiltration and activity of immune cells (eosinophils, neutrophils, macrophages, lymphocytes, and mast cells). Furthermore, it enhances the vesicular activity of uterine telocytes and their communication with various immune cells. NETA also influences decidualization and the immunoexpression of progesterone receptors in uterine epithelial and immune cells. This study concludes that the primary mechanism by which NETA controls pregnancy is through decidual (pregnancy-like) effects or improper decidualization, which inhibits fertilization and implantation respectively. Our research provides evidence of the contraceptive mechanism of NETA from an immunological perspective in an animal model.
Collapse
Affiliation(s)
- Mahmoud Abd-Elkareem
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt.
| | - Sulaiman Mohammed Alnasser
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, 52571, Buraydah, Saudi Arabia
| | - Alotaibi Meshal
- Pharmacy Practice, College of Pharmacy, University of Hafr Albatin, Hafr Albatin,, Saudi Arabia
| | - Mohamed H Kotob
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090, Vienna, Austria
- Department of Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Ayman S Amer
- Department of Human Anatomy and Embryology, Faculty of Medicine, Assiut University, Assiut, 71526, Egypt
| | - Raghda Ismail Abdullah
- Department of Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, New Valley University, El Kharga, Egypt
| | - Ahmed U Ali
- Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
29
|
Zhao ZW, Wang YC, Chen PC, Tzeng SF, Chen PS, Kuo YM. Dopamine D1 receptor agonist alleviates post-weaning isolation-induced neuroinflammation and depression-like behaviors in female mice. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2025; 21:6. [PMID: 40065395 PMCID: PMC11895232 DOI: 10.1186/s12993-025-00269-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Major depressive disorder is a significant global cause of disability, particularly among adolescents. The dopamine system and nearby neuroinflammation, crucial for regulating mood and processing rewards, are central to the frontostriatal circuit, which is linked to depression. This study aimed to investigate the effect of post-weaning isolation (PWI) on depression in adolescent mice, with a focus on exploring the involvement of microglia and dopamine D1 receptor (D1R) in the frontostriatal circuit due to their known links with mood disorders. RESULTS Adolescent mice underwent 8 weeks of PWI before evaluating their depression-like behaviors and the activation status of microglia in the frontostriatal regions. Selective D1-like dopamine receptor agonist SKF-81,297 was administered into the medial prefrontal cortex (mPFC) of PWI mice to assess its antidepressant and anti-microglial activation properties. The effects of SKF-81,297 on inflammatory signaling pathways were examined in BV2 microglial cells. After 8 weeks of PWI, female mice exhibited more severe depression-like behaviors than males, with greater microglial activation in the frontostriatal regions. Microglial activation in mPFC was the most prominent among the three frontostriatal regions examined, and it was positively correlated with the severity of depression-like behaviors. Female PWI mice exhibited increased expression of dopamine D2 receptors (D2R). SKF-81,297 treatment alleviated depression-like behaviors and local microglial activation induced by PWI; however, SKF-81,297 induced these alterations in naïve mice. In vitro, SKF-81,297 decreased pro-inflammatory cytokine release and phosphorylations of JNK and ERK induced by lipopolysaccharide, while in untreated BV2 cells, SKF-81,297 elicited inflammation. CONCLUSIONS This study highlights a sex-specific susceptibility to PWI-induced neuroinflammation and depression. While targeting the D1R shows potential in alleviating PWI-induced changes, further investigation is required to evaluate potential adverse effects under normal conditions.
Collapse
Affiliation(s)
- Zi-Wei Zhao
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yun-Chen Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Pei-Chun Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Shun-Fen Tzeng
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Po-See Chen
- Department of Psychiatry, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, 70101, Taiwan
- Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yu-Min Kuo
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
30
|
Khatiz A, Tomlinson C, Ruzhytska B, Croft EK, Amrani A, Dunn S, Mendrek A, Gris D. Real-time behavioral monitoring of C57BL/6J mice during reproductive cycle. Front Neurosci 2025; 19:1509822. [PMID: 40098987 PMCID: PMC11911481 DOI: 10.3389/fnins.2025.1509822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction The present study aims to identify differences in behavioral profiles in post-pubertal C57BL/6J males and female mice across distinct phases of the reproductive cycle in a home cage environment. Methods To reduce human bias, we used an automated behavioral analysis system HomeCageScan from CleverSys Inc. Mice were monitored continuously, and resulting data were summarized across 24-h, light, and dark cycles. Behavioral activities of each period were analyzed using hierarchical clustering, factor analysis, and principal component analysis. Results Females exhibited higher levels of physically demanding activities, including ambulatory and exploratory movements, particularly during estrus and metestrus, with estrus showing up to 30% more activity than males. In contrast, males consistently engaged in more sleep-related behaviors across all phases, with significantly higher engagement during the light cycle compared to females in proestrus and estrus (p < 0.0001); the extent of this sex difference was greater during proestrus and estrus than in metestrus and diestrus (p < 0.01). Notably, distinct patterns of sleep fragmentation were observed, with females experiencing greater disruptions during the light cycle, while males showed similar disruptions during the dark cycle. Feeding and resourcing behaviors were highest in males, showing up to 20% increase compared to cycling females, as well as significantly engaging in habituation-related behaviors such as feeding and digging. Interphase differences were observed within females, such as a significant increase of habituation-related activities during estrus compared to proestrus and diestrus (p < 0.05), while during the dark cycle, these activities peaked during the diestrus phase (p < 0.05). Female mice in the metestrus phase exhibited more sleep-related behaviors than those in proestrus. Discussion Our study has revealed prevalent behavioral differences due to sex, and inter-phase variations by employing a continuous monitoring approach designed to reduce bias. This methodology ensures a comprehensive understanding of natural behavioral patterns and strategies.
Collapse
Affiliation(s)
- Ariane Khatiz
- Program of Physiology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Cassidy Tomlinson
- Program of Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Bohdana Ruzhytska
- Program of Translational Medical Bioengineering, National Technical University of Ukraine, Kyiv, Ukraine
| | - Erika Kathe Croft
- Department of Pediatrics, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Abdelaziz Amrani
- Department of Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Shannon Dunn
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Biological Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Adrianna Mendrek
- Department of Psychology, Bishop’s University, Sherbrooke, QC, Canada
| | - Denis Gris
- Department of Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
- Department of Physiology and Pharmacology, University of Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
31
|
Brunette MA, Wall MA, Sinko D, Machlin JH, Blevins GM, Leo M, Tan A, Ray B, Cascalho M, Padmanabhan V, Shikanov A. Restoration of ovarian endocrine function with encapsulated immune isolated human ovarian xenograft in ovariectomized mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640622. [PMID: 40093140 PMCID: PMC11908119 DOI: 10.1101/2025.02.27.640622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Anti-cancer treatments cause premature depletion of the non-renewable ovarian reserve of follicles, the source of key steroid hormones, leading to premature ovarian insufficiency (POI) in 50% of pediatric cancer survivors. Patients with POI, especially at the onset of pubertal development, experience significant endocrine complications, including delayed growth, elevated risks of obesity and diabetes, and accelerated cardiovascular, musculoskeletal and neurological disorders as adults. The only approved pharmacological treatment for POI is an off-label prescribed hormone replacement therapy, which does not replace physiologically functioning ovaries. To restore production of ovarian hormones and protect against immune-mediated injury, we developed a hydrogel-based capsule for implantation of donor ovarian tissue. We evaluated the restoration of ovarian endocrine function in ovariectomized immunodeficient (NOD scid gamma, NSG) mice implanted with encapsulated xenografts over 20 weeks through daily vaginal cytology, hormone measurements and histological analysis of explanted human xenografts. The encapsulated xenografts integrated into the murine hypothalamus-pituitary-gonad (HPG) axis responding to circulating murine gonadotropins and restoring ovarian endocrine function. As controls, we implanted non encapsulated human ovarian xenografts comparable in size. Without the need for exogeneous stimulation, the estrous cyclicity resumed in both groups of mice 12 weeks post implantation and all mice regularly cycled experiencing between 3 to 8 estrous cycles in 20 weeks. The levels of estradiol gradually increased reaching on average 50pg/mL 20 weeks post implantation. Morphological analysis of the encapsulated grafts revealed presence of large antral follicles, ∼3mm in diameter, consistent with regular cyclicity and measurable levels of circulating hormones. This work demonstrates that endocrine function of encapsulated human ovarian tissue was not affected by the encapsulation and integrated with the host physiology similarly to the non-encapsulated controls.
Collapse
|
32
|
Bishnoi IR, Bordt EA. Sex and Region-Specific Differences in Microglial Morphology and Function Across Development. NEUROGLIA (BASEL, SWITZERLAND) 2025; 6:2. [PMID: 40181886 PMCID: PMC11967618 DOI: 10.3390/neuroglia6010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Microglia are exceptionally dynamic resident innate immune cells within the central nervous system, existing on a continuum of morphologies and functions throughout their lifespan. They play vital roles in response to injuries and infections, clearing cellular debris, and maintaining neural homeostasis throughout development. Emerging research suggests that microglia are strongly influenced by biological factors, including sex, developmental stage, and their local environment. This review synthesizes findings on sex differences in microglial morphology and function in key brain regions, including the frontal cortex, hippocampus, amygdala, hypothalamus, basal ganglia, and cerebellum, across the lifespan. Where available, we examine how gonadal hormones influence these microglial characteristics. Additionally, we highlight the limitations of relying solely on morphology to infer function and underscore the need for comprehensive, multimodal approaches to guide future research. Ultimately, this review aims to advance the dialogue on these spatiotemporally heterogeneous cells and their implications for sex differences in brain function and vulnerability to neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Indra R. Bishnoi
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Evan A. Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
33
|
Tucker JAL, McCarthy SF, Bornath DPD, Khoja JS, Hazell TJ. The Effect of the Menstrual Cycle on Energy Intake: A Systematic Review and Meta-analysis. Nutr Rev 2025; 83:e866-e876. [PMID: 39008822 PMCID: PMC11819481 DOI: 10.1093/nutrit/nuae093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024] Open
Abstract
CONTEXT Energy intake may differ across the menstrual cycle, with some studies identifying greater energy intake in the luteal phase (LP) compared with the follicular phase (FP) and others finding no clear differences. To date, no study has systematically synthesized the available data to draw more definite conclusions while considering any methodological inconsistencies between studies. OBJECTIVE The aim was to conduct a systematic review/meta-analysis in an effort to determine if there are differences in energy intake between the FP and LP. DATA SOURCES A systematic search strategy was developed and the search was conducted in 5 databases for studies that investigated any changes in energy intake across menstrual phases. DATA EXTRACTION Using Covidence, studies were identified and included if they contained individuals between the ages of 18 and 45 years, maintained an average body mass index (BMI) of 18.5-25 kg/m2, had no history of disordered eating, and included energy intake and menstrual cycle measurements in the FP and LP. DATA ANALYSIS Effect sizes were calculated for each study and a random-effects model was used to pool the results of each study. RESULTS Fifteen datasets were included consisting of 330 female participants with a mean age of 26 ± 4 years and mean BMI of 22.4 ± 2.3 kg/m2. Overall, there was a statistically significant difference (standardized mean difference = 0.69; P = .039) with increased energy intake in the LP compared with the FP (crude 168 kcal⋅d-1 average difference between phases). CONCLUSION Energy intake was found to be greater in the LP compared with the FP, providing insight into the effect of the menstrual cycle on energy intake. However, there were repeated methodological inconsistencies and future work should strive to utilize best practices for both energy intake measurement and menstrual phase specification.
Collapse
Affiliation(s)
- Jessica A L Tucker
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, Waterloo, ON N2L 3C5, Canada
| | - Seth F McCarthy
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, Waterloo, ON N2L 3C5, Canada
| | - Derek P D Bornath
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, Waterloo, ON N2L 3C5, Canada
| | - Jenna S Khoja
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, Waterloo, ON N2L 3C5, Canada
| | - Tom J Hazell
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, Waterloo, ON N2L 3C5, Canada
| |
Collapse
|
34
|
Krivoshein G, Rivera-Mancilla E, MaassenVanDenBrink A, Giniatullin R, van den Maagdenberg AMJM. Sex difference in TRPM3 channel functioning in nociceptive and vascular systems: an emerging target for migraine therapy in females? J Headache Pain 2025; 26:40. [PMID: 39994546 PMCID: PMC11853570 DOI: 10.1186/s10194-025-01966-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Transient Receptor Potential Melastatin 3 (TRPM3) channels are Ca2+ permeable ion channels that act as polymodal sensors of mechanical, thermal, and various chemical stimuli. TRPM3 channels are highly expressed in the trigeminovascular system, including trigeminal neurons and the vasculature. Their presence in dural afferents suggests that they are potential triggers of migraine pain, which is originating from the meningeal area. This area is densely innervated by autonomous and trigeminal nerves that contain the major migraine mediator calcitonin gene-related peptide (CGRP) in peptidergic nerve fibers. Co-expression of TRPM3 channels and CGRP receptors in meningeal nerves suggests a potential interplay between both signalling systems. Compared to other members of the TRP family, TRPM3 channels have a high sensitivity to sex hormones and to the endogenous neurosteroid pregnenolone sulfate (PregS). The predominantly female sex hormones estrogen and progesterone, of which the levels drop during menses, act as natural inhibitors of TRPM3 channels, while PregS is a known endogenous agonist of these channels. A decrease in sex hormone levels has also been suggested as trigger for attacks of menstrually-related migraine. Notably, there is a remarkable sex difference in TRPM3-mediated effects in trigeminal nociceptive signalling and the vasculature. In line with this, the relaxation of human isolated meningeal arteries induced by the activation of TRPM3 channels is greater in females. Additionally, the sex-dependent vasodilatory responses to CGRP in meningeal arteries seem to be influenced by age-related hormonal changes, which could contribute to sex differences in migraine pathology. Consistent with these observations, activation of TRPM3 channels triggers nociceptive sensory firing much more prominently in female than male mouse meninges, suggesting that pain processing in female patients with migraine may differ. Overall, the combined TRPM3-related neuronal and vascular mechanisms could provide a possible explanation for the higher prevalence and even the more severe quality of migraine attacks in females. This narrative review summarizes recent data on the sex-dependent roles of TRPM3 channels in migraine pathophysiology, the potential interplay between TRPM3 and CGRP signalling, and highlights the prospects for translational therapies targeting TRPM3 channels, which may be of particular relevance for women with migraine.
Collapse
Affiliation(s)
- Georgii Krivoshein
- Departments of Human Genetics and Neurology, Leiden University Medical Center, PO Box 9600 2300 RC, Leiden, The Netherlands
| | - Eduardo Rivera-Mancilla
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Antoinette MaassenVanDenBrink
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Rashid Giniatullin
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Arn M J M van den Maagdenberg
- Departments of Human Genetics and Neurology, Leiden University Medical Center, PO Box 9600 2300 RC, Leiden, The Netherlands.
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
35
|
Maleki MH, Miladpour B, Mazhari SA, Far MH, Rajabi M, Alinejad M, Dehghanian A, Beigmohammadi F, Esmaeli N, Siri M, Aryanian Z. Exploring Isotretinoin's Unexpected Acceleration of wound Healing: A rat model study. Int Immunopharmacol 2025; 148:114145. [PMID: 39889411 DOI: 10.1016/j.intimp.2025.114145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 01/05/2025] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND There have been clinical observations indicating that wound healing could be affected in patients undergoing systemic isotretinoin treatment. However, the precise role of retinoids in wound healing is still unclear and controversial. It is generally assumed that systemic retinoids could be harmful to wound healing, but this requires further investigation. METHODS Sprague-Dawley rats were gavaged with 2 mg/Kg/day of Isotretinoin and divided into three groups: Control, Isotretinoin/1month and Isotretinoin/2month. Photographic documentation and histomorphometric investigation were performed. The mRNA expressions of IL-6, MCP-1, VEGF, ICAM1, L-Selectin, TGF-1β, IL-10, IL-1α, and IL-8 were examined by qRT-PCR. RESULTS There was no significant impact on the rate of wound closure in Isotretinoin/1month group. However, a two-month regimen accelerated the wound-healing process. RT-PCR results revealed increased expression of IL-6, IL-8, IL-1α, TGF-β1, IL-10 MCP-1, ICAM1, L-Selectin, and VEGF rats that were administered Isotretinoin. Histological observations showed an increased number of mast cells in the wound areas of rats treated with Isotretinoin. CONCLUSION Our research indicated that taking Isotretinoin did not slow down wound healing and may even help the growth phase. Additionally, we did not observe any keloid formation during our histopathological analysis, suggesting that it may not be necessary to postpone invasive surgical procedures for six months after Isotretinoin therapy.
Collapse
Affiliation(s)
- Mohammad Hasan Maleki
- Autoimmune Bullous Diseases Research Center Razi Hospital Tehran University of Medical Sciences Tehran Iran; Endocrinology and Metabolism Research Center Shiraz University of Medical Science Shiraz Iran
| | - Behnoosh Miladpour
- Department of Clinical Biochemistry Fasa University of Medical Sciences Fasa Iran
| | | | - Mohammad Hojjati Far
- Department of physiology School of Medicine Shiraz University of Medical Sciences Shiraz Iran
| | - Mahsa Rajabi
- Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Mahdi Alinejad
- Department of Gastroenterology Kerman University of Medical Sciences Kerman Iran
| | - Amirreza Dehghanian
- Trauma Research Center Shiraz University of Medical Sciences Shiraz Iran; Molecular Pathology and Cytogenetics Division Department of Pathology School of Medicine Shiraz University of Medical Sciences Shiraz Iran
| | - Fereshteh Beigmohammadi
- Autoimmune Bullous Diseases Research Center Razi Hospital Tehran University of Medical Sciences Tehran Iran
| | - Nafiseh Esmaeli
- Autoimmune Bullous Diseases Research Center Razi Hospital Tehran University of Medical Sciences Tehran Iran; Department of Dermatology Razi Hospital School of Medicine Tehran University of Medical Sciences Iran
| | - Morvarid Siri
- Endocrinology and Metabolism Research Center Shiraz University of Medical Science Shiraz Iran; Autophagy Research Centre Shiraz University of Medical Sciences Shiraz Iran.
| | - Zeinab Aryanian
- Autoimmune Bullous Diseases Research Center Razi Hospital Tehran University of Medical Sciences Tehran Iran; Department of Dermatology Babol University of Medical Sciences Babol Iran.
| |
Collapse
|
36
|
Gilfarb RA, Ranade S, Dybas E, Biddle A, Stewart M, Rajesh A, Leuner B, Lenz KM. Hormonal contraceptives in adolescence impact the neuroimmune environment of the medial prefrontal cortex and hippocampus in female rats. Brain Behav Immun 2025; 127:315-328. [PMID: 39978694 DOI: 10.1016/j.bbi.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 02/02/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Adolescence is a period of protracted neurodevelopment, during which the prefrontal cortex (PFC) undergoes significant remodeling. Microglia are integral to neurodevelopment and are sensitive to gonadal hormones, which increase during adolescence. Microglia and gonadal hormones can interact to influence adolescent development of the PFC (or medial prefrontal cortex [mPFC] in rodents). In females, gonadal hormones can be perturbed by using hormonal contraceptives (HCs). We predicted that HC administration over adolescence could affect microglia, other immunocompetent cells, and the neuroimmune environment of the developing mPFC. We also assessed HC effects on neuroimmune measures in the hippocampus, as the hippocampus also matures throughout adolescence and is sensitive to ovarian hormones. Intact post-pubertal female Sprague-Dawley rats received daily subcutaneous injections of vehicle or 10 ug ethinyl estradiol + 20 ug levonorgestrel (HCs) throughout adolescence from postnatal day (PND) 35-56. On PND 57 or 58, brains were collected for immunohistochemistry and qPCR. In the mPFC, HC-treated rats showed less Iba1 (microglia) immunolabeling and fewer Iba1+ cells. HC treatment also altered microglia morphology and reduced the spacing between microglia in the mPFC. In the hippocampus, HC-treated rats had reduced Iba1 immunolabeling in the dorsal CA1 and reductions in microglial cell complexity in dorsal CA1, ventral CA1, and ventral CA3. There were no effects of HCs on GFAP (astrocyte) immunolabeling in the mPFC or on astrocytes in any hippocampal subregion analyzed, except an increase in astrocyte number in the dorsal dentate gyrus. mPFC expression of genes related to phagocytosis (Cd68, Trem2) and neuroimmune signaling (Cx3cr1, Cx3cl1) were reduced in rats treated with HCs, but no gene expression changes were seen in the hippocampus. These data provide the first evidence that HCs given during the critical developmental period of adolescence can affect microglia properties in limbic brain regions.
Collapse
Affiliation(s)
- Rachel A Gilfarb
- Neuroscience Graduate Program, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA
| | - Sanjana Ranade
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Elizabeth Dybas
- Neuroscience Graduate Program, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA
| | - Abigail Biddle
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Meredith Stewart
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Abhishek Rajesh
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Benedetta Leuner
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, 460 W. 12th Ave, Columbus, OH 43210, The Ohio State University, USA
| | - Kathryn M Lenz
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, 460 W. 12th Ave, Columbus, OH 43210, The Ohio State University, USA.
| |
Collapse
|
37
|
Dufresne K, Al KF, Craig HC, Coleman CEM, Kasper KJ, Burton JP, McCormick JK. TSST-1 promotes colonization of Staphylococcus aureus within the vaginal tract by activation of CD8 + T cells. Infect Immun 2025; 93:e0043924. [PMID: 39840991 PMCID: PMC11834441 DOI: 10.1128/iai.00439-24] [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: 10/02/2024] [Accepted: 12/14/2024] [Indexed: 01/23/2025] Open
Abstract
Toxic shock syndrome toxin-1 (TSST-1) is a superantigen produced by Staphylococcus aureus and is the determinant of menstrual toxic shock syndrome (mTSS); however, the impact of TSST-1 on the vaginal environment beyond mTSS is not understood. Herein, we assessed how TSST-1 affects vaginal colonization by S. aureus, host inflammatory responses, and changes in microbial communities within the murine vagina. We demonstrated that TSST-1 induced a CD8+ T-cell-dependent inflammatory response in 24 h that correlated with S. aureus persistence within the vaginal tract. This increase was due to superantigen-dependent T-cell activation that triggered a change in microbial composition within the vaginal tract. Altogether, this study demonstrates that within the vaginal tract, TSST-1 modulates the vaginal microbiota to favor the survival of S. aureus in the absence of mTSS.IMPORTANCEToxic shock syndrome toxin-1 (TSST-1) is a superantigen toxin produced from Staphylococcus aureus that causes the menstrual form of toxic shock syndrome. This research demonstrates that TSST-1 also has a wider function within the vaginal tract than previously expected. We show that TSST-1, by activating CD8+ T cells, induces an inflammatory environment that modifies the vaginal microbiota to favor colonization by S. aureus. These are important findings as S. aureus can colonize the human vaginal tract efficiently and subsequently trigger dysbiosis within the microbial communities leading to several adverse outcomes such as decreased fertility, increased risks for sexually transmitted diseases, and issues related to pregnancy and birth.
Collapse
Affiliation(s)
- Karine Dufresne
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Kait F. Al
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotics Research, Lawson Health Research Institute, London, Ontario, Canada
| | - Heather C. Craig
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Charlotte E. M. Coleman
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Katherine J. Kasper
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Jeremy P. Burton
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotics Research, Lawson Health Research Institute, London, Ontario, Canada
| | - John K. McCormick
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotics Research, Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
38
|
Rousselle D, Silveyra P. Acute Exposure to Ozone Affects Circulating Estradiol Levels and Gonadotropin Gene Expression in Female Mice. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2025; 22:222. [PMID: 40003448 PMCID: PMC11855596 DOI: 10.3390/ijerph22020222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025]
Abstract
Ozone, a critical air pollutant, has been shown to lead to systemic inflammation that can alter bodily functions, including hormone secretion, fertility, and the hypothalamic-pituitary-gonadal (HPG) axis. This study aimed to quantify changes in hormone production and follicle development after acute exposure to ozone using an animal model to identify the potential mechanisms underlying the observed effects of air pollution exposures on fertility and hormone secretion. To accomplish this, regularly cycling 8-week-old female C57BL/6J mice were exposed to 2 ppm of ozone or filtered air (control) for 3 h on the day of proestrus. Blood, ovaries, brain tissues, and pituitary glands were collected at 4 h after exposure to evaluate hormone levels, ovarian follicle distribution, and gene expression. Ovaries were also harvested at 24 h post-exposure. We found that at 4 h after ozone exposure, mice had significantly higher (30%) circulating estradiol levels than mice exposed to filtered air. This effect was accompanied by a decrease in mRNA expression of gonadotropin genes (LH, FSH) and gonadotropin-releasing hormone in the pituitary gland. Analysis of ovarian tissue at 4 h and 24 h after exposure showed no significant changes in follicle composition or the expression of steroidogenesis genes. We conclude that acute ozone exposure affects sex hormone levels and disrupts the HPG axis. Future studies addressing chronic or long-term effects of air pollution exposure are needed to elucidate the mechanisms by which ambient ozone affects endocrine function.
Collapse
Affiliation(s)
- Dustin Rousselle
- Department of Environmental and Occupational Health, School of Public Health, Indiana University, Bloomington, IN 47405, USA;
| | - Patricia Silveyra
- Department of Environmental and Occupational Health, School of Public Health, Indiana University, Bloomington, IN 47405, USA;
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
39
|
Amir M, Yimer N, Hiew M, Yusoff MSM, Babatunde SM, Quddus A. Edible Bird's Nest (EBN) Ameliorates the Effects of Indomethacin (IMC)-Induced Embryo Implantation Dysfunction in Rats. BIOLOGY 2025; 14:159. [PMID: 40001927 PMCID: PMC11851620 DOI: 10.3390/biology14020159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 02/27/2025]
Abstract
IMC has been reported to influence embryo implantation negatively in animals including rats. While EBN has been known to have a potential protective effect against reproductive toxicity, there is limited study on the effect of EBN on IMC toxicity in reproduction. This study aimed to ascertain whether pretreatment with a natural substance, Edible Bird's Nest (EBN), will reduce IMC-induced toxicity in pregnant rats. Thirty Sprague-Dawley rats divided into five equal groups were treated with EBN and IMC as follows: G1 = Control, G2 = IMC (4.33 mg/kg), G3 = IMC + EBN (4.33 mg/kg + 60 mg/kg), G4 = IMC + EBN (4.33 mg/kg + 90 mg/kg), and G5 = IMC + EBN (4.33 mg/kg +120 mg/kg). EBN was administered once daily for 8 weeks while IMC was injected subcutaneously. On day 8 after mating, all rats were sacrificed for blood sampling and embryo implantation rate (EIR) assessment; the uterine tissues were also subjected to immunohistochemical and histological analyses. G5 recorded significantly higher EIR, fertility index, and expression of epidermal growth factor receptor (EGFR) in the uterine section, across stroma cells, the glandular epithelium, and the luminal epithelium compared to control and other groups. IMC-induced inflammatory alterations, endometrial atrophy, vacuolar degeneration, and atrophy were not detected in uterine tissue sections in G4 and G5, with the latter group demonstrating the highest EIR with protective effects on uterine tissues. Thus, EBN supplementation might be of great benefit in guarding the fertility of individuals who depend on IMC for the treatment of chronic inflammatory illness.
Collapse
Affiliation(s)
- Maria Amir
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.A.); (M.H.)
- Department of Physiology and Biochemistry, Faculty of Veterinary and Animal Sciences, Ziauddin University, Karachi 75600, Sindh, Pakistan
| | - Nurhusien Yimer
- Department of Veterinary Sciences, School of Medicine, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
- Department of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, East Java, Indonesia
| | - Mark Hiew
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.A.); (M.H.)
| | - Md Sabri Mohd Yusoff
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Sadiq Mohammed Babatunde
- Department of Farm and Exotic Animal Medicine and Surgery, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Abdul Quddus
- Faculty of Veterinary and Animal Science, Lasbela University of Agriculture Water and Marine Science, Uthal 90150, Balochistan, Pakistan;
| |
Collapse
|
40
|
Tso P, Bernier-Latmani J, Petrova TV, Liu M. Transport functions of intestinal lymphatic vessels. Nat Rev Gastroenterol Hepatol 2025; 22:127-145. [PMID: 39496888 DOI: 10.1038/s41575-024-00996-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 11/06/2024]
Abstract
Lymphatic vessels are crucial for fluid absorption and the transport of peripheral immune cells to lymph nodes. However, in the small intestine, the lymphatic fluid is rich in diet-derived lipids incorporated into chylomicrons and gut-specific immune cells. Thus, intestinal lymphatic vessels have evolved to handle these unique cargoes and are critical for systemic dietary lipid delivery and metabolism. This Review covers mechanisms of lipid absorption from epithelial cells to the lymphatics as well as unique features of the gut microenvironment that affect these functions. Moreover, we discuss details of the intestinal lymphatics in gut immune cell trafficking and insights into the role of inter-organ communication. Lastly, we highlight the particularities of fat absorption that can be harnessed for efficient lipid-soluble drug distribution for novel therapies, including the ability of chylomicron-associated drugs to bypass first-pass liver metabolism for systemic delivery. In all, this Review will help to promote an understanding of intestinal lymphatic-systemic interactions to guide future research directions.
Collapse
Affiliation(s)
- Patrick Tso
- Department of Pathology & Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA.
| | - Jeremiah Bernier-Latmani
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Min Liu
- Department of Pathology & Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
41
|
Pierce-Messick Z, Shipman ML, Desilets GL, Corbit LH. Outcome devaluation as a method for identifying goal-directed behaviors in rats. Nat Protoc 2025; 20:518-538. [PMID: 39443709 DOI: 10.1038/s41596-024-01054-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/24/2024] [Indexed: 10/25/2024]
Abstract
Goal-directed behaviors allow animals to act to satisfy needs and desires. The outcome devaluation task is an effective method for identifying goal-directed behaviors and distinguishing these from other types of behavior. Rats can be trained to lever-press for one or multiple distinct food rewards. During testing, the previously earned food-or a control food for comparison-is devalued by allowing the animal to freely feed on it until they are sated before testing lever-press performance under extinction conditions (no rewards are delivered). Behavior that adapts to reflect the new value of the outcome is considered goal-directed, whereas behavior that continues as in previous training despite the change in outcome value, is not. As more research groups have used this task, variability in the procedures used has increased. Here, we provide a reliable procedure for conducting the outcome devaluation task with appropriate controls. We describe the most common variants of the task and control conditions and discuss troubleshooting measures such as outcome pre-exposure, habituation to pre-feeding chambers and attention to animals' hunger levels. The method outlined can be executed in ~2 weeks including training (~8 d) and testing (1-4 d) by researchers who are familiar with performing behavioral tasks in laboratory rodents, although longer training may be considered for those who are interested in observing habitual control of behavior. This protocol should facilitate the comparison of results from different studies and laboratories, while allowing flexibility in the application of the outcome devaluation task to different research questions.
Collapse
Affiliation(s)
| | - Megan L Shipman
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | | | - Laura H Corbit
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
42
|
Kacar E, Oz ZD, Serhatlioglu I, Kaya Tektemur N, Ozdede MR, Yalcin T, Ozbeg G, Ozgen A, Tan F, Orhan SU, Zorlu O, Ucer A, Yasar A, Yilmaz B, Kelestimur H. Asprosin-induced alterations in female rat puberty and reproductive hormonal profiles. Arch Physiol Biochem 2025; 131:24-32. [PMID: 39092983 DOI: 10.1080/13813455.2024.2386279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/06/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVE To investigate the comprehensive effects of daily chronic asprosin administration on various pubertal and reproductive parameters in female rats. This study aims to elucidate the role of asprosin in regulating the onset of puberty and its influence on hormonal profiles and ovarian histology. METHODS Asprosin was administered intraperitoneally (i.p.) at a dose of 500 ng/kg daily for eight weeks. Hormonal assays and histological analyses were performed to evaluate the effects of asprosin on the onset of puberty and reproductive function. RESULTS Daily chronic administration of asprosin accelerated the onset of the first oestrus. Hormonal assays revealed significant elevations in serum levels of Follicle-Stimulating Hormone (FSH) and Oestradiol (E2), while Inhibin B levels decreased. Histological evaluations demonstrated an increased number of primary and secondary follicles in ovarian tissue, without affecting primordial follicle counts or reproductive organ weights. CONCLUSIONS Role of adipokines in regulating puberty and reproductive function has increasingly gained recognition. This study aimed to provide the first comprehensive examination of the effects of daily chronic asprosin administration on pubertal and reproductive parameters in female rats. Utilising hormonal assays and histological analyses, asprosin was administered intraperitoneally (i.p.) at a dose of 500 ng/kg, daily, for eight weeks. Our findings revealed that daily chronic administration of asprosin accelerated the onset of the first oestrus. Hormonal assays showed significant elevations in serum levels of Follicle-Stimulating Hormone (FSH) and Oestradiol (E2), while Inhibin B levels decreased. Histological evaluations demonstrated an increased number of primary and secondary follicles in ovarian tissue, without affecting primordial follicle counts or reproductive organ weights. These results provide new insights into asprosin's role in advancing the age of first oestrus and modulating hormonal profiles, thereby offering potential benefits to the female reproductive system.
Collapse
Affiliation(s)
- Emine Kacar
- Department of Physiology, Firat University, Elazig, Turkey
| | - Zeynep Dila Oz
- Department of Physiology, Firat University, Elazig, Turkey
| | | | | | | | - Tugce Yalcin
- Department of Physiology, Firat University, Elazig, Turkey
| | - Gulendam Ozbeg
- Department of Physiology, Firat University, Elazig, Turkey
| | - Aslisah Ozgen
- Department of Physiology, Firat University, Elazig, Turkey
| | - Fatih Tan
- Vocational School of Health Services, Osmaniye Korkut Ata University, Osmaniye, Turkey
| | | | - Ozge Zorlu
- Department of Biophysics, Firat University, Elazig, Turkey
| | - Aysun Ucer
- Department of Biophysics, Firat University, Elazig, Turkey
| | - Abdullah Yasar
- Vocational School of Health Services, Firat University, Elazig, Turkey
| | - Bayram Yilmaz
- Department of Physiology, Yeditepe University, Istanbul, Turkey
| | - Haluk Kelestimur
- Department of Physiology, Istanbul Okan University, Istanbul, Turkey
| |
Collapse
|
43
|
Holalagoudar S, Kisielewski S, Martini A, Johnson K, Leoni AL, Demminger C, Brosel S. Rodent estrous cycle pattern: Harmonizing the cycle evaluation and interpretation. Regul Toxicol Pharmacol 2025; 156:105768. [PMID: 39716560 DOI: 10.1016/j.yrtph.2024.105768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/27/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
The estrous cycle is a sensitive endpoint from an endocrine disruptor perspective and is included in rodent reproductive toxicity studies. In this paper, the methods for estrous cycle timing and different approaches followed by testing laboratories for evaluating the days of the estrous cycle were reviewed. No major differences are identified for counting 4-day estrous cycle. However, when extended episodes of estrus (E) stages occur, the cycle counting differs between testing laboratories potentially resulting in misinterpretation and inaccurate outcome. Appearance of extended episodes of two to three E in an estrous cycle need explanation. Therefore, the following are proposed, 1) follow OECD guidance document for normal 4/5 day cycles, 2) recommends an update of the OECD document as in the current one it is unclear for a 5-day cycle ending with Proestrus(P) followed by episode(s) of estrus(E), 3) two episodes of E after P in a 5-day cycle and three consecutive episodes of E should be considered abnormal, and 4) stages prior to first E of the first cycle or after last E (last P of a 5-day) of last cycle should not be assumed a complete cycle. Additionally, call for collaboration is made to harmonize the cycle counting approach.
Collapse
Affiliation(s)
| | - Susan Kisielewski
- Corteva Agriscience, Haskell R&D Center, 1090 Elkton Road, Newark, DE, 19711, USA.
| | - Austin Martini
- Corteva Agriscience, Haskell R&D Center, 1090 Elkton Road, Newark, DE, 19711, USA.
| | - Kamin Johnson
- Corteva Agriscience LLC, Predictive Safety Center, Indianapolis, IN 46268, USA.
| | - Anne-Laure Leoni
- BSL BIOSERVICE Scientific Laboratories Munich GmbH, Behringstraße 6/8, 82152, Planegg, Germany.
| | - Corinna Demminger
- BSL BIOSERVICE Scientific Laboratories Munich GmbH, Behringstraße 6/8, 82152, Planegg, Germany.
| | - Sonja Brosel
- BSL BIOSERVICE Scientific Laboratories Munich GmbH, Behringstraße 6/8, 82152, Planegg, Germany.
| |
Collapse
|
44
|
Khan D, Sridhar A, Moffett CR. GLP-1R/NPY2R regulate gene expression, ovarian and adrenal morphology in HFD mice. J Endocrinol 2025; 264:e240189. [PMID: 39692365 PMCID: PMC11798413 DOI: 10.1530/joe-24-0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/25/2024] [Accepted: 12/18/2024] [Indexed: 12/19/2024]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) and neuropeptide Y receptors (NPYRs) are expressed in reproductive tissues contributing to the regulation of gonadal function. This exploratory study examines the potential impact of their modulation by assessing the effects of exendin-4 (Ex-4) and peptide YY (PYY) (3-36) on endocrine ovaries and adrenals in high-fat diet (HFD) mice. Ex-4 and PYY(3-36) reduced blood glucose and energy intake, with no effects on body weight. While HFD did not impact the estrous cycle, Ex-4 increased metestrus frequency and decreased diestrus frequency resulting in 0% mice experiencing repeated diestrus or becoming acyclic. Luteinizing hormone levels were significantly higher in the Ex-4 and PYY(3-36) groups compared to the normal diet and HFD controls. In the adrenals, reduced capsule and zona glomerulosa thickness caused by HFD was reversed after peptide treatments. Within the ovaries, HFD increased the number of atretic follicles, an effect that disappeared after Ex-4 and PYY(3-36) treatments. Ex-4 also increased the number of corpora lutea owing to the prolonged metestrus phase. Gene expression analysis within the adrenals revealed the upregulation of Insr and the downregulation of Prgtr in HFD mice, while Ex-4 downregulated the expression of Gipr. The ovarian gene expression of Gipr, Npy1r and Prgtr was downregulated by Ex-4 treatment, while PYY(3-36) significantly downregulated the Prgtr expression compared to HFD mice. These data indicate that manipulating GLP-1R and NPY2R leads to changes in the reproductive physiology of mice. In addition, the observed alterations in the morphology and gene expression in the adrenals and ovaries imply a direct impact of these peptides on female reproductive function.
Collapse
Affiliation(s)
- Dawood Khan
- Diabetes Research Group, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | | | | |
Collapse
|
45
|
Zhu Z, Lei M, Guo R, Xu Y, Zhao Y, Wei C, Yang Q, Sun Y. Nicotinamide riboside supplementation ameliorates ovarian dysfunction in a PCOS mouse model. J Ovarian Res 2025; 18:9. [PMID: 39833950 PMCID: PMC11749135 DOI: 10.1186/s13048-025-01596-4] [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: 09/19/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is the leading cause of anovulatory infertility among women of reproductive age, yet the range of effective treatment options remains limited. Our previous study revealed that reduced levels of nicotinamide adenine dinucleotide (NAD+) in ovarian granulosa cells (GCs) of women with PCOS resulted in the accumulation of reactive oxygen species (ROS) and mitochondrial dysfunction. However, it is still uncertain whether increasing NAD+ levels in the ovaries could improve ovarian function in PCOS. In this study, we demonstrated that supplementation with the NAD+ precursor nicotinamide riboside (NR) prevented the decrease in ovarian NAD+ levels, normalized estrous cycle irregularities, and enhanced ovulation potential in dehydroepiandrosterone (DHEA)-induced PCOS mice. Moreover, NR supplementation alleviated ovarian fibrosis and enhanced mitochondrial function in ovarian stromal cells of PCOS mice. Furthermore, NR supplementation improved oocyte quality in PCOS mice, as evidenced by reduced abnormal mitochondrial clustering, enhanced mitochondrial membrane potential, decreased ROS levels, reduced spindle abnormality rates, and increased early embryonic development potential in fertilized oocytes. These findings suggest that supplementing with NAD+ precursors could be a promising therapeutic strategy for addressing ovarian infertility associated with PCOS.
Collapse
Affiliation(s)
- Zhenye Zhu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Min Lei
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruizhi Guo
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yining Xu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanqing Zhao
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chenlu Wei
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingling Yang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Yingpu Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
46
|
da Silva TQM, Barbosa EM, Santos LC, de Oliveira LS, Cunha MCDSG, de Macedo IO, Martins BGC, Oliveira CL, Rodrigues NP, Araújo-Lopes R, Szawka RE, Silva JF. Hypothyroidism Alters Uterine Kisspeptin System and Activity Modulators in Cyclic Rats. Int J Mol Sci 2025; 26:543. [PMID: 39859259 PMCID: PMC11765193 DOI: 10.3390/ijms26020543] [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: 10/11/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Hypothyroidism causes ovarian dysfunction and infertility in women and animals and impairs the hypothalamic expression of kisspeptin (Kp). However, kisspeptin is also expressed in the genital system, and the lack of the Kp receptor (Kiss1r) in the uterus is linked to reduced implantation rates. This study investigated the impact of hypothyroidism on the uterine expression of Kp and Kiss1r in female rats throughout the estrous cycle and the associated changes in uterine activity modulators. Hypothyroidism was induced through daily administration of propylthiouracil (PTU) over a period of 14 days. Plasma levels of LH, E2, and P4, cyclicity, body and uterine weight, uterine histomorphometry, and the gene and/or protein expression of Kiss1, Kiss1r, estrogen receptor α (ERα), progesterone receptor (PR), and thyroid hormone receptor α (TRα) were assessed. Additionally, proliferative activity (CDC-47) and the gene expression of uterine receptivity mediators (SMO, WNT4, BMP2, HAND2, MUC1, and LIF) were evaluated. Hypothyroidism prolonged the diestrus and increased progesterone levels during this phase, while decreasing luteinizing hormone and estradiol on proestrus. In the uterus, hypothyroidism reduced Kp immunostaining on diestrus and KISS1R mRNA levels on proestrus. These changes were accompanied by reduced endometrial glands, reduced uterine proliferative activity, and reduced ERα gene and protein expression. Additionally, hypothyroidism led to reduced uterine gene expression of LIF, BMP2, WNT4, and HAND2. On the other hand, thyroid hypofunction increased uterine PR and TRα immunostaining, while it reduced PGR gene expression on diestrus. These findings demonstrate that hypothyroidism reduces the expression of Kiss1/Kiss1r system in the uterus, which is associated with disrupted uterine estrogen and progesterone signaling and reduced expression of uterine receptivity mediators across the rat estrous cycle.
Collapse
Affiliation(s)
- Thayná Queiroz Menezes da Silva
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Erikles Macêdo Barbosa
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Luciano Cardoso Santos
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Luciana Santos de Oliveira
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Maria Clara da Silva Galrão Cunha
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Isabella Oliveira de Macedo
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Brenda Geovana Campos Martins
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Cibele Luz Oliveira
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Natalia Panhoca Rodrigues
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Roberta Araújo-Lopes
- Endocrinology and Metabolism Laboratory, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; (R.A.-L.); (R.E.S.)
| | - Raphael Escorsim Szawka
- Endocrinology and Metabolism Laboratory, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; (R.A.-L.); (R.E.S.)
| | - Juneo Freitas Silva
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| |
Collapse
|
47
|
Sziva RE, Kollarics R, Pál É, Bányai B, Korsós-Novák Á, Fontányi Z, Magyar P, Süli A, Nádasy GL, Ács N, Horváth EM, Hadjadj L, Várbíró S. Increased Oxidative and Nitrative Stress and Decreased Sex Steroid Relaxation in a Vitamin D-Deficient Hyperandrogenic Rodent Model-And a Validation of the Polycystic Ovary Syndrome Model. Nutrients 2025; 17:201. [PMID: 39861332 PMCID: PMC11767255 DOI: 10.3390/nu17020201] [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: 11/30/2024] [Revised: 01/03/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Both hyperandrogenism (HA) and vitamin D deficiency (VDD) can separately lead to impaired vascular reactivity and ovulatory dysfunction in fertile females. The aim was to examine the early interactions of these states in a rat model of PCOS. METHODS Four-week-old adolescent female rats were divided into four groups: vitamin D (VD)-supplemented (n = 12); VD-supplemented and testosterone-treated (n = 12); VDD- (n = 11) and VDD-and-testosterone-treated (n = 11). Animals underwent transdermal testosterone treatment for 8 weeks. Target VD levels were achieved with oral VD supplementation and a VD-free diet. Estrous cycles were followed by vaginal smear, and quantitative histomorphometric measurements of the ovaries were also taken. In the 8th week, testosterone- and estrogen-induced relaxation of coronary arterioles was examined with pressure angiography. Estrogen receptor (ER) density and oxidative and nitrative stress parameters (Poly-(ADP-Ribose)-Polymerase and 3-nitrotyrosine) in the vessel wall were investigated with immunohistochemistry. RESULTS VDD caused impaired estrous cycles, and testosterone caused anovulatory cycles (the cycles were stopped at the diestrous phase). VDD combined with testosterone treatment resulted in reduced testosterone and estrogen vasorelaxation, lower ER density, and higher oxidative and nitrative stress in the vessel wall. CONCLUSIONS PCOS with vitamin D deficiency may be associated with increased oxidative-nitrative stress in coronary arterioles. This oxidative and nitrative stress, potentially caused by hyperandrogenism and/or vitamin D deficiency, could impair estrogen-induced relaxation of the coronary arterioles, possibly by decreasing NO bioavailability and disrupting the estrogen-induced relaxation pathway.
Collapse
Affiliation(s)
- Réka Eszter Sziva
- Department of Obstetrics and Gynaecology, Semmelweis University, Üllői Street 78/a, 1082 Budapest, Hungary; (R.E.S.); (Z.F.); (A.S.); (N.Á.); (S.V.)
- Department of Physiology, Semmelweis University, Tűzoltó Street 37–47, 1094 Budapest, Hungary; (B.B.); (G.L.N.); (E.M.H.)
- Workgroup of Research Management, Doctoral School, Semmelweis University, 1085 Budapest, Hungary
| | - Réka Kollarics
- Department of Obstetrics and Gynaecology, Semmelweis University, Üllői Street 78/a, 1082 Budapest, Hungary; (R.E.S.); (Z.F.); (A.S.); (N.Á.); (S.V.)
- Department of Physiology, Semmelweis University, Tűzoltó Street 37–47, 1094 Budapest, Hungary; (B.B.); (G.L.N.); (E.M.H.)
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Street 37–47, 1094 Budapest, Hungary; (É.P.); (L.H.)
- Department of Obstetrics and Gynaecology, Szeged University, Semmelweis Street 1, 6725 Szeged, Hungary
| | - Éva Pál
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Street 37–47, 1094 Budapest, Hungary; (É.P.); (L.H.)
- HUN-REN-SU Cerebrovascular and Neurocognitive Diseases Research Group, 1094 Budapest, Hungary
| | - Bálint Bányai
- Department of Physiology, Semmelweis University, Tűzoltó Street 37–47, 1094 Budapest, Hungary; (B.B.); (G.L.N.); (E.M.H.)
| | - Ágnes Korsós-Novák
- Department of Pathology, Toldy Ferenc Hospital and Outpatient Clinic of Cegléd, Törteli Street 1–3, 2700 Cegléd, Hungary;
| | - Zoltán Fontányi
- Department of Obstetrics and Gynaecology, Semmelweis University, Üllői Street 78/a, 1082 Budapest, Hungary; (R.E.S.); (Z.F.); (A.S.); (N.Á.); (S.V.)
| | - Péter Magyar
- Medical Imaging Centre, Faculty of Medicine, Semmelweis University, Üllői Street 78/a, 1083 Budapest, Hungary;
| | - Anita Süli
- Department of Obstetrics and Gynaecology, Semmelweis University, Üllői Street 78/a, 1082 Budapest, Hungary; (R.E.S.); (Z.F.); (A.S.); (N.Á.); (S.V.)
- Department of Physiology, Semmelweis University, Tűzoltó Street 37–47, 1094 Budapest, Hungary; (B.B.); (G.L.N.); (E.M.H.)
| | - György L. Nádasy
- Department of Physiology, Semmelweis University, Tűzoltó Street 37–47, 1094 Budapest, Hungary; (B.B.); (G.L.N.); (E.M.H.)
| | - Nándor Ács
- Department of Obstetrics and Gynaecology, Semmelweis University, Üllői Street 78/a, 1082 Budapest, Hungary; (R.E.S.); (Z.F.); (A.S.); (N.Á.); (S.V.)
| | - Eszter Mária Horváth
- Department of Physiology, Semmelweis University, Tűzoltó Street 37–47, 1094 Budapest, Hungary; (B.B.); (G.L.N.); (E.M.H.)
| | - Leila Hadjadj
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Street 37–47, 1094 Budapest, Hungary; (É.P.); (L.H.)
| | - Szabolcs Várbíró
- Department of Obstetrics and Gynaecology, Semmelweis University, Üllői Street 78/a, 1082 Budapest, Hungary; (R.E.S.); (Z.F.); (A.S.); (N.Á.); (S.V.)
- Workgroup of Research Management, Doctoral School, Semmelweis University, 1085 Budapest, Hungary
- Department of Obstetrics and Gynaecology, Szeged University, Semmelweis Street 1, 6725 Szeged, Hungary
| |
Collapse
|
48
|
Zhou Z, Han SY, Pardo-Navarro M, Wall EG, Desai R, Vas S, Handelsman DJ, Herbison AE. GnRH pulse generator activity in mouse models of polycystic ovary syndrome. eLife 2025; 13:RP97179. [PMID: 39761106 DOI: 10.7554/elife.97179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
One in ten women in their reproductive age suffer from polycystic ovary syndrome (PCOS) that, alongside subfertility and hyperandrogenism, typically presents with increased luteinizing hormone (LH) pulsatility. As such, it is suspected that the arcuate kisspeptin (ARNKISS) neurons that represent the GnRH pulse generator are dysfunctional in PCOS. We used here in vivo GCaMP fiber photometry and other approaches to examine the behavior of the GnRH pulse generator in two mouse models of PCOS. We began with the peripubertal androgen (PPA) mouse model of PCOS but found that it had a reduction in the frequency of ARNKISS neuron synchronization events (SEs) that drive LH pulses. Examining the prenatal androgen (PNA) model of PCOS, we observed highly variable patterns of pulse generator activity with no significant differences detected in ARNKISS neuron SEs, pulsatile LH secretion, or serum testosterone, estradiol, and progesterone concentrations. However, a machine learning approach identified that the ARNKISS neurons of acyclic PNA mice continued to exhibit cyclical patterns of activity similar to that of normal mice. The frequency of ARNKISS neuron SEs was significantly increased in algorithm-identified 'diestrous stage' PNA mice compared to controls. In addition, ARNKISS neurons exhibited reduced feedback suppression to progesterone in PNA mice and their gonadotrophs were also less sensitive to GnRH. These observations demonstrate the importance of understanding GnRH pulse generator activity in mouse models of PCOS. The existence of cyclical GnRH pulse generator activity in the acyclic PNA mouse indicates the presence of a complex phenotype with deficits at multiple levels of the hypothalamo-pituitary-gonadal axis.
Collapse
Affiliation(s)
- Ziyue Zhou
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Su Young Han
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Maria Pardo-Navarro
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Ellen G Wall
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Reena Desai
- ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Szilvia Vas
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | | | - Allan E Herbison
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
49
|
Maheshwar KV, Stuart AE, Kay LM. Sex differences in olfactory behavior and neurophysiology in Long Evans rats. J Neurophysiol 2025; 133:257-267. [PMID: 39698988 PMCID: PMC11918302 DOI: 10.1152/jn.00222.2024] [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: 05/28/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
In many species, olfactory abilities in females are more acute than those in males. Studies in humans show that women have lower olfactory thresholds and are better able to discriminate and identify odors than men. In mice, odorants elicit faster activation from a larger number of olfactory bulb glomeruli in females than in males. Our study explores sex differences in olfaction in Long Evans rats from a behavioral and electrophysiological perspective. Local field potentials (LFPs) in the olfactory bulb (OB) represent the coordinated activity of bulbar neurons. Olfactory gamma (65-120 Hz) and beta (15-30 Hz) oscillations have been functionally linked to odor perception. Spontaneous and odor-evoked OB LFPs were recorded from awake rats at the same time for 12 days. Odors used included urine of both sexes and monomolecular odorants characterized previously for correlation of volatility with behavior and OB oscillations. Sampling duration in a habituation context, baseline gamma and beta power, and odor-elicited beta and gamma power were analyzed. We find that females sample odorants for a shorter duration than males (just over 1-s difference). Although baseline gamma and beta power do not show significant differences between the two sexes, odor-elicited gamma and beta power in females is significantly lower than in males. Neither sampling duration nor beta and gamma power in females varied systematically with day of estrus. We further verify that variance of these behavioral and physiological measures is not different across sexes, adding to growing evidence that researchers need not be concerned about often-claimed additional variance in female subjects.NEW & NOTEWORTHY Olfaction plays a large role in evolutionary processes. However, we know little about sex differences in olfactory bulb neurophysiology, and many scientists believe that females are more variable because of estrus. We show that female rats sniff odors for shorter durations than males and have lower power in neural oscillations related to cognition. Estrus was not related to variance in any measures. Finally, males and females show equal variance on these behavioral and physiological processes.
Collapse
Affiliation(s)
- Kruthika V Maheshwar
- Master of Arts Program in the Social Sciences, The University of Chicago, Chicago, Illinois, United States
- Institute for Mind and Biology, The University of Chicago, Chicago, Illinois, United States
| | - Abigail E Stuart
- Institute for Mind and Biology, The University of Chicago, Chicago, Illinois, United States
| | - Leslie M Kay
- Institute for Mind and Biology, The University of Chicago, Chicago, Illinois, United States
- Department of Psychology, The University of Chicago, Chicago, Illinois, United States
| |
Collapse
|
50
|
Bornes L, van Winden LJ, Geurts VCM, de Bruijn B, Azarang L, Lanfermeijer M, Caruso M, Proost N, Boeije M, Lohuis JO, Belthier G, Noguera Delgado E, de Gruil N, Kroep JR, van de Ven M, Menezes R, Wesseling J, Kok M, Linn S, Broeks A, van Rossum HH, Scheele CLGJ, van Rheenen J. The oestrous cycle stage affects mammary tumour sensitivity to chemotherapy. Nature 2025; 637:195-204. [PMID: 39633046 PMCID: PMC11666466 DOI: 10.1038/s41586-024-08276-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/23/2024] [Indexed: 12/07/2024]
Abstract
The response of breast cancer to neoadjuvant chemotherapy (NAC) varies substantially, even when tumours belong to the same molecular or histological subtype1. Here we identify the oestrous cycle as an important contributor to this heterogeneity. In three mouse models of breast cancer, we show reduced responses to NAC when treatment is initiated during the dioestrus stage, when compared with initiation during the oestrus stage. Similar findings were observed in retrospective premenopausal cohorts of human patients. Mechanistically, the dioestrus stage exhibits systemic and localized changes, including (1) an increased number of cells undergoing epithelial-to-mesenchymal transition linked to chemoresistance2-4 and (2) decreased tumour vessel diameter, suggesting potential constraints to drug sensitivity and delivery. In addition, an elevated presence of macrophages, previously associated with chemoresistance induction5, characterizes the dioestrus phase. Whereas NAC disrupts the oestrous cycle, this elevated macrophage prevalence persists and depletion of macrophages mitigates the reduced therapy response observed when initiating treatment during dioestrus. Our data collectively demonstrate the oestrous cycle as a crucial infradian rhythm determining chemosensitivity, warranting future clinical studies to exploit optimal treatment initiation timing for enhanced chemotherapy outcomes.
Collapse
Affiliation(s)
- Laura Bornes
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lennart J van Winden
- Laboratory of Clinical Chemistry and Hematology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Veerle C M Geurts
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Beaunelle de Bruijn
- VIB Center for Cancer Biology, KU Leuven Department of Oncology, Leuven, Belgium
| | - Leyla Azarang
- Biostatistics Centre & Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Mirthe Lanfermeijer
- Laboratory of Clinical Chemistry and Hematology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marika Caruso
- VIB Center for Cancer Biology, KU Leuven Department of Oncology, Leuven, Belgium
| | - Natalie Proost
- Mouse Clinic for Cancer and Aging (MCCA) Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Manon Boeije
- Mouse Clinic for Cancer and Aging (MCCA) Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jeroen O Lohuis
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Guillaume Belthier
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Eulàlia Noguera Delgado
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Nadia de Gruil
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marieke van de Ven
- Mouse Clinic for Cancer and Aging (MCCA) Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Renee Menezes
- Biostatistics Centre & Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marleen Kok
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sabine Linn
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pathology, Utrecht University Medical Center, Utrecht, the Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology & Biobanking, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Huub H van Rossum
- Laboratory of Clinical Chemistry and Hematology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Jacco van Rheenen
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|