1
|
Wu W, Wu J, Hou Z, Yan Q, Qin K, Zhao Y, Zhang H, Zhu Y, He J, Li J. Association between ethylene oxide exposure and serum sex hormone levels measured in a reference sample of the US general population. Front Endocrinol (Lausanne) 2025; 16:1533516. [PMID: 40290307 PMCID: PMC12023753 DOI: 10.3389/fendo.2025.1533516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Ethylene oxide (EO) is a crucial organic compound commonly utilized in industrial and medical products. Food and Drug Administration (FDA)-approved EO sterilization sterilizes about 50% of sterile medical devices in the U.S. Animal and human studies have suggested that EO exposure may result in severe health problem. However, studies evaluating the relationship between EO exposure and sex hormones in human populations are still lacking. Therefore, further investigation into EO's effects on humans is essential. This cross-sectional study within the U.S. National Health and Nutrition Examination Survey (NHANES),2013-2016 examined the relationship between EO-hemoglobin adducts (HbEO) and sex hormones. HbEO was found to be inversely associated with estradiol (E2) and positively associated with the ratio of total testosterone (TT) to E2 and sex hormone-binding globulin (SHBG) levels in adult males. Such associations HbEO and E2 and SHBG were non-linear in male adults. However, no significant associations were found between HbEO and sex steroids across various age groups of females and all male age groups except for adults. Thus, our study provides evidence that EO may potentially serve as an endocrine disruptor in the environment, affecting the levels of sex hormones in adult males.
Collapse
Affiliation(s)
- Wenhao Wu
- Department of Endocrinology and Metabolism, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jingna Wu
- Department of Endocrinology and Metabolism, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zebin Hou
- Department of Thyroid Surgery, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi, China
| | - Qi Yan
- Department of Endocrinology and Metabolism, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Kaixin Qin
- Department of Endocrinology and Metabolism, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuan Zhao
- Department of Endocrinology and Metabolism, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hua Zhang
- Department of Endocrinology and Metabolism, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yikun Zhu
- Department of Endocrinology and Metabolism, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Junhua He
- Department of Endocrinology and Metabolism, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jin Li
- Department of Endocrinology and Metabolism, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
2
|
Liufu B, Su Q, Hong K, Wei J, Wang Y, Han Z, Yu L. 17α-Methyltestosterone Affected Growth, Gonadal Development, and Intestinal Microbial Analysis in the Giant Freshwater Prawn ( Macrobrachium rosenbergii). Animals (Basel) 2025; 15:870. [PMID: 40150399 PMCID: PMC11939226 DOI: 10.3390/ani15060870] [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: 02/11/2025] [Revised: 03/14/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
17α-methyltestosterone (MT) is known for its ability to suppress ovaries and induce spermatogenesis; yet, its effects in crustaceans are underexplored. This study investigates the impact of varying MT concentrations on the gonadal development and intestinal microbiota of juvenile Macrobrachium rosenbergii. Feeds containing different MT doses were provided, and the sex ratios, histological observations, reproductive gene expression, and intestinal microbial composition were analyzed. The results revealed short-term feeding (60 days) of 1000 mg/kg MT resulted in the highest male ratio, while long-term feeding (150 days) of 500 mg/kg MT achieved the same outcome. Conversely, long-term feeding of 1500 mg/kg MT led to the lowest male ratio and retarded male germ cell development. An intestinal microbiota analysis showed that MT supplementation significantly increased microbial abundance and altered the intestinal microbial community structure. Additionally, MT suppressed the expression of female reproductive-related genes. This study provides insights into the effects of MT on reproductive development and gut microbiota in juvenile prawns, offering a valuable reference for the application of MT in crustacean aquaculture.
Collapse
Affiliation(s)
- Bai Liufu
- School of Fishery, Zhejiang Ocean University, Zhoushan 316000, China; (B.L.); (Z.H.)
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China; (Q.S.); (K.H.); (J.W.); (Y.W.)
| | - Qiyao Su
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China; (Q.S.); (K.H.); (J.W.); (Y.W.)
| | - Kunhao Hong
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China; (Q.S.); (K.H.); (J.W.); (Y.W.)
| | - Jie Wei
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China; (Q.S.); (K.H.); (J.W.); (Y.W.)
| | - Yakun Wang
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China; (Q.S.); (K.H.); (J.W.); (Y.W.)
| | - Zhiqiang Han
- School of Fishery, Zhejiang Ocean University, Zhoushan 316000, China; (B.L.); (Z.H.)
| | - Lingyun Yu
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China; (Q.S.); (K.H.); (J.W.); (Y.W.)
| |
Collapse
|
3
|
Jalalizadeh M, Buosi K, Giacomelli CF, Leme PAF, Ferrari KL, Dionato FAV, Brito WRS, Brunetti NS, Maia AR, Morari J, Pagliarone AC, Farias AS, Velloso LA, Queiroz MAF, Vallinoto ACR, Bajgelman MC, Reis LO. Therapeutic BCG vaccine protects against long COVID: The BATTLE randomized clinical trial. J Intern Med 2025; 297:60-78. [PMID: 39560319 DOI: 10.1111/joim.20033] [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] [Indexed: 11/20/2024]
Abstract
BACKGROUND Bacillus Calmette-Guérin (BCG) injected during the COVID-19 convalescence period was safe and enhanced recovery from anosmia and dysgeusia in the acute phase. OBJECTIVES To report the long-term results of the BATTLE trial, BCG vaccine in adults with mild COVID-19. METHODS Design: Double-blind, placebo-controlled, randomized (1:1) clinical trial. INTERVENTION BCG intradermal vaccine and placebo. PATIENTS A total of 157 BCG and 142 placebo recipients participated in the 6-month follow-up, and 97 BCG and 95 placebo recipients participated in the 12-month follow-up. MEASUREMENTS Long COVID symptoms and mechanistic analyses. RESULTS BCG reduced hearing problems at 6 months (odds ratio [OR] = 0.26) and sleeping, concentration, memory, and vision problems at 12 months (OR = 0.45, 0.36, 0.38, and 0.36, respectively). Sensitivity analyses confirmed that long COVID-19 symptoms were reduced at the 6- and 12-month follow-ups (p = 0.010 and 0.031, respectively). BCG's crossover interaction paradoxically increased hair loss in women and decreased it in men at 6 months (p = 0.032). BCG immunomodulation is likely mediated through inhibition of Fas ligand expression in the blood and increased induction of IL6, IL10, interferon-induced transmembrane protein 3, and angiotensin-converting enzyme 2 in cultured human macrophages. CONCLUSION Long-term follow-up of the BATTLE trial participants revealed that BCG protects against long COVID development if administered within the COVID-19 convalescence period. The response to BCG was subject-specific, including a paradoxical crossover interaction based on sex. LIMITATIONS Not tested for previous mycobacterial exposure; loss to follow-up, particularly at 12 months.
Collapse
Grants
- 88887.506617/2020-00 Coordination for the Improvement of Higher Education Personnel, CAPES, Federal Government, Brazil
- 88887.657670/2021-00 Coordination for the Improvement of Higher Education Personnel, CAPES, Federal Government, Brazil
- General Coordination of the National Immunization Program - CGPNI/DEIDT/SVS/MS
- 465/2020 Ministry of Health, Brazil
- 304747/2018-1 National Council for Scientific and Technological Development-CNPq, Research Productivity
- 310135/2022-2 National Council for Scientific and Technological Development-CNPq, Research Productivity
- 302935/2021-5 National Council for Scientific and Technological Development-CNPq, Research Productivity
Collapse
Affiliation(s)
- Mehrsa Jalalizadeh
- UroScience, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Keini Buosi
- UroScience, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | | | - Patricia A F Leme
- UroScience, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Karen L Ferrari
- UroScience, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | | | - Wandrey R S Brito
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, Pará, Brazil
| | - Natália S Brunetti
- Biology Institute, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Aline R Maia
- Obesity and Comorbidities Research Center, University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Joseane Morari
- Obesity and Comorbidities Research Center, University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Ana C Pagliarone
- UroScience, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Alessandro S Farias
- Biology Institute, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Licio A Velloso
- Obesity and Comorbidities Research Center, University of Campinas, Unicamp, Campinas, São Paulo, Brazil
| | - Maria A F Queiroz
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, Pará, Brazil
| | - Antonio C R Vallinoto
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, Pará, Brazil
| | - Marcio C Bajgelman
- Brazilian Biosciences National Laboratory, LNBio, CNPEM, Campinas, São Paulo, Brazil
| | - Leonardo O Reis
- UroScience, State University of Campinas, Unicamp, Campinas, São Paulo, Brazil
- ImmunOncology, Pontifical Catholic University of Campinas, PUC-Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
4
|
Zhu T, Jin M, Luo J, Yang Y, Li X, Peng H, Shen Y, Zhou Q. Mating behaviour and cholesterol nutritional strategies promoted ovarian development of female swimming crab ( Portunus trituberculatus). Br J Nutr 2024; 132:835-850. [PMID: 39391922 DOI: 10.1017/s0007114524001193] [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: 10/12/2024]
Abstract
Female crabs enter a stage of rapid ovarian development after mating, and cholesterol is a substrate for steroid hormone synthesis. Therefore, in this experiment, an 8-week feeding trial was conducted to investigate the effects of mating treatments (mated crab and unmated crab) and three dietary cholesterol levels (0·09 %, 0·79 % and 1·40 %) on ovarian development, cholesterol metabolism and steroid hormones metabolism of adult female swimming crab (Portunus trituberculatus). The results indicated that crabs fed the diet with 0·79 % cholesterol significantly increased gonadosomatic index (GSI) and vitellogenin (VTG) content than other treatments in the same mating status. Moreover, mated crabs had markedly increased GSI and VTG content in the ovary and hepatopancreas than unmated crabs. The histological observation found that exogenous vitellogenic oocytes appeared in the mated crabs, while previtellogenic oocytes and endogenous vitellogenic oocytes were the primary oocytes in unmated crabs. The transmission electron microscopy analysis showed that when fed diet with 0·79 % cholesterol, the unmated crabs contained more rough endoplasmic reticulum and mated crabs had higher yolk content than other treatments. Furthermore, mating treatment and dietary 0·79 % cholesterol level both promoted cholesterol deposition by up-regulation of the mRNA and protein expression levels of class B scavenger receptors 1 (Srb1), while stimulating the secretion of steroid hormones by up-regulation of the mRNA and protein expression of steroidogenic acute regulatory protein (Star). Overall, the present results indicated that mating behaviour plays a leading role in promoting ovarian development, and dietary 0·79 % cholesterol level can further promote ovarian development after mating.
Collapse
Affiliation(s)
- Tingting Zhu
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Aquaculture Biotechnology Ministry of Education, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo315211, People's Republic of China
| | - Min Jin
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Aquaculture Biotechnology Ministry of Education, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo315211, People's Republic of China
| | - Jiaxiang Luo
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Aquaculture Biotechnology Ministry of Education, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo315211, People's Republic of China
| | - Yuhang Yang
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Aquaculture Biotechnology Ministry of Education, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo315211, People's Republic of China
| | - Xiangkai Li
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Aquaculture Biotechnology Ministry of Education, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo315211, People's Republic of China
| | - Hongyu Peng
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Aquaculture Biotechnology Ministry of Education, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo315211, People's Republic of China
| | - Yuedong Shen
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Aquaculture Biotechnology Ministry of Education, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo315211, People's Republic of China
| | - Qicun Zhou
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Aquaculture Biotechnology Ministry of Education, Ningbo University, Ningbo315211, People's Republic of China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo315211, People's Republic of China
| |
Collapse
|
5
|
Pavlinek A, Adhya D, Tsompanidis A, Warrier V, Vernon AC, Lancaster M, Mill J, Srivastava DP, Baron-Cohen S. Using Organoids to Model Sex Differences in the Human Brain. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100343. [PMID: 39092139 PMCID: PMC11292257 DOI: 10.1016/j.bpsgos.2024.100343] [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: 10/18/2023] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 08/04/2024] Open
Abstract
Sex differences are widespread during neurodevelopment and play a role in neuropsychiatric conditions such as autism, which is more prevalent in males than females. In humans, males have been shown to have larger brain volumes than females with development of the hippocampus and amygdala showing prominent sex differences. Mechanistically, sex steroids and sex chromosomes drive these differences in brain development, which seem to peak during prenatal and pubertal stages. Animal models have played a crucial role in understanding sex differences, but the study of human sex differences requires an experimental model that can recapitulate complex genetic traits. To fill this gap, human induced pluripotent stem cell-derived brain organoids are now being used to study how complex genetic traits influence prenatal brain development. For example, brain organoids from individuals with autism and individuals with X chromosome-linked Rett syndrome and fragile X syndrome have revealed prenatal differences in cell proliferation, a measure of brain volume differences, and excitatory-inhibitory imbalances. Brain organoids have also revealed increased neurogenesis of excitatory neurons due to androgens. However, despite growing interest in using brain organoids, several key challenges remain that affect its validity as a model system. In this review, we discuss how sex steroids and the sex chromosomes each contribute to sex differences in brain development. Then, we examine the role of X chromosome inactivation as a factor that drives sex differences. Finally, we discuss the combined challenges of modeling X chromosome inactivation and limitations of brain organoids that need to be taken into consideration when studying sex differences.
Collapse
Affiliation(s)
- Adam Pavlinek
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Dwaipayan Adhya
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Alex Tsompanidis
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Varun Warrier
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Anthony C. Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | | | - Jonathan Mill
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Simon Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
6
|
Murillo Ramos AM, Wilson JY. Is there potential for estradiol receptor signaling in lophotrochozoans? Gen Comp Endocrinol 2024; 354:114519. [PMID: 38677339 DOI: 10.1016/j.ygcen.2024.114519] [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: 03/07/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024]
Abstract
Estrogen receptors (ERs) are thought to be the ancestor of all steroid receptors and are present in most lophotrochozoans studied to date, including molluscs, annelids, and rotifers. A number of studies have investigated the functional role of estrogen receptors in invertebrate species, although most are in molluscs, where the receptor is constitutively active. In vitro experiments provided evidence for ligand-activated estrogen receptors in annelids, raising important questions about the role of estrogen signalling in lophotrochozoan lineages. Here, we review the concordant and discordant evidence of estradiol receptor signalling in lophotrochozoans, with a focus on annelids and rotifers. We explore the de novo synthesis of estrogens, the evolution and expression of estrogen receptors, and physiological responses to activation of estrogen receptors in the lophotrochozoan phyla Annelida and Rotifera. Key data are missing to determine if de novo biosynthesis of estradiol in non-molluscan lophotrochozoans is likely. For example, an ortholog for the CYP11 gene is present, but confirmation of substrate conversion and measured tissue products is lacking. Orthologs CYP17 and CYP19 are lacking, yet intermediates or products (e.g. estradiol) in tissues have been measured. Estrogen receptors are present in multiple species, and for a limited number, in vitro data show agonist binding of estradiol and/or transcriptional activation. The expression patterns of the lophotrochozoan ERs suggest developmental, reproductive, and digestive roles but are highly species dependent. E2 exposures suggest that lophotrochozoan ERs may play a role in reproduction, but no strong dose-response relationship has been established. Therefore, we expect most lophotrochozoan species, outside of perhaps platyhelminths, to have an ER but their physiological role remains elusive. Mining genomes for orthologs gene families responsible for steroidogenesis, coupled with in vitro and in vivo studies of the steroid pathway are needed to better assess whether lophotrochozoans are capable of estradiol biosynthesis. One major challenge is that much of the data are divided across a diversity of species. We propose that the polychaetes Capitella teleta or Platyneris dumerilii, and rotifer Brachionus manjavacas may be strong species choices for studies of estrogen receptor signalling, because of available genomic data, established laboratory culture techniques, and gene knockout potential.
Collapse
Affiliation(s)
- A M Murillo Ramos
- Department of Biology, McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada.
| | - J Y Wilson
- Department of Biology, McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada.
| |
Collapse
|
7
|
Wu T, Wu Y, Li Y, Du Y, Feng S, Wang D, Zhou L. Genome-wide analysis of two different regions of brain reveals the molecular changes of fertility related genes in rln3a -/- mutants in male Nile tilapia (Oreochromis niloticus). Gen Comp Endocrinol 2024; 354:114543. [PMID: 38692521 DOI: 10.1016/j.ygcen.2024.114543] [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: 02/04/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/03/2024]
Abstract
Relaxin3 (rln3) has been associated with various emotional and cognitive processes, including stress, anxiety, learning, memory, motivational behavior, and circadian rhythm. Notably, previous report revealed that Rln3a played an indispensable role in testicular development and male fertility in Nile tilapia (Oreochromis niloticus). However, the underlying molecular mechanisms remain largely unknown. We found that Rln3a is expressed exclusively in the diencephalon* (Di*) of the brain. Deficiency of Rln3a resulted in a significant increase in serum dopamine level and an upregulation of gene expression of gnrh1 and kisspeptin2. To further elucidate the role of Rln3a in fish fertility, we collected two different regions of Di* and hypothalamus (Hyp) tissues for subsequent RNA-seq analysis of both wild-type (rln3a+/+) and rln3a-/- male tilapia. Upon the transcriptomic data, 1136 and 755 differentially expressed genes (DEGs) were identified in the Di* and Hyp tissues, respectively. In Di*, the up-regulated genes were enriched in circadian rhythm, chemical carcinogenesis, while the down-regulated genes were enriched in type II diabetes mellitus, dopaminergic synapse, and other pathways. In Hyp, the up-regulated genes were enriched in circadian rhythm, pyrimidine metabolism, while the down-regulated genes were enriched in type I diabetes mellitus, autoimmune thyroid disease, and other pathways. Subsequently, the results of both qRT-PCR and FISH assays highlighted a pronounced up-regulation of core circadian rhythm genes, cry1b and per3, whereas genes such as clocka, clockb, and arntl exhibited down-regulation. Furthermore, the genes associated with dopamine biosynthesis were significantly increased in the Hyp. In summary, the mutation of rln3a in male tilapia resulted in notable changes in circadian rhythm and disease-linked signaling pathways in the Di* and Hyp. These changes might account for the fertility defects observed in rln3a-/- male mutants in tilapia.
Collapse
Affiliation(s)
- Tengfei Wu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Science, Southwest University, Chongqing 400715, China
| | - You Wu
- Fisheries Engineering Institute, Chinese Academy of Fishery Sciences, Beijing, China; Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Science, Southwest University, Chongqing 400715, China
| | - Yanlong Li
- Fisheries Engineering Institute, Chinese Academy of Fishery Sciences, Beijing, China; Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Science, Southwest University, Chongqing 400715, China
| | - Yiyun Du
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Science, Southwest University, Chongqing 400715, China
| | - Saining Feng
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Science, Southwest University, Chongqing 400715, China
| | - Deshou Wang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Science, Southwest University, Chongqing 400715, China.
| | - Linyan Zhou
- Fisheries Engineering Institute, Chinese Academy of Fishery Sciences, Beijing, China.
| |
Collapse
|
8
|
Lecce E, Conti A, Nuccio S, Felici F, Bazzucchi I. Characterising sex-related differences in lower- and higher-threshold motor unit behaviour through high-density surface electromyography. Exp Physiol 2024; 109:1317-1329. [PMID: 38888901 PMCID: PMC11291872 DOI: 10.1113/ep091823] [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: 02/14/2024] [Accepted: 05/28/2024] [Indexed: 06/20/2024]
Abstract
Emerging questions in neuromuscular physiology revolve around whether males and females share similar neural control in diverse tasks across a broad range of intensities. In order to explore these features, high-density electromyography was used to record the myoelectrical activity of biceps brachii during trapezoidal isometric contractions at 35% and 70% of maximal voluntary force (MVF) on 11 male and 13 female participants. Identified motor units were then classified as lower-threshold (recruited at ≤30%MVF) and higher-threshold (recruited at >30%MVF). The discharge rate, interspike interval variability, recruitment and derecruitment thresholds, and estimates of neural drive to motor neurons were assessed. Female lower-threshold motor units showed higher neural drive (P < 0.001), accompanied by higher discharge rate at recruitment (P = 0.006), plateau (P = 0.001) and derecruitment (P = 0.001). On the other hand, male higher-threshold motor units showed greater neural drive (P = 0.04), accompanied by higher discharge rate at recruitment (P = 0.005), plateau (P = 0.04) and derecruitment (P = 0.01). Motor unit discharge rate normalised by the recruitment threshold was significantly higher in female lower-threshold motor units (P < 0.001), while no differences were observed in higher-threshold motor units. Recruitment and derecruitment thresholds are higher in males across all intensities (P < 0.01). However, males and females have similar activation and deactivation strategies, as evidenced by similar recruitment-to-derecruitment ratios (P > 0.05). This study encompasses a broad intensity range to analyse motor unit sex-related differences, highlighting higher neural drive and discharge rates in female lower-threshold motor units, elevated recruitment and derecruitment thresholds in males, and convergences in activation and deactivation strategies. HIGHLIGHTS: What is the central question of the study? Do male and female motor units behave similarly in low- and high-intensity contractions? What is the main finding and its importance? Female motor units show higher discharge rates in low-intensity tasks and lower discharge rates in high-intensity tasks, with no differences in recruitment behaviour. A broader inter-spike interval variability was also observed in females. These findings underline that there are sex-specific differences concern the firing strategies based on task intensity.
Collapse
Affiliation(s)
- Edoardo Lecce
- Department of Movement, Human and Health Sciences, Laboratory of Exercise PhysiologyUniversity of Rome ‘Foro Italico’RomeItaly
| | - Alessandra Conti
- Department of Movement, Human and Health Sciences, Laboratory of Exercise PhysiologyUniversity of Rome ‘Foro Italico’RomeItaly
| | - Stefano Nuccio
- Department of Movement, Human and Health Sciences, Laboratory of Exercise PhysiologyUniversity of Rome ‘Foro Italico’RomeItaly
| | - Francesco Felici
- Department of Movement, Human and Health Sciences, Laboratory of Exercise PhysiologyUniversity of Rome ‘Foro Italico’RomeItaly
| | - Ilenia Bazzucchi
- Department of Movement, Human and Health Sciences, Laboratory of Exercise PhysiologyUniversity of Rome ‘Foro Italico’RomeItaly
| |
Collapse
|
9
|
Clark KL, Shukla M, George JW, Gustin S, Rowley MJ, Davis JS. An environmentally relevant mixture of per- and polyfluoroalkyl substances (PFAS) impacts proliferation, steroid hormone synthesis, and gene transcription in primary human granulosa cells. Toxicol Sci 2024; 200:57-69. [PMID: 38603627 PMCID: PMC11199914 DOI: 10.1093/toxsci/kfae049] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024] Open
Abstract
Per- and polyfluoroalkyl substances (PFAS) are a group of synthetic chemicals that are resistant to biodegradation and are environmentally persistent. PFAS are found in many consumer products and are a major source of water and soil contamination. This study investigated the effects of an environmentally relevant PFAS mixture (perfluorooctanoic acid [PFOA], perfluorooctanesulfonic acid [PFOS], perfluorohexanesulfonic acid [PFHxS]) on the transcriptome and function of human granulosa cells (hGCs). Primary hGCs were harvested from follicular aspirates of healthy, reproductive-age women who were undergoing oocyte retrieval for in vitro fertilization. Liquid Chromatography with tandem mass spectrometry (LC/MS-MS) was performed to identify PFAS compounds in pure follicular fluid. Cells were cultured with vehicle control or a PFAS mixture (2 nM PFHxS, 7 nM PFOA, 10 nM PFOS) for 96 h. Analyses of cell proliferation/apoptosis, steroidogenesis, and gene expression were measured via 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays/immunofluorescence, ELISA/western blotting, and RNA sequencing/bioinformatics, respectively. PFOA, PFOS, and PFHxS were detected in 100% of follicle fluid samples. Increased cell proliferation was observed in hGCs treated with the PFAS mixture with no impacts on cellular apoptosis. The PFAS mixture also altered steroid hormone synthesis, increasing both follicle-stimulating hormone-stimulated and basal progesterone secretion and concomitant upregulation of STAR protein. RNA sequencing revealed inherent differences in transcriptomic profiles in hGCs after PFAS exposure. This study demonstrates functional and transcriptomic changes in hGCs after exposure to a PFAS mixture, improving our knowledge about the impacts of PFAS exposures and female reproductive health. These findings suggest that PFAS compounds can disrupt normal granulosa cell function with possible long-term consequences on overall reproductive health.
Collapse
Affiliation(s)
- Kendra L Clark
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska 68105, USA
| | - Mamta Shukla
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Jitu W George
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska 68105, USA
| | - Stephanie Gustin
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
- Heartland Center for Reproductive Medicine, Omaha, Nebraska 68138, USA
| | - M Jordan Rowley
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - John S Davis
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska 68105, USA
| |
Collapse
|
10
|
Piasecki J, Škarabot J, Spillane P, Piasecki M, Ansdell P. Sex Differences in Neuromuscular Aging: The Role of Sex Hormones. Exerc Sport Sci Rev 2024; 52:54-62. [PMID: 38329342 DOI: 10.1249/jes.0000000000000335] [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] [Indexed: 02/09/2024]
Abstract
Males and females experience different trajectories of neuromuscular function across the lifespan, with females demonstrating accelerated deconditioning in later life. We hypothesize that the menopause is a critical period in the female lifespan, during which the dramatic reduction in sex hormone concentrations negatively impacts synaptic input to the motoneuron pool, as well as motor unit discharge properties.
Collapse
Affiliation(s)
- Jessica Piasecki
- Sport, Health and Performance Enhancement Research Centre, Nottingham Trent University, Nottingham, UK
| | - Jakob Škarabot
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Padraig Spillane
- Department of Sport, Exercise and Rehabilitation, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Mathew Piasecki
- Centre of Metabolism, Ageing and Physiology (COMAP), MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Paul Ansdell
- Department of Sport, Exercise and Rehabilitation, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| |
Collapse
|
11
|
Dai Z, Gong Z, Wang C, Long W, Liu D, Zhang H, Lei A. The role of hormones in ILC2-driven allergic airway inflammation. Scand J Immunol 2024; 99:e13357. [PMID: 39008023 DOI: 10.1111/sji.13357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/23/2023] [Accepted: 01/05/2024] [Indexed: 07/16/2024]
Abstract
Group 2 innate lymphoid cells (ILC2s) are a type of innate immune cells that produce a large amount of IL-5 and IL-13 and two cytokines that are crucial for various processes such as allergic airway inflammation, tissue repair and tissue homeostasis. It is known that damaged epithelial-derived alarmins, such as IL-33, IL-25 and thymic stromal lymphopoietin (TSLP), are the predominant ILC2 activators that mediate the production of type 2 cytokines. In recent years, abundant studies have found that many factors can regulate ILC2 development and function. Hormones synthesized by the body's endocrine glands or cells play an important role in immune response. Notably, ILC2s express hormone receptors and their proliferation and function can be modulated by multiple hormones during allergic airway inflammation. Here, we summarize the effects of multiple hormones on ILC2-driven allergic airway inflammation and discuss the underlying mechanisms and potential therapeutic significance.
Collapse
Affiliation(s)
- Zhongling Dai
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Zhande Gong
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Cui Wang
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - WeiXiang Long
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Duo Liu
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Haijun Zhang
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Aihua Lei
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
12
|
Unger CA, Hope MC, Aladhami AK, Cotham WE, Socia CE, Rice BC, Clegg DJ, Velázquez KT, LaVoie HA, Hollis F, Enos RT. A Novel Tissue-Specific Insight into Sex Steroid Fluctuations Throughout the Murine Estrous Cycle. Endocrinology 2023; 165:bqad175. [PMID: 37967240 PMCID: PMC11032246 DOI: 10.1210/endocr/bqad175] [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: 10/13/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/17/2023]
Abstract
Serum sex steroid levels fluctuate throughout the reproductive cycle. However, the degree to which sex steroid tissue content mimics circulating content is unknown. Understanding the flux and physiological quantity of tissue steroid content is imperative for targeted hormonal therapy development. Utilizing a gold-standard ultrasensitive liquid chromatography-mass spectrometry (LC/MS) method we determined sex steroid (17β-estradiol [E2], testosterone, androstenedione, and progesterone) fluctuations in serum and in 15 tissues throughout the murine estrous cycle (proestrus, estrus, and diestrus I) and in ovariectomized (OVX) mice. We observed dynamic fluctuations in serum and tissue steroid content throughout the estrous cycle with proestrus generally presenting the highest content of E2, testosterone, and androstenedione, and lowest content of progesterone. In general, the trend in circulating steroid content between the stages of the estrous cycle was mimicked in tissue. However, the absolute amounts of steroid levels when normalized to tissue weight were found to be significantly different between the tissues with the serum steroid quantity often being significantly lower than the tissue quantity. Additionally, we found that OVX mice generally displayed a depletion of all steroids in the various tissues assessed, except in the adrenal glands which were determined to be the main site of peripheral E2 production after ovary removal. This investigation provides a comprehensive analysis of steroid content throughout the estrous cycle in a multitude of tissues and serum. We believe this information will help serve as the basis for the development of physiologically relevant, tissue-specific hormonal therapies.
Collapse
Affiliation(s)
- Christian A Unger
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29209, USA
| | - Marion C Hope
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29209, USA
| | - Ahmed K Aladhami
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29209, USA
| | - William E Cotham
- Department of Chemistry and Biochemistry, College of Arts and Science, University of South Carolina, Columbia, SC 29208, USA
| | - Cassidy E Socia
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29209, USA
| | - Barton C Rice
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29209, USA
| | - Deborah J Clegg
- Department of Internal Medicine, Texas Tech Health Sciences Center, El Paso, TX 7995, USA
| | - Kandy T Velázquez
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29209, USA
| | - Holly A LaVoie
- Department of Cell Biology and Anatomy, University of South Carolina, School of Medicine, Columbia, SC 29209, USA
| | - Fiona Hollis
- Department of Pharmacology, Physiology, and Neuroscience, School of Medicine, Columbia, SC 29209, USA
| | - Reilly T Enos
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29209, USA
| |
Collapse
|
13
|
Fancher AT, Hua Y, Close DA, Xu W, McDermott LA, Strock CJ, Santiago U, Camacho CJ, Johnston PA. Characterization of allosteric modulators that disrupt androgen receptor co-activator protein-protein interactions to alter transactivation-Drug leads for metastatic castration resistant prostate cancer. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023; 28:325-343. [PMID: 37549772 DOI: 10.1016/j.slasd.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/06/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Three series of compounds were prioritized from a high content screening campaign that identified molecules that blocked dihydrotestosterone (DHT) induced formation of Androgen Receptor (AR) protein-protein interactions (PPIs) with the Transcriptional Intermediary Factor 2 (TIF2) coactivator and also disrupted preformed AR-TIF2 PPI complexes; the hydrobenzo-oxazepins (S1), thiadiazol-5-piperidine-carboxamides (S2), and phenyl-methyl-indoles (S3). Compounds from these series inhibited AR PPIs with TIF2 and SRC-1, another p160 coactivator, in mammalian 2-hybrid assays and blocked transcriptional activation in reporter assays driven by full length AR or AR-V7 splice variants. Compounds inhibited the growth of five prostate cancer cell lines, with many exhibiting differential cytotoxicity towards AR positive cell lines. Representative compounds from the 3 series substantially reduced both endogenous and DHT-enhanced expression and secretion of the prostate specific antigen (PSA) cancer biomarker in the C4-2 castration resistant prostate cancer (CRPC) cell line. The comparatively weak activities of series compounds in the H3-DHT and/or TIF2 box 3 LXXLL-peptide binding assays to the recombinant ligand binding domain of AR suggest that direct antagonism at the orthosteric ligand binding site or AF-2 surface respectively are unlikely mechanisms of action. Cellular enhanced thermal stability assays (CETSA) indicated that compounds engaged AR and reduced the maximum efficacy and right shifted the EC50 of DHT-enhanced AR thermal stabilization consistent with the effects of negative allosteric modulators. Molecular docking of potent representative hits from each series to AR structures suggest that S1-1 and S2-6 engage a novel binding pocket (BP-1) adjacent to the orthosteric ligand binding site, while S3-11 occupies the AR binding function 3 (BF-3) allosteric pocket. Hit binding poses indicate spaces and residues adjacent to the BP-1 and BF-3 pockets that will be exploited in future medicinal chemistry optimization studies. Small molecule allosteric modulators that prevent/disrupt AR PPIs with coactivators like TIF2 to alter transcriptional activation in the presence of orthosteric agonists might evade the resistance mechanisms to existing prostate cancer drugs and provide novel starting points for medicinal chemistry lead optimization and future development into therapies for metastatic CRPC.
Collapse
Affiliation(s)
- Ashley T Fancher
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; Nucleus Global, 2 Ravinia Drive, Suite 605, Atlanta, GA 30346, USA
| | - Yun Hua
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - David A Close
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Wei Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lee A McDermott
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; PsychoGenics Inc, 215 College Road, Paramus, NJ 07652, USA
| | | | - Ulises Santiago
- Department of Computational and Systems Biology, School of Medicine, at the University of Pittsburgh, USA
| | - Carlos J Camacho
- Department of Computational and Systems Biology, School of Medicine, at the University of Pittsburgh, USA
| | - Paul A Johnston
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA 15232, USA.
| |
Collapse
|
14
|
Wang W, Wu B, Liu Z, Sun X, Zhou L, Xu W, Yu T, Zheng Y, Zhang S. Comprehensive analysis on the regulation of differentially expressed of mRNA and ncRNA in different ovarian stages of ark shell Scapharca broughtonii. BMC Genomics 2023; 24:563. [PMID: 37736709 PMCID: PMC10515027 DOI: 10.1186/s12864-023-09648-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/04/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Ovarian development is an important prerequisite and basis for animal reproduction. In many vertebrates, it is regulated by multiple genes and influenced by sex steroid hormones and environmental factors. However, relative information is limited in shellfish. To explore the biological functions and molecular mechanisms of mRNA and non-coding RNA that regulate ovarian development in Scapharca broughtonii, we performed whole transcriptome sequencing analysis on ovaries at three developmental stages. Furthermore, the biological processes involved in the differential expression of mRNA and ncRNA were analyzed. RESULTS A total of 11,342 mRNAs, 6897 lncRNAs, 135 circRNAs, and 275 miRNAs were differentially expressed. By mapping the differentially expressed RNAs from the three developmental stages of Venn diagram, multiple groups of shared mRNAs and lncRNAs were found to be associated with ovarian development, with some mRNA and ncRNA functions associated with steroid hormone. In addition, we constructed and visualized the lncRNA/circRNA-miRNA-mRNA network based on ceRNA targeting relationships. CONCLUSIONS These findings may facilitate our further understanding the mRNA and ncRNAs roles in the regulation of shellfish reproduction.
Collapse
Affiliation(s)
- Wenjing Wang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 266071, Qingdao, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, 201306, Shanghai, China
| | - Biao Wu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 266071, Qingdao, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, 266237, Qingdao, China.
| | - Zhihong Liu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 266071, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, 266237, Qingdao, China
| | - Xiujun Sun
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 266071, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, 266237, Qingdao, China
| | - Liqing Zhou
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 266071, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, 266237, Qingdao, China
| | - Wandong Xu
- Administrative Examination and Approval Service Bureau of Kenli District, Dongying, China, 257500
| | - Tao Yu
- Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Sciences, Yantai, China, 265800
| | - Yanxin Zheng
- Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Sciences, Yantai, China, 265800
| | - Shihao Zhang
- Shandong Anhai lnvestment , Jinan, China, Co., Ltd, 250013
| |
Collapse
|
15
|
Stanelle-Bertram S, Beck S, Mounogou NK, Schaumburg B, Stoll F, Al Jawazneh A, Schmal Z, Bai T, Zickler M, Beythien G, Becker K, de la Roi M, Heinrich F, Schulz C, Sauter M, Krasemann S, Lange P, Heinemann A, van Riel D, Leijten L, Bauer L, van den Bosch TPP, Lopuhaä B, Busche T, Wibberg D, Schaudien D, Goldmann T, Lüttjohann A, Ruschinski J, Jania H, Müller Z, Pinho Dos Reis V, Krupp-Buzimkic V, Wolff M, Fallerini C, Baldassarri M, Furini S, Norwood K, Käufer C, Schützenmeister N, von Köckritz-Blickwede M, Schroeder M, Jarczak D, Nierhaus A, Welte T, Kluge S, McHardy AC, Sommer F, Kalinowski J, Krauss-Etschmann S, Richter F, von der Thüsen J, Baumgärtner W, Klingel K, Ondruschka B, Renieri A, Gabriel G. CYP19A1 mediates severe SARS-CoV-2 disease outcome in males. Cell Rep Med 2023; 4:101152. [PMID: 37572667 PMCID: PMC10518605 DOI: 10.1016/j.xcrm.2023.101152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 05/10/2023] [Accepted: 07/18/2023] [Indexed: 08/14/2023]
Abstract
Male sex represents one of the major risk factors for severe COVID-19 outcome. However, underlying mechanisms that mediate sex-dependent disease outcome are as yet unknown. Here, we identify the CYP19A1 gene encoding for the testosterone-to-estradiol metabolizing enzyme CYP19A1 (also known as aromatase) as a host factor that contributes to worsened disease outcome in SARS-CoV-2-infected males. We analyzed exome sequencing data obtained from a human COVID-19 cohort (n = 2,866) using a machine-learning approach and identify a CYP19A1-activity-increasing mutation to be associated with the development of severe disease in men but not women. We further analyzed human autopsy-derived lungs (n = 86) and detect increased pulmonary CYP19A1 expression at the time point of death in men compared with women. In the golden hamster model, we show that SARS-CoV-2 infection causes increased CYP19A1 expression in the lung that is associated with dysregulated plasma sex hormone levels and reduced long-term pulmonary function in males but not females. Treatment of SARS-CoV-2-infected hamsters with a clinically approved CYP19A1 inhibitor (letrozole) improves impaired lung function and supports recovery of imbalanced sex hormones specifically in males. Our study identifies CYP19A1 as a contributor to sex-specific SARS-CoV-2 disease outcome in males. Furthermore, inhibition of CYP19A1 by the clinically approved drug letrozole may furnish a new therapeutic strategy for individualized patient management and treatment.
Collapse
Affiliation(s)
| | - Sebastian Beck
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Nancy Kouassi Mounogou
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Berfin Schaumburg
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Fabian Stoll
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Amirah Al Jawazneh
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Zoé Schmal
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Tian Bai
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Martin Zickler
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Georg Beythien
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Kathrin Becker
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Madeleine de la Roi
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Fabian Heinrich
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Schulz
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Martina Sauter
- Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Susanne Krasemann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Core Facility Experimental Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Philine Lange
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Axel Heinemann
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Debby van Riel
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Lonneke Leijten
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Lisa Bauer
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Boaz Lopuhaä
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Tobias Busche
- Medical School East Westphalia-Lippe & Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Daniel Wibberg
- Microbial Genomics and Biotechnology, Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
| | - Torsten Goldmann
- Pathology of the University Medical Center Schleswig-Holstein, Campus Lübeck and the Research Center Borstel, Research Center Borstel, Leibniz Center for Medicine and Biosciences, German Center for Lung Research (DZL), Borstel, Germany
| | - Anna Lüttjohann
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Jenny Ruschinski
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Hanna Jania
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Zacharias Müller
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | | | - Vanessa Krupp-Buzimkic
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Martin Wolff
- Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Chiara Fallerini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Medical Genetics, University of Siena, Siena, Italy
| | - Margherita Baldassarri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Medical Genetics, University of Siena, Siena, Italy
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Katrina Norwood
- Department for Computational Biology of Infection Research, Helmholtz Center for Infection Research, Braunschweig, Germany; Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Christopher Käufer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Maren von Köckritz-Blickwede
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany; Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Maria Schroeder
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dominik Jarczak
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Axel Nierhaus
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease, Member of the German Center for Lung Research, Hannover, Germany
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alice C McHardy
- German Center for Infection Research (DZIF), Braunschweig, Germany; Department for Computational Biology of Infection Research, Helmholtz Center for Infection Research, Braunschweig, Germany; Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany; Cluster of Excellence RESIST (EXC 2355), Hannover Medical School, Hannover, Germany
| | - Frank Sommer
- Division Men's Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jörn Kalinowski
- Microbial Genomics and Biotechnology, Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Susanne Krauss-Etschmann
- Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, Member of the German Center for Lung Research (DZL), Borstel, Germany; Institute for Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Jan von der Thüsen
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Karin Klingel
- Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alessandra Renieri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Medical Genetics, University of Siena, Siena, Italy; Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Gülsah Gabriel
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany; Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany; German Center for Infection Research (DZIF), Braunschweig, Germany.
| |
Collapse
|
16
|
Tabor JB, Penner LC, Cooper JG, Ghodsi M, Galarneau JM, Fraser DD, Emery CA, Wellington CL, Debert CT. Characterizing Factors Influencing Baseline Plasma Biomarkers for Sport-Related Concussion in Adolescents. J Neurotrauma 2023; 40:1638-1650. [PMID: 36852497 DOI: 10.1089/neu.2022.0501] [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: 03/01/2023] Open
Abstract
Abstract Developing objective measures to diagnose sport-related concussion (SRC) is a top priority, particularly in the pediatric context, given the vulnerability of the developing brain. While advances in SRC blood biomarkers are being made in adult populations, less data are available for adolescents. Clinical validation of blood biomarkers post-SRC will first require investigation in a healthy uninjured state. Further, rapid pubertal changes during adolescence may implicate possible interactions with circulating sex hormones and the menstrual cycle for females. This cross-sectional study aimed to characterize pre-injury plasma levels of glial fibrillary acidic protein (GFAP), neurofilament light (NF-L), ubiquitin C-terminal hydrolase-L1 (UCH-L1), total tau (T-tau), and phosphorylated tau-181 (P-tau-181), considering previous concussion, age, and sex in healthy adolescent sport participants. Possible associations with menstrual cycle phase and circulating sex hormone levels (i.e., progesterone, estradiol, testosterone) were also explored. Pre-injury blood samples were obtained from 149 healthy adolescents (48% female, ages 11-18) participating in a larger Surveillance in High Schools and Community Sports to Reduce Concussions and their Consequences (SHRed Concussions) multi-site longitudinal cohort study. Main outcomes were natural log (ln) transformed plasma GFAP, NF-L, UCH-L1, T-tau, and P-tau-181 concentrations, quantified on the Quanterix Simoa HD-X platform. Mixed-effects multi-variable linear regression was used to assess associations between biomarkers and self-reported previous concussion (yes/no), age (years), sex (male/female), objectively determined menstrual cycle phase (follicular/luteal), plasma progesterone, estradiol, and testosterone. Males had 19.8% lower UCH-L1 (β = -0.221, 95% confidence interval [CI; -0.396, -0.046]), 18.9% lower GFAP (β = -0.210, 95% CI [-0.352, -0.068]), and 21.8% higher P-tau-181 (β = 0.197, 95% CI [0.048, 0.346]) compared with females, adjusting for age and previous concussion. GFAP decreased 9.5% with each 1-year increase in age, adjusting for previous concussion and sex (β = -0.100, 95% CI [-0.152, -0.049]). No biomarkers were associated with a history of previous concussion. Exploratory investigations found no associations between biomarkers and menstrual cycle phase. Females displayed an age-adjusted negative association between T-tau and progesterone (β = -0.010, 95% CI [-0.018, -0.002]), whereas males had a negative age-adjusted association between UCH-L1 and testosterone (β = -0.020, 95% CI [-0.037, -0.002]). As such, age- and sex-specific reference intervals may be warranted for pediatric athlete populations prior to clinical validation of blood biomarkers for SRC. Additionally, hormonal associations highlight the need to consider puberty and development in adolescent studies. Overall, findings suggest these biomarkers are resilient to a history of previous concussion and menstrual cycle phase, supporting continued investigation in adolescent SRC.
Collapse
Affiliation(s)
- Jason Benjamin Tabor
- Sport Injury Prevention Research Center, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Linden Chase Penner
- Sport Injury Prevention Research Center, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jennifer Gradi Cooper
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mohammad Ghodsi
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jean-Michel Galarneau
- Sport Injury Prevention Research Center, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Douglas Dale Fraser
- Department of Pediatrics and Clinical Neurological Sciences, Western University, London, Ontario, Canada
| | - Carolyn Ann Emery
- Sport Injury Prevention Research Center, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Cheryl Lea Wellington
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Chantel Teresa Debert
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
17
|
Kasarinaite A, Sinton M, Saunders PTK, Hay DC. The Influence of Sex Hormones in Liver Function and Disease. Cells 2023; 12:1604. [PMID: 37371074 PMCID: PMC10296738 DOI: 10.3390/cells12121604] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
The liver performs a multitude of bodily functions, whilst retaining the ability to regenerate damaged tissue. In this review, we discuss sex steroid biology, regulation of mammalian liver physiology and the development of new model systems to improve our understanding of liver biology in health and disease. A major risk factor for the development of liver disease is hepatic fibrosis. Key drivers of this process are metabolic dysfunction and pathologic activation of the immune system. Although non-alcoholic fatty liver disease (NAFLD) is largely regarded as benign, it does progress to non-alcoholic steatohepatitis in a subset of patients, increasing their risk of developing cirrhosis and hepatocellular carcinoma. NAFLD susceptibility varies across the population, with obesity and insulin resistance playing a strong role in the disease development. Additionally, sex and age have been identified as important risk factors. In addition to the regulation of liver biochemistry, sex hormones also regulate the immune system, with sexual dimorphism described for both innate and adaptive immune responses. Therefore, sex differences in liver metabolism, immunity and their interplay are important factors to consider when designing, studying and developing therapeutic strategies to treat human liver disease. The purpose of this review is to provide the reader with a general overview of sex steroid biology and their regulation of mammalian liver physiology.
Collapse
Affiliation(s)
- Alvile Kasarinaite
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Matthew Sinton
- School of Biodiversity, One Health, and Veterinary Medicine, University of Glasgow, Glasgow G61 1QH, UK
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow G12 9TA, UK
| | - Philippa T. K. Saunders
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - David C. Hay
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| |
Collapse
|
18
|
Wei Y, Sun Y, Jia S, Yan P, Xiong C, Qi M, Wang C, Du Z, Jiang H. Identification of endogenous carbonyl steroids in human serum by chemical derivatization, hydrogen/deuterium exchange mass spectrometry and the quantitative structure-retention relationship. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1226:123776. [PMID: 37311272 DOI: 10.1016/j.jchromb.2023.123776] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/09/2023] [Accepted: 05/30/2023] [Indexed: 06/15/2023]
Abstract
Steroids are tetracyclic aliphatic compounds, and most of them contain carbonyl groups. The disordered homeostasis of steroids is closely related to the occurrence and progression of various diseases. Due to high structural similarity, low concentrations in vivo, poor ionization efficiency, and interference from endogenous substances, it is very challenging to comprehensively and unambiguously identify endogenous steroids in biological matrix. Herein, an integrated strategy was developed for the characterization of endogenous steroids in serum based on chemical derivatization, ultra-performance liquid chromatography quadrupole Exactive mass spectrometry (UPLC-Q-Exactive-MS/MS), hydrogen/deuterium (H/D) exchange, and a quantitative structure-retention relationship (QSRR) model. To enhance the mass spectrometry (MS) response of carbonyl steroids, the ketonic carbonyl group was derivatized by Girard T (GT). Firstly, the fragmentation rules of derivatized carbonyl steroid standards by GT were summarized. Then, carbonyl steroids in serum were derivatized by GT and identified based on the fragmentation rules or by comparing retention time and MS/MS spectra with those of standards. H/D exchange MS was utilized to distinguish derivatized steroid isomers for the first time. Finally, a QSRR model was constructed to predict the retention time of the unknown steroid derivatives. With this strategy, 93 carbonyl steroids were identified from human serum, and 30 of them were determined to be dicarbonyl steroids by the charge number of characteristic ions and the number of exchangeable hrdrogen or comparing with standards. The QSRR model built by the machine learning algorithms has an excellent regression correlation, thus the accurate structures of 14 carbonyl steroids were determined, among which three steroids were reported for the first time in human serum. This study provides a new analytical method for the comprehensive and reliable identification of carbonyl steroids in biological matrix.
Collapse
Affiliation(s)
- Yinyu Wei
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yi Sun
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shuailong Jia
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Pan Yan
- Department of Pharmacy, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410028, China
| | - Chaomei Xiong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Meiling Qi
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chenxi Wang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhifeng Du
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Hongliang Jiang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
19
|
Weng X, Zhu Q, Liao C, Jiang G. Cumulative Exposure to Phthalates and Their Alternatives and Associated Female Reproductive Health: Body Burdens, Adverse Outcomes, and Underlying Mechanisms. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023. [PMID: 37196176 DOI: 10.1021/acs.est.3c00823] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
The global birth rate has recently shown a decreasing trend, and exposure to environmental pollutants has been identified as a potential factor affecting female reproductive health. Phthalates have been widely used as plasticizers in plastic containers, children's toys, and medical devices, and their ubiquitous presence and endocrine-disrupting potential have already raised particular concerns. Phthalate exposure has been linked to various adverse health outcomes, including reproductive diseases. Given that many phthalates are gradually being banned, a growing number of phthalate alternatives are becoming popular, such as di(isononyl) cyclohexane-1,2-dicarboxylate (DINCH), di(2-ethylhexyl) adipate (DEHA), and di(2-ethylhexyl) terephthalate (DEHTP), and they are beginning to have a wide range of environmental effects. Studies have shown that many phthalate alternatives may disrupt female reproductive function by altering the estrous cycle, causing ovarian follicular atresia, and prolonging the gestational cycle, which raises growing concerns about their potential health risks. Herein, we summarize the effects of phthalates and their common alternatives in different female models, the exposure levels that influence the reproductive system, and the effects on female reproductive impairment, adverse pregnancy outcomes, and offspring development. Additionally, we scrutinize the effects of phthalates and their alternatives on hormone signaling, oxidative stress, and intracellular signaling to explore the underlying mechanisms of action on female reproductive health, because these chemicals may affect reproductive tissues directly or indirectly through endocrine disruption. Given the declining global trends of female reproductive capacity and the potential ability of phthalates and their alternatives to negatively impact female reproductive health, a more comprehensive study is needed to understand their effects on the human body and their underlying mechanisms. These findings may have an important role in improving female reproductive health and in turn decreasing the number of complications during pregnancy.
Collapse
Affiliation(s)
- Xueyu Weng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingqing Zhu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunyang Liao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
20
|
Ocañas SR, Ansere VA, Kellogg CM, Isola JVV, Chucair-Elliott AJ, Freeman WM. Chromosomal and gonadal factors regulate microglial sex effects in the aging brain. Brain Res Bull 2023; 195:157-171. [PMID: 36804773 PMCID: PMC10810555 DOI: 10.1016/j.brainresbull.2023.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023]
Abstract
Biological sex contributes to phenotypic sex effects through genetic (sex chromosomal) and hormonal (gonadal) mechanisms. There are profound sex differences in the prevalence and progression of age-related brain diseases, including neurodegenerative diseases. Inflammation of neural tissue is one of the most consistent age-related phenotypes seen with healthy aging and disease. The pro-inflammatory environment of the aging brain has primarily been attributed to microglial reactivity and adoption of heterogeneous reactive states dependent upon intrinsic (i.e., sex) and extrinsic (i.e., age, disease state) factors. Here, we review sex effects in microglia across the lifespan, explore potential genetic and hormonal molecular mechanisms of microglial sex effects, and discuss currently available models and methods to study sex effects in the aging brain. Despite recent attention to this area, significant further research is needed to mechanistically understand the regulation of microglial sex effects across the lifespan, which may open new avenues for sex informed prevention and treatment strategies.
Collapse
Affiliation(s)
- Sarah R Ocañas
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Victor A Ansere
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Collyn M Kellogg
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jose V V Isola
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Ana J Chucair-Elliott
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
21
|
Reddy KD, Oliver BGG. Sexual dimorphism in chronic respiratory diseases. Cell Biosci 2023; 13:47. [PMID: 36882807 PMCID: PMC9993607 DOI: 10.1186/s13578-023-00998-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Sex differences in susceptibility, severity, and progression are prevalent for various diseases in multiple organ systems. This phenomenon is particularly apparent in respiratory diseases. Asthma demonstrates an age-dependent pattern of sexual dimorphism. However, marked differences between males and females exist in other pervasive conditions such as chronic obstructive pulmonary disease (COPD) and lung cancer. The sex hormones estrogen and testosterone are commonly considered the primary factors causing sexual dimorphism in disease. However, how they contribute to differences in disease onset between males and females remains undefined. The sex chromosomes are an under-investigated fundamental form of sexual dimorphism. Recent studies highlight key X and Y-chromosome-linked genes that regulate vital cell processes and can contribute to disease-relevant mechanisms. This review summarises patterns of sex differences in asthma, COPD and lung cancer, highlighting physiological mechanisms causing the observed dimorphism. We also describe the role of the sex hormones and present candidate genes on the sex chromosomes as potential factors contributing to sexual dimorphism in disease.
Collapse
Affiliation(s)
- Karosham Diren Reddy
- Respiratory and Cellular Molecular Biology Group, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.
- School of Life Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| | - Brian Gregory George Oliver
- Respiratory and Cellular Molecular Biology Group, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia
- School of Life Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| |
Collapse
|
22
|
Rangsrikitphoti P, Marquez-Garban DC, Pietras RJ, McGowan E, Boonyaratanakornkit V. Sex steroid hormones and DNA repair regulation: Implications on cancer treatment responses. J Steroid Biochem Mol Biol 2023; 227:106230. [PMID: 36450315 DOI: 10.1016/j.jsbmb.2022.106230] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
The role of sex steroid hormones (SSHs) has been shown to modulate cancer cytotoxic treatment sensitivity. Dysregulation of DNA repair associated with genomic instability, abnormal cell survival and not only promotes cancer progression but also resistance to cancer treatment. The three major SSHs, androgen, estrogen, and progesterone, have been shown to interact with several essential DNA repair components. The presence of androgens directly regulates key molecules in DNA double-strand break (DSB) repair. Estrogen can promote cell proliferation and DNA repair, allowing cancer cells to tolerate chemotherapy and radiotherapy. Information on the role of progesterone in DNA repair is limited: progesterone interaction with some DNA repair components has been identified, but the biological significance is still unknown. Here, we review the roles of how each SSH affects DNA repair regulation and modulates response to genotoxic therapies and discuss future research that can be beneficial when combining SSHs with cancer therapy. We also provide preliminary analysis from publicly available databases defining the link between progesterone/PR and DDRs & DNA repair regulation that plausibly contribute to chemotherapy response and breast cancer patient survival.
Collapse
Affiliation(s)
- Pattarasiri Rangsrikitphoti
- Graduate Program in Clinical Biochemistry and Molecular Medicine and Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Diana C Marquez-Garban
- UCLA Jonsson Comprehensive Cancer and Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Richard J Pietras
- UCLA Jonsson Comprehensive Cancer and Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Eileen McGowan
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Viroj Boonyaratanakornkit
- Graduate Program in Clinical Biochemistry and Molecular Medicine and Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Age-related Inflammation and Degeneration Research Unit, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
23
|
Di Cosola M, Spirito F, Zhurakivska K, Nocini R, Lovero R, Sembronio S, Santacroce L, Brauner E, Storto G, Lo Muzio L, Cazzolla AP. Congenital adrenal hyperplasia. Role of dentist in early diagnosis. Open Med (Wars) 2022; 17:1699-1704. [PMID: 36382053 PMCID: PMC9616050 DOI: 10.1515/med-2022-0524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/24/2022] [Accepted: 06/21/2022] [Indexed: 11/07/2022] Open
Abstract
Congenital adrenal hyperplasia (CAH) is a genetic disorder characterized by an impairment of steroid synthesis due to an altered production of 21-hydroxylase enzyme. Corticoid hormones are involved in the development and functioning of many organs. The aim of the present study was to review the international literature to collect data regarding oral manifestations of CAH. A review of the literature describing oral features of patients affected by CAH was performed using electronic databases (PubMed and Scopus). The data about number of patients, form of CAH, and oral findings were extracted and analyzed. Seven studies were included in the final analysis. The principal findings reported regarded an advanced dental development observed in patients with CAH. One paper reported amelogenesis imperfecta and periodontal issues. The dentist could be the first specialist involved in the CAH syndrome diagnosis, identifying the characteristic features described above, especially for the classical simple virilizing and non-classical form.
Collapse
Affiliation(s)
- Michele Di Cosola
- Department of Clinical and Experimental Medicine, University of Foggia , Foggia , Italy
| | - Francesca Spirito
- Department of Clinical and Experimental Medicine, University of Foggia , Foggia , Italy
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana,” University of Salerno , Via Allende, 84081 , Baronissi (SA) , Italy
| | - Khrystyna Zhurakivska
- Department of Clinical and Experimental Medicine, University of Foggia , Foggia , Italy
| | - Riccardo Nocini
- ENT Department, University of Verona , 37100 , Verona , Italy
| | - Roberto Lovero
- Clinical Pathology Unit, AOU Policlinico Consorziale di Bari - Ospedale Giovanni XXIII , 70124 , Bari , Italy
| | - Salvatore Sembronio
- Department of Maxillofacial Surgery, University of Udine, Academic Hospital “Santa Maria della Misericordia,” , 33100 , Udine , Italy
| | - Luigi Santacroce
- Department of Interdisciplinary Medicine, Microbiology and Virology Laboratory, University Hospital of Bari, Università degli Studi di Bari , 70124 , Bari , Italy
| | - Edoardo Brauner
- Department of Dental and Maxillary Facial Sciences, “Sapienza” University of Rome , 00185 , Rome , Italy
| | - Giovanni Storto
- Nuclear Medicine Unit, IRCCS Regional Cancer Hospital CROB, Referral Cancer Center of Basilicata , 85028 , Potenza , Italy
| | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, University of Foggia , Foggia , Italy
| | - Angela Pia Cazzolla
- Department of Clinical and Experimental Medicine, University of Foggia , Foggia , Italy
| |
Collapse
|
24
|
Paredes-Amaya CC, Manzano-Gayosso P, Hernández-Hernández F. Identification of Differentially Expressed Genes in Nocardia brasiliensis Induced by Progesterone and Dihydrotestosterone Using Differential Display PCR. Curr Microbiol 2022; 79:335. [PMID: 36201047 DOI: 10.1007/s00284-022-03028-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022]
Abstract
Sex steroid hormones have an important physiological role in humans. They can also affect the gene expression of many organisms, including bacteria. In Mexico, Nocardia brasiliensis is the main causative agent of actinomycetoma, a granulomatous disease more frequent in men than women, which is thought to be related to a higher occupational risk in men. Therefore, it has been suggested that differences in clinical presentation could be related to sex steroid hormone levels. Attempting to explain the differences in actinomycetoma prevalence between men and women, in this work, the effect of progesterone and dihydrotestosterone on the genetic expression of N. brasiliensis was investigated using a differential display polymerase chain reaction assay. The results showed that both hormones affected the expression of genes encoding proteins related to central metabolism and hypothetical proteins with unknown functions. This study also demonstrated the utility of differential display in this modern era and provided a first approach to the effect of sex hormones on N. brasiliensis gene expression.
Collapse
Affiliation(s)
- Claudia C Paredes-Amaya
- Departamento de Microbiología, Escuela de Ciencias Básicas, Facultad de Salud, Universidad del Valle, Cali, Colombia.,Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Pontificia Universidad Javeriana Cali, Cali, Colombia
| | - Patricia Manzano-Gayosso
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Francisca Hernández-Hernández
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
25
|
Beneficial Effects of Flaxseed and/or Mulberry Extracts Supplementation in Ovariectomized Wistar Rats. Nutrients 2022; 14:nu14153238. [PMID: 35956414 PMCID: PMC9370575 DOI: 10.3390/nu14153238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/14/2022] [Accepted: 07/14/2022] [Indexed: 11/29/2022] Open
Abstract
Low endogenous estrogen action causes several injuries. Medicinal plants, such as flaxseed and mulberry, contain substances that have been shown to be effective to the organism. The aim was to verify the effects of flaxseed and/or mulberry extracts on ovariectomized Wistar rats. The animals received supplements of extracts and estrogen or saline by gavage for 60 days and were weighed weekly. Vaginal wash, blood, pituitary, uterus, liver, and kidneys were collected. Phenolic compounds and the antioxidant activity of the extracts, lipid profile, uric acid, liver enzymes, and pituitary weight were measured. Histomorphometric for uterine wall and histopathological analyses for liver and kidney were performed. Flaxseed and mulberry extracts showed great antioxidant activity and large amounts of phenolic compounds. The treatment with extracts had less weight gain, increased pituitary weight, the predominance of vaginal epithelial cells, and reduced TC, LDL-c and lipase activity, similar to estrogen animals. Estrogen or flaxseed + mulberry animals reduced VLDL-c and TAG. HDL-c, uric acid, and liver enzymes did not differ. Estrogen or extracts demonstrated trophic action on the endometrial thickness and have not shown hepatotoxicity or nephrotoxicity. We suggested the beneficial effects of flaxseed and mulberry extract as an alternative to reduce and/or prevent the negative effects caused by low estrogenic action.
Collapse
|
26
|
Winek K, Tzur Y, Soreq H. Biological underpinnings of sex differences in neurological disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 164:27-67. [PMID: 36038206 DOI: 10.1016/bs.irn.2022.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The importance of sex differences in neurological disorders has been increasingly acknowledged in recent clinical and basic research studies, but the complex biology and genetics underlying sex-linked biological heterogeneity and its brain-to-body impact remained incompletely understood. Men and women differ substantially in their susceptibility to certain neurological diseases, in the severity of symptoms, prognosis as well as the nature and efficacy of their response to treatments. The detailed mechanisms underlying these differences, especially at the molecular level, are being addressed in many studies but leave a lot to be further revealed. Here, we provide an overview of recent advances in our understanding of how sex differences in the brain and brain-body signaling contribute to neurological disorders and further present some future prospects entailed in terms of diagnostics and therapeutics.
Collapse
Affiliation(s)
- Katarzyna Winek
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel; The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yonat Tzur
- The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hermona Soreq
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel; The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
27
|
Krentzel AA, Proaño SB, Dorris DM, Setzer B, Meitzen J. The estrous cycle and 17β-estradiol modulate the electrophysiological properties of rat nucleus accumbens core medium spiny neurons. J Neuroendocrinol 2022; 34:e13122. [PMID: 35365910 PMCID: PMC9250601 DOI: 10.1111/jne.13122] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/02/2022] [Accepted: 02/22/2022] [Indexed: 12/03/2022]
Abstract
The nucleus accumbens core is a key nexus within the mammalian brain for integrating the premotor and limbic systems and regulating important cognitive functions such as motivated behaviors. Nucleus accumbens core functions show sex differences and are sensitive to the presence of hormones such as 17β-estradiol (estradiol) in normal and pathological contexts. The primary neuron type of the nucleus accumbens core, the medium spiny neuron (MSN), exhibits sex differences in both intrinsic excitability and glutamatergic excitatory synapse electrophysiological properties. Here, we provide a review of recent literature showing how estradiol modulates rat nucleus accumbens core MSN electrophysiology within the context of the estrous cycle. We review the changes in MSN electrophysiological properties across the estrous cycle and how these changes can be mimicked in response to exogenous estradiol exposure. We discuss in detail recent findings regarding how acute estradiol exposure rapidly modulates excitatory synapse properties in nucleus accumbens core but not caudate-putamen MSNs, which mirror the natural changes seen across estrous cycle phases. These recent insights demonstrate the strong impact of sex-specific estradiol action upon nucleus accumbens core neuron electrophysiology.
Collapse
Affiliation(s)
- Amanda A. Krentzel
- Department of Biological SciencesNorth Carolina State UniversityRaleighNCUSA
| | - Stephanie B. Proaño
- Neurobiology LaboratoryNational Institute of Environmental Health Sciences, NIHResearch Triangle ParkNCUSA
| | - David M. Dorris
- Department of Biological SciencesNorth Carolina State UniversityRaleighNCUSA
| | - Beverly Setzer
- Graduate Program for Neuroscience and Department of Biomedical EngineeringBoston UniversityBostonMAUSA
| | - John Meitzen
- Department of Biological SciencesNorth Carolina State UniversityRaleighNCUSA
- Comparative Medicine InstituteNorth Carolina State UniversityRaleighNCUSA
- Center for Human Health and the EnvironmentNorth Carolina State UniversityRaleighNCUSA
| |
Collapse
|
28
|
Jiang J, Su S, Lai T, Feng W, Li F, Tian C, Gao Y, Munganga BP, Tang Y, Xu P. Recognition of Gonadal Development in Eriocheir sinensis Based on the Impulse of Love at First Sight. Front Physiol 2022; 13:793699. [PMID: 35574457 PMCID: PMC9091178 DOI: 10.3389/fphys.2022.793699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 03/21/2022] [Indexed: 12/02/2022] Open
Abstract
Given the difficulty in identifying individuals with different degrees of ovarian development, we developed a new device utilizing the hypothesis of mutual attraction behavior between male and female crabs with mature gonads by releasing the sexual pheromone so they could be examined. From a total of 40 female crabs, 10 were isolated within half an hour. Histological analysis showed that the ovaries of crabs in the isolated group were in stage IV, while those of the control groups were in stage III. In addition, progesterone (PROG) in experimental groups was significantly reduced compared with the control group (p < 0.05), but no significant difference was detected in estradiol (E2). In response to the different developmental stages, hemolymph biochemical indices and the determination of gonadal fatty acids profiles were explored. The results indicated only C18:4 showed a significant difference between these two groups. A transcriptome was generated to determine the genes involved in the mutual attraction process; differentially expressed genes (DEGs) were significantly related to gonadal development. Therefore, the device can be used to isolate Chinese mitten crabs with stage IV ovarian development.
Collapse
Affiliation(s)
- Jingjing Jiang
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Shengyan Su
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China.,Key Laboratory of Genetic Breeding and Aquaculture Biology of Freshwater Fishes, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Ting Lai
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Wenrong Feng
- Key Laboratory of Genetic Breeding and Aquaculture Biology of Freshwater Fishes, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Feifan Li
- Key Laboratory of Genetic Breeding and Aquaculture Biology of Freshwater Fishes, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Can Tian
- National Demonstration Center for Expermental Fisherise Science Education, Shanghai Ocean University, Shanghai, China
| | - Yang Gao
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | | | - Yongkai Tang
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China.,Key Laboratory of Genetic Breeding and Aquaculture Biology of Freshwater Fishes, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Pao Xu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China.,Key Laboratory of Genetic Breeding and Aquaculture Biology of Freshwater Fishes, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| |
Collapse
|
29
|
Liang X, Luo X, Lin H, Han F, Qin JG, Chen L, Xu C, Li E. Effects and Mechanism of Different Phospholipid Diets on Ovary Development in Female Broodstock Pacific White Shrimp, Litopenaeus vannamei. Front Nutr 2022; 9:830934. [PMID: 35252307 PMCID: PMC8894211 DOI: 10.3389/fnut.2022.830934] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/19/2022] [Indexed: 01/15/2023] Open
Abstract
Research on nutrition and feed development for the broodstock of the Pacific white shrimp, Litopenaeus vannamei, is rare, and a poor broodstock quality is a critical factor restricting the seed supply in shrimp farming. As an essential nutrient for the gonadal development of L. vannamei, one control diet (no phospholipid) and three typical phospholipids (soybean lecithin, egg yolk lecithin, and krill oil) were evaluated in a semipurified diet of 4% phospholipid for a 28-day trial (initial weight 34.7 ± 4.2 g). Dietary phospholipid supplementation significantly promoted the ovarian maturation of female L. vannamei. Compared with soybean lecithin and egg yolk lecithin, krill oil showed the best positive results. Shrimp fed with a diet krill oil has obtained a significantly higher gonadosomatic index, yolk particle deposition, lipid accumulation, and estrogen secretion than from other sources. Ovary lipidomic analysis showed that the krill oil enriched the lipid composition of the ovary. The “glycerophospholipid metabolism” and “sphingolipid metabolism” pathways were significantly varied via topological pathway analysis. Genes and hub genes, with significantly different expression levels, were significantly enriched in the “fatty acid metabolism pathway,” “glycerophospholipid metabolism,” and “arachidonic acid metabolism” pathways by transcriptomic analysis. Correlation analysis of the transcriptome and lipidomics showed that the differential gene “hormone-sensitive lipase-like” (HSL) was positively correlated with various lipids [triglycerides (TG), phosphatidic acid (PA), phosphatidylserine (P), phosphatidylethanolamine (PE), glucosylceramide (GlcCer), phosphatidylglycerol (PG), and phosphatidylinositol (PI)] but was negatively correlated with diacylglycerol (DG), lysophosphatidylethanolamine (LPE), and sphingomyelin (SM). In conclusion, the dietary phospholipids, especially krill oil as a phospholipid source, can promote the development of L. vannamei ovaries by increasing the accumulation of nutrients such as triglycerides and sterols, and the secretion of estrogen or related hormones, such as estradiol and methylfarneside, by affecting the metabolism of glycerol phospholipids and some key fatty acids.
Collapse
Affiliation(s)
- Xiaolong Liang
- Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan Aquaculture Breeding Engineering Research Center, College of Marine Sciences, Hainan University, Haikou, China
| | - Xiaolong Luo
- Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan Aquaculture Breeding Engineering Research Center, College of Marine Sciences, Hainan University, Haikou, China
| | - Hongxing Lin
- Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan Aquaculture Breeding Engineering Research Center, College of Marine Sciences, Hainan University, Haikou, China
| | - Fenglu Han
- Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan Aquaculture Breeding Engineering Research Center, College of Marine Sciences, Hainan University, Haikou, China
| | - Jian G Qin
- School of Biological Sciences, Flinders University, Adelaide, SA, Australia
| | - Liqiao Chen
- School of Life Sciences, East China Normal University, Shanghai, China
| | - Chang Xu
- Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan Aquaculture Breeding Engineering Research Center, College of Marine Sciences, Hainan University, Haikou, China
| | - Erchao Li
- Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan Aquaculture Breeding Engineering Research Center, College of Marine Sciences, Hainan University, Haikou, China
| |
Collapse
|
30
|
Targeting Estrogens and Various Estrogen-Related Receptors against Non-Small Cell Lung Cancers: A Perspective. Cancers (Basel) 2021; 14:cancers14010080. [PMID: 35008242 PMCID: PMC8750572 DOI: 10.3390/cancers14010080] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/08/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022] Open
Abstract
Non-small cell lung cancers (NSCLCs) account for ~85% of lung cancer cases worldwide. Mammalian lungs are exposed to both endogenous and exogenous estrogens. The expression of estrogen receptors (ERs) in lung cancer cells has evoked the necessity to evaluate the role of estrogens in the disease progression. Estrogens, specifically 17β-estradiol, promote maturation of several tissue types including lungs. Recent epidemiologic data indicate that women have a higher risk of lung adenocarcinoma, a type of NSCLC, when compared to men, independent of smoking status. Besides ERs, pulmonary tissues both in healthy physiology and in NSCLCs also express G-protein-coupled ERs (GPERs), epidermal growth factor receptor (EGFRs), estrogen-related receptors (ERRs) and orphan nuclear receptors. Premenopausal females between the ages of 15 and 50 years synthesize a large contingent of estrogens and are at a greater risk of developing NSCLCs. Estrogen-ER/GPER/EGFR/ERR-mediated activation of various cell signaling molecules regulates NSCLC cell proliferation, survival and apoptosis. This article sheds light on the most recent achievements in the elucidation of sequential biochemical events in estrogen-activated cell signaling pathways involved in NSCLC severity with insight into the mechanism of regulation by ERs/GPERs/EGFRs/ERRs. It further discusses the success of anti-estrogen therapies against NSCLCs.
Collapse
|
31
|
Ding J, You S, Zhang J, Zhang H, Wang H, Zhang W, Qi W, Su R, He Z. Rational design of 17β-hydroxysteroid dehydrogenase type3 for improving testosterone production with an engineered Pichia pastoris. BIORESOURCE TECHNOLOGY 2021; 341:125833. [PMID: 34455250 DOI: 10.1016/j.biortech.2021.125833] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 06/13/2023]
Abstract
Testosterone (TS) is a critical androgenic steroid that regulates human metabolism and maintains secondary sexual characteristics. The biotransformation from 4-androstene-3,17-done (4-AD) to TS is limited by the poor catalytic activity of 17β-hydroxysteroid dehydrogenase type 3 (17β-HSD3). Herein, we explored the structural characteristics and catalytic mechanism of 17β-HSD3 and adopted the rational design strategy to improve its catalytic activity. Molecular docking and molecular dynamics simulations revealed the substrate-binding pocket and the binding mode of 4-AD to 17β-HSD3. We located the pivotal residues and regulated their hydrophobicity and polarity. The obtained G186R/Y195W variant formed additional electrostatic interaction and hydrogen bond with 4-AD, increasing the binding affinity between the variant and 4-AD. Therefore, the G186R/Y195W variant produced 3.98 g/L of TS, which increased to 297%. The combination of structural and mechanism resolution drives the implementation of the rational design strategy, which provides guidance for bioproduction of TS catalyzed by 17β-HSD3.
Collapse
Affiliation(s)
- Juanjuan Ding
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China
| | - Shengping You
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China; Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, PR China
| | - Jiaxing Zhang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China
| | - Hongtao Zhang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China
| | - Hui Wang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China
| | - Wei Zhang
- Ever-Sky Bioscience (Tianjin) Co., Ltd., PR China; Biosyn Healthy Pharma Co., Ltd, PR China
| | - Wei Qi
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China; State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300350, PR China; Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, PR China; Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, PR China.
| | - Rongxin Su
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China; State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300350, PR China; Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, PR China; Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, PR China
| | - Zhimin He
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China; State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300350, PR China
| |
Collapse
|
32
|
Lipoldová M, Demant P. Gene-Specific Sex Effects on Susceptibility to Infectious Diseases. Front Immunol 2021; 12:712688. [PMID: 34721380 PMCID: PMC8553003 DOI: 10.3389/fimmu.2021.712688] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammation is an integral part of defense against most infectious diseases. These pathogen-induced immune responses are in very many instances strongly influenced by host’s sex. As a consequence, sexual dimorphisms were observed in susceptibility to many infectious diseases. They are pathogen dose-dependent, and their outcomes depend on pathogen and even on its species or subspecies. Sex may differentially affect pathology of various organs and its influence is modified by interaction of host’s hormonal status and genotype: sex chromosomes X and Y, as well as autosomal genes. In this Mini Review we summarize the major influences of sex in human infections and subsequently focus on 22 autosomal genes/loci that modify in a sex-dependent way the response to infectious diseases in mouse models. These genes have been observed to influence susceptibility to viruses, bacteria, parasites, fungi and worms. Some sex-dependent genes/loci affect susceptibility only in females or only in males, affect both sexes, but have stronger effect in one sex; still other genes were shown to affect the disease in both sexes, but with opposite direction of effect in females and males. The understanding of mechanisms of sex-dependent differences in the course of infectious diseases may be relevant for their personalized management.
Collapse
Affiliation(s)
- Marie Lipoldová
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Peter Demant
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
33
|
Jacoby C, Goerke M, Bezold D, Jessen H, Boll M. A fully reversible 25-hydroxy steroid kinase involved in oxygen-independent cholesterol side-chain oxidation. J Biol Chem 2021; 297:101105. [PMID: 34425106 PMCID: PMC8449060 DOI: 10.1016/j.jbc.2021.101105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 11/29/2022] Open
Abstract
The degradation of cholesterol and related steroids by microbes follows fundamentally different strategies in aerobic and anaerobic environments. In anaerobic bacteria, the primary C26 of the isoprenoid side chain is hydroxylated without oxygen via a three-step cascade: (i) water-dependent hydroxylation at the tertiary C25, (ii) ATP-dependent dehydration to form a subterminal alkene, and (iii) water-dependent hydroxylation at the primary C26 to form an allylic alcohol. However, the enzymes involved in the ATP-dependent dehydration have remained unknown. Here, we isolated an ATP-dependent 25-hydroxy-steroid kinase (25-HSK) from the anaerobic bacterium Sterolibacterium denitrificans. This highly active enzyme preferentially phosphorylated the tertiary C25 of steroid alcohols, including metabolites of cholesterol and sitosterol degradation or 25-OH-vitamin D3. Kinetic data were in agreement with a sequential mechanism via a ternary complex. Remarkably, 25-HSK readily catalyzed the formation of γ-(18O)2-ATP from ADP and the C25-(18O)2-phosphoester. The observed full reversibility of 25-HSK with an equilibrium constant below one can be rationalized by an unusual high phosphoryl transfer potential of tertiary steroid C25-phosphoesters, which is ≈20 kJ mol−1 higher than that of standard sugar phosphoesters and even slightly greater than the β,γ-phosphoanhydride of ATP. In summary, 25-HSK plays an essential role in anaerobic bacterial degradation of zoo- and phytosterols and shows only little similarity to known phosphotransferases.
Collapse
Affiliation(s)
- Christian Jacoby
- Faculty of Biology, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Malina Goerke
- Faculty of Biology, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Dominik Bezold
- Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Henning Jessen
- Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Matthias Boll
- Faculty of Biology, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany.
| |
Collapse
|
34
|
Raut S, Kumar AV, Deshpande S, Khambata K, Balasinor NH. Sex hormones regulate lipid metabolism in adult Sertoli cells: A genome-wide study of estrogen and androgen receptor binding sites. J Steroid Biochem Mol Biol 2021; 211:105898. [PMID: 33845154 DOI: 10.1016/j.jsbmb.2021.105898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/16/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022]
Abstract
Optimal functioning of Sertoli cells is crucial for spermatogenesis which is under tight regulation of sex hormones, estrogen and androgen. Adult rat Sertoli cells expresses estrogen receptor beta (ERβ) and androgen receptor (AR), both of which regulate gene transcription by binding to the DNA. The present study is aimed to acquire a genome-wide map of estrogen- and androgen-regulated genes in adult Sertoli cells. ChIP-Seq was performed for ERβ and AR in Sertoli cells under physiological conditions. 30,859 peaks in ERβ and 9,594 peaks in AR were identified with a fold enrichment >2 fold. Pathway analysis for the genes revealed metabolic pathways to be significantly enriched. Since Sertoli cells have supportive functions and provide energy substrates to germ cells during spermatogenesis, significantly enriched metabolic pathways were explored further. Peaks of the genes involved in lipid metabolism, like fatty acid, glyceride, leucine, and sphingosine metabolism were validated. Motif analysis confirmed the presence of estrogen- and androgen-response elements (EREs and AREs). Moreover, transcript levels of enzymes involved in the lipid metabolic pathways were significantly altered in cultured Sertoli cells treated with estrogen and androgen receptor agonists, demonstrating functional significance of these binding sites. This study elucidates a mechanism by which sex hormones regulate lipid metabolism in Sertoli cells by transcriptionally controlling the expression of these genes, thereby shedding light on the roles of these hormones in male fertility.
Collapse
Affiliation(s)
- Sanketa Raut
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Anita V Kumar
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Sharvari Deshpande
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Kushaan Khambata
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Nafisa H Balasinor
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive Health, Mumbai, India.
| |
Collapse
|
35
|
You HH, Song G. Review of endocrine disruptors on male and female reproductive systems. Comp Biochem Physiol C Toxicol Pharmacol 2021; 244:109002. [PMID: 33610819 DOI: 10.1016/j.cbpc.2021.109002] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/03/2021] [Accepted: 02/11/2021] [Indexed: 12/21/2022]
Abstract
Endocrine disruptors (EDs) interfere with different hormonal and metabolic processes and disrupt the development of organs and tissues, as well as the reproductive system. In toxicology research, various animal models have been utilized to compare and characterize the effects of EDs. We reviewed studies assessing the effect of ED exposure in humans, zebrafish, and mouse models and the adverse effects of EDs on male and female reproductive systems. This review outlines the distinctive morphological characteristics, as well as gene expression, factors, and mechanisms that are known to occur in response to EDs. In each animal model, disturbances in the reproductive system were associated with certain factors of apoptosis, the hypothalamic-pituitary-gonadal axis, estrogen receptor pathway-induced meiotic disruption, and steroidogenesis. The effects of bisphenol A, phthalate, and 17α-ethinylestradiol have been investigated in animal models, each providing supporting outcomes and elaborating the key regulators of male and female reproductive systems.
Collapse
Affiliation(s)
- Hyekyoung Hannah You
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
36
|
Hall JM, Korach KS. Endocrine disrupting chemicals (EDCs) and sex steroid receptors. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 92:191-235. [PMID: 34452687 DOI: 10.1016/bs.apha.2021.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sex-steroid receptors (SSRs) are essential mediators of estrogen, progestin, and androgen signaling that are critical in vast aspects of human development and multi-organ homeostasis. Dysregulation of SSR function has been implicated in numerous pathologies including cancers, obesity, Type II diabetes mellitus, neuroendocrine disorders, cardiovascular disease, hyperlipidemia, male and female infertility, and other reproductive disorders. Endocrine disrupting chemicals (EDCs) modulate SSR function in a wide variety of cell and tissues. There exists strong experimental, clinical, and epidemiological evidence that engagement of EDCs with SSRs may disrupt endogenous hormone signaling leading to physiological abnormalities that may manifest in disease. In this chapter, we discuss the molecular mechanisms by which EDCs interact with estrogen, progestin, and androgen receptors and alter SSR functions in target cells. In addition, the pathological consequences of disruption of SSR action in reproductive and other organs by EDCs is described with an emphasis on underlying mechanisms of receptors dysfunction.
Collapse
Affiliation(s)
- Julianne M Hall
- Quinnipiac University Frank H. Netter MD School of Medicine, Hamden, CT, United States.
| | - Kenneth S Korach
- National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, United States
| |
Collapse
|
37
|
Sim CB, Phipson B, Ziemann M, Rafehi H, Mills RJ, Watt KI, Abu-Bonsrah KD, Kalathur RK, Voges HK, Dinh DT, ter Huurne M, Vivien CJ, Kaspi A, Kaipananickal H, Hidalgo A, Delbridge LM, Robker RL, Gregorevic P, dos Remedios CG, Lal S, Piers AT, Konstantinov IE, Elliott DA, El-Osta A, Oshlack A, Hudson JE, Porrello ER. Sex-Specific Control of Human Heart Maturation by the Progesterone Receptor. Circulation 2021; 143:1614-1628. [PMID: 33682422 PMCID: PMC8055196 DOI: 10.1161/circulationaha.120.051921] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/29/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Despite in-depth knowledge of the molecular mechanisms controlling embryonic heart development, little is known about the signals governing postnatal maturation of the human heart. METHODS Single-nucleus RNA sequencing of 54 140 nuclei from 9 human donors was used to profile transcriptional changes in diverse cardiac cell types during maturation from fetal stages to adulthood. Bulk RNA sequencing and the Assay for Transposase-Accessible Chromatin using sequencing were used to further validate transcriptional changes and to profile alterations in the chromatin accessibility landscape in purified cardiomyocyte nuclei from 21 human donors. Functional validation studies of sex steroids implicated in cardiac maturation were performed in human pluripotent stem cell-derived cardiac organoids and mice. RESULTS Our data identify the progesterone receptor as a key mediator of sex-dependent transcriptional programs during cardiomyocyte maturation. Functional validation studies in human cardiac organoids and mice demonstrate that the progesterone receptor drives sex-specific metabolic programs and maturation of cardiac contractile properties. CONCLUSIONS These data provide a blueprint for understanding human heart maturation in both sexes and reveal an important role for the progesterone receptor in human heart development.
Collapse
Affiliation(s)
- Choon Boon Sim
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Belinda Phipson
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre (B.P., A.O.), University of Melbourne, Victoria, Australia
| | - Mark Ziemann
- Department of Diabetes, Central Clinical School, Alfred Centre, Monash University, Melbourne, Victoria, Australia (M.Z., H.R., A.K., H.K., A.E.-O.)
- School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Victoria, Australia (M.Z.)
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.Z., H.R., K.I.W., A.K., H.K., P.G., A.E.-O.)
| | - Haloom Rafehi
- Department of Diabetes, Central Clinical School, Alfred Centre, Monash University, Melbourne, Victoria, Australia (M.Z., H.R., A.K., H.K., A.E.-O.)
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.Z., H.R., K.I.W., A.K., H.K., P.G., A.E.-O.)
| | - Richard J. Mills
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia (R.J.M., J.E.H.)
| | - Kevin I. Watt
- Peter MacCallum Cancer Centre (B.P., A.O.), University of Melbourne, Victoria, Australia
- Centre for Muscle Research (K.I.W., P.G., E.R.P.), University of Melbourne, Victoria, Australia
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.Z., H.R., K.I.W., A.K., H.K., P.G., A.E.-O.)
| | - Kwaku D. Abu-Bonsrah
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences, and Department of Paediatrics (K.D.A.-B., H.K.V., A.H., I.E.K., D.A.E.), University of Melbourne, Victoria, Australia
| | - Ravi K.R. Kalathur
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Holly K. Voges
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences, and Department of Paediatrics (K.D.A.-B., H.K.V., A.H., I.E.K., D.A.E.), University of Melbourne, Victoria, Australia
| | - Doan T. Dinh
- Robinson Research Institute, The University of Adelaide, South Australia, Australia (D.T.D., R.L.R.)
| | - Menno ter Huurne
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Celine J. Vivien
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Antony Kaspi
- Department of Diabetes, Central Clinical School, Alfred Centre, Monash University, Melbourne, Victoria, Australia (M.Z., H.R., A.K., H.K., A.E.-O.)
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.Z., H.R., K.I.W., A.K., H.K., P.G., A.E.-O.)
| | - Harikrishnan Kaipananickal
- Department of Diabetes, Central Clinical School, Alfred Centre, Monash University, Melbourne, Victoria, Australia (M.Z., H.R., A.K., H.K., A.E.-O.)
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.Z., H.R., K.I.W., A.K., H.K., P.G., A.E.-O.)
| | - Alejandro Hidalgo
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences, and Department of Paediatrics (K.D.A.-B., H.K.V., A.H., I.E.K., D.A.E.), University of Melbourne, Victoria, Australia
| | - Leanne M.D. Delbridge
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology (K.I.W., L.M.D.D., P.G., E.R.P.), University of Melbourne, Victoria, Australia
| | - Rebecca L. Robker
- Robinson Research Institute, The University of Adelaide, South Australia, Australia (D.T.D., R.L.R.)
| | - Paul Gregorevic
- Department of Anatomy and Physiology (K.I.W., L.M.D.D., P.G., E.R.P.), University of Melbourne, Victoria, Australia
- Centre for Muscle Research (K.I.W., P.G., E.R.P.), University of Melbourne, Victoria, Australia
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.Z., H.R., K.I.W., A.K., H.K., P.G., A.E.-O.)
| | - Cristobal G. dos Remedios
- School of Medical Sciences, The University of Sydney, New South Wales, Australia (C.G.d.R., S.L.)
- Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia (C.G.d.R.)
| | - Sean Lal
- School of Medical Sciences, The University of Sydney, New South Wales, Australia (C.G.d.R., S.L.)
| | - Adam T. Piers
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Igor E. Konstantinov
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Department of Cardiac Surgery (I.E.K.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences, and Department of Paediatrics (K.D.A.-B., H.K.V., A.H., I.E.K., D.A.E.), University of Melbourne, Victoria, Australia
| | - David A. Elliott
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences, and Department of Paediatrics (K.D.A.-B., H.K.V., A.H., I.E.K., D.A.E.), University of Melbourne, Victoria, Australia
| | - Assam El-Osta
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Alicia Oshlack
- Department of Diabetes, Central Clinical School, Alfred Centre, Monash University, Melbourne, Victoria, Australia (M.Z., H.R., A.K., H.K., A.E.-O.)
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.Z., H.R., K.I.W., A.K., H.K., P.G., A.E.-O.)
- Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, Li Ka Shing Institute of Health Sciences, and The Chinese University of Hong Kong, China (A.E.-O.)
| | - James E. Hudson
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia (R.J.M., J.E.H.)
- Centre for Cardiac and Vascular Biology, School of Biomedical Sciences, The University of Queensland, Brisbane, Australia (J.E.H., E.R.P.)
| | - Enzo R. Porrello
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology (K.I.W., L.M.D.D., P.G., E.R.P.), University of Melbourne, Victoria, Australia
- Centre for Muscle Research (K.I.W., P.G., E.R.P.), University of Melbourne, Victoria, Australia
- Centre for Cardiac and Vascular Biology, School of Biomedical Sciences, The University of Queensland, Brisbane, Australia (J.E.H., E.R.P.)
| |
Collapse
|
38
|
Ambhore NS, Kalidhindi RSR, Sathish V. Sex-Steroid Signaling in Lung Diseases and Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:243-273. [PMID: 33788197 DOI: 10.1007/978-3-030-63046-1_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sex/gender difference exists in the physiology of multiple organs. Recent epidemiological reports suggest the influence of sex-steroids in modulating a wide variety of disease conditions. Sex-based discrepancies have been reported in pulmonary physiology and various chronic inflammatory responses associated with lung diseases like asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis, and rare lung diseases. Notably, emerging clinical evidence suggests that several respiratory diseases affect women to a greater degree, with increased severity and prevalence than men. Although sex-specific differences in various lung diseases are evident, such differences are inherent to sex-steroids, which are major biological variables in men and women who play a central role to control these differences. The focus of this chapter is to comprehend the sex-steroid biology in inflammatory lung diseases and to understand the mechanistic role of sex-steroids signaling in regulating these diseases. Exploring the roles of sex-steroid signaling in the regulation of lung diseases and inflammation is crucial for the development of novel and effective therapy. Overall, we will illustrate the importance of differential sex-steroid signaling in lung diseases and their possible clinical implications for the development of complementary and alternative medicine to treat lung diseases.
Collapse
Affiliation(s)
- Nilesh Sudhakar Ambhore
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | | | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA.
| |
Collapse
|
39
|
Methodological Considerations for Studies in Sport and Exercise Science with Women as Participants: A Working Guide for Standards of Practice for Research on Women. Sports Med 2021; 51:843-861. [PMID: 33725341 PMCID: PMC8053180 DOI: 10.1007/s40279-021-01435-8] [Citation(s) in RCA: 327] [Impact Index Per Article: 81.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2021] [Indexed: 12/25/2022]
Abstract
Until recently, there has been less demand for and interest in female-specific sport and exercise science data. As a result, the vast majority of high-quality sport and exercise science data have been derived from studies with men as participants, which reduces the application of these data due to the known physiological differences between the sexes, specifically with regard to reproductive endocrinology. Furthermore, a shortage of specialist knowledge on female physiology in the sport science community, coupled with a reluctance to effectively adapt experimental designs to incorporate female-specific considerations, such as the menstrual cycle, hormonal contraceptive use, pregnancy and the menopause, has slowed the pursuit of knowledge in this field of research. In addition, a lack of agreement on the terminology and methodological approaches (i.e., gold-standard techniques) used within this research area has further hindered the ability of researchers to adequately develop evidenced-based guidelines for female exercisers. The purpose of this paper was to highlight the specific considerations needed when employing women (i.e., from athletes to non-athletes) as participants in sport and exercise science-based research. These considerations relate to participant selection criteria and adaptations for experimental design and address the diversity and complexities associated with female reproductive endocrinology across the lifespan. This statement intends to promote an increase in the inclusion of women as participants in studies related to sport and exercise science and an enhanced execution of these studies resulting in more high-quality female-specific data.
Collapse
|
40
|
Do Sex Differences in Physiology Confer a Female Advantage in Ultra-Endurance Sport? Sports Med 2021; 51:895-915. [PMID: 33502701 DOI: 10.1007/s40279-020-01417-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 01/09/2023]
Abstract
Ultra-endurance has been defined as any exercise bout that exceeds 6 h. A number of exceptional, record-breaking performances by female athletes in ultra-endurance sport have roused speculation that they might be predisposed to success in such events. Indeed, while the male-to-female performance gap in traditional endurance sport (e.g., marathon) remains at ~ 10%, the disparity in ultra-endurance competition has been reported as low as 4% despite the markedly lower number of female participants. Moreover, females generally outperform males in extreme-distance swimming. The issue is complex, however, with many sports-specific considerations and caveats. This review summarizes the sex-based differences in physiological functions and draws attention to those which likely determine success in extreme exercise endeavors. The aim is to provide a balanced discussion of the female versus male predisposition to ultra-endurance sport. Herein, we discuss sex-based differences in muscle morphology and fatigability, respiratory-neuromechanical function, substrate utilization, oxygen utilization, gastrointestinal structure and function, and hormonal control. The literature indicates that while females exhibit numerous phenotypes that would be expected to confer an advantage in ultra-endurance competition (e.g., greater fatigue resistance, greater substrate efficiency, and lower energetic demands), they also exhibit several characteristics that unequivocally impinge on performance (e.g., lower O2-carrying capacity, increased prevalence of GI distress, and sex-hormone effects on cellular function/injury risk). Crucially, the advantageous traits may only manifest as ergogenic in the extreme endurance events which, paradoxically, are those that females less often contest. The title question should be revisited in the coming years, when/if the number of female participants increases.
Collapse
|
41
|
Fontaine R, Royan MR, von Krogh K, Weltzien FA, Baker DM. Direct and Indirect Effects of Sex Steroids on Gonadotrope Cell Plasticity in the Teleost Fish Pituitary. Front Endocrinol (Lausanne) 2020; 11:605068. [PMID: 33365013 PMCID: PMC7750530 DOI: 10.3389/fendo.2020.605068] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/12/2020] [Indexed: 12/26/2022] Open
Abstract
The pituitary gland controls many important physiological processes in vertebrates, including growth, homeostasis, and reproduction. As in mammals, the teleost pituitary exhibits a high degree of plasticity. This plasticity permits changes in hormone production and secretion necessary to meet the fluctuating demands over the life of an animal. Pituitary plasticity is achieved at both cellular and population levels. At the cellular level, hormone synthesis and release can be regulated via changes in cell composition to modulate both sensitivity and response to different signals. At the cell population level, the number of cells producing a given hormone can change due to proliferation, differentiation of progenitor cells, or transdifferentiation of specific cell types. Gonadotropes, which play an important role in the control of reproduction, have been intensively investigated during the last decades and found to display plasticity. To ensure appropriate endocrine function, gonadotropes rely on external and internal signals integrated at the brain level or by the gonadotropes themselves. One important group of internal signals is the sex steroids, produced mainly by the gonadal steroidogenic cells. Sex steroids have been shown to exert complex effects on the teleost pituitary, with differential effects depending on the species investigated, physiological status or sex of the animal, and dose or method of administration. This review summarizes current knowledge of the effects of sex steroids (androgens and estrogens) on gonadotrope cell plasticity in teleost anterior pituitary, discriminating direct from indirect effects.
Collapse
Affiliation(s)
- Romain Fontaine
- Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Muhammad Rahmad Royan
- Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Kristine von Krogh
- Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Finn-Arne Weltzien
- Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Dianne M. Baker
- Department of Biological Sciences, University of Mary Washington, Fredericksburg, VA, United States
| |
Collapse
|
42
|
Schuler G, Feller S, Schwandt HJ. [Determination of sex steroids in shed skin of the beaded lizard Heloderma suspectum ("Gila Monster")]. TIERAERZTLICHE PRAXIS AUSGABE KLEINTIERE HEIMTIERE 2020; 48:410-419. [PMID: 33276391 DOI: 10.1055/a-1274-9146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Measurement of steroid hormones in skin appendages such as mammalian hair or claws and in avian feathers represents a recognized non-invasive method for the determination of these parameters. The aim of this pilot study in the Gila Monster was to investigate whether the measurement of sex steroids in shed skin may be employed for the monitoring of endocrine gonadal function or sex determination in reptiles. MATERIAL AND METHODS Shed skins were available from 11 female and 7 male adult and sexually mature animals. Large pieces of skin were initially cut into smaller pieces with scissors. The resultant dermal fragments were finely ground under liquid nitrogen and finally extracted with organic solvents. The following parameters were determined radioimmunologically in the dried and re-dissolved extracts: progesterone (P4), estradiol-17β (E2), testosterone (T), free total estrogens (fGÖ) and free plus conjugated total estrogens (fkGÖ). RESULTS For P4 (p = 0.0052) and E2 (p = 0.0079) significant sex differences were found with higher concentrations in females compared to males. Unexpectedly, the measured values for T were also significantly higher in females (p = 0.0232) than in males, with the concentrations overall only slightly above the detection limit. Compared to fGÖ, the concentrations of fkGÖ were only slightly higher, with no significant differences between both sexes. CONCLUSION AND CLINICAL RELEVANCE In this pilot study, the methods employed did not allow for reliable sex determination in individual animals, neither alone nor in combination, due to an overlap between the sexes. In principle, however, the measurement of sex steroids in shed skins could represent a useful method for non-invasive sex determination or endocrine gonadal function assessment in certain reptile species.
Collapse
Affiliation(s)
- Gerhard Schuler
- Klinik für Geburtshilfe, Gynäkologie und Andrologie der Groß- und Kleintiere mit Tierärztlicher Ambulanz, Justus-Liebig-Universität Gießen
| | | | | |
Collapse
|
43
|
Rattan S, Flaws JA. The epigenetic impacts of endocrine disruptors on female reproduction across generations†. Biol Reprod 2020; 101:635-644. [PMID: 31077281 DOI: 10.1093/biolre/ioz081] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/18/2019] [Accepted: 05/09/2019] [Indexed: 02/06/2023] Open
Abstract
Humans and animals are repeatedly exposed to endocrine disruptors, many of which are ubiquitous in the environment. Endocrine disruptors interfere with hormone action; thus, causing non-monotonic dose responses that are atypical of standard toxicant exposures. The female reproductive system is particularly susceptible to the effects of endocrine disruptors. Likewise, exposures to endocrine disruptors during developmental periods are particularly concerning because programming during development can be adversely impacted by hormone level changes. Subsequently, developing reproductive tissues can be predisposed to diseases in adulthood and these diseases can be passed down to future generations. The mechanisms of action by which endocrine disruptors cause disease transmission to future generations are thought to include epigenetic modifications. This review highlights the effects of endocrine disruptors on the female reproductive system, with an emphasis on the multi- and transgenerational epigenetic effects of these exposures.
Collapse
Affiliation(s)
- Saniya Rattan
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Jodi A Flaws
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Illinois, USA
| |
Collapse
|
44
|
Hlisníková H, Petrovičová I, Kolena B, Šidlovská M, Sirotkin A. Effects and Mechanisms of Phthalates' Action on Reproductive Processes and Reproductive Health: A Literature Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E6811. [PMID: 32961939 PMCID: PMC7559247 DOI: 10.3390/ijerph17186811] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022]
Abstract
The production of plastic products, which requires phthalate plasticizers, has resulted in the problems for human health, especially that of reproductive health. Phthalate exposure can induce reproductive disorders at various regulatory levels. The aim of this review was to compile the evidence concerning the association between phthalates and reproductive diseases, phthalates-induced reproductive disorders, and their possible endocrine and intracellular mechanisms. Phthalates may induce alterations in puberty, the development of testicular dysgenesis syndrome, cancer, and fertility disorders in both males and females. At the hormonal level, phthalates can modify the release of hypothalamic, pituitary, and peripheral hormones. At the intracellular level, phthalates can interfere with nuclear receptors, membrane receptors, intracellular signaling pathways, and modulate gene expression associated with reproduction. To understand and to treat the adverse effects of phthalates on human health, it is essential to expand the current knowledge concerning their mechanism of action in the organism.
Collapse
Affiliation(s)
- Henrieta Hlisníková
- Department of Zoology and Anthropology, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia; (I.P.); (B.K.); (M.Š.); (A.S.)
| | | | | | | | | |
Collapse
|
45
|
de Almeida MM, Dias-Rocha CP, Reis-Gomes CF, Wang H, Cordeiro A, Pazos-Moura CC, Joss-Moore L, Trevenzoli IH. Maternal high-fat diet up-regulates type-1 cannabinoid receptor with estrogen signaling changes in a sex- and depot- specific manner in white adipose tissue of adult rat offspring. Eur J Nutr 2020; 60:1313-1326. [PMID: 32671459 DOI: 10.1007/s00394-020-02318-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 06/25/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Obesity and high-fat (HF) diet are associated with over activation of the endocannabinoid system (ECS). We have demonstrated that maternal HF diet induces early obesity and modulates cannabinoid signaling in visceral (VIS) and subcutaneous (SUB) white adipose tissue (WAT) in weanling rat offspring. We hypothesized that perinatal maternal HF diet would program the expression of ECS in adipose tissue in a long-term way in parallel to alterations in epigenetic markers and sex hormone signaling. METHODS Progenitor female rats received control diet (C, 9% fat) or isocaloric high-fat diet (HF, 28% fat) for 8 weeks before mating, gestation, and lactation. All pups were weaned to C diet and they were euthanized at 180 days old. RESULTS Maternal HF diet induced overweight and increased SUB WAT mass of male and female adult offspring. Maternal HF diet induced hypertrophy of VIS and SUB adipocytes only in female offspring associated with increased type 1 cannabinoid receptor protein (CB1) and mRNA (Cnr1) levels. These changes were associated with increased estrogen receptor α binding to Cnr1 promoter in SUB WAT of adult female offspring, which may contribute to higher expression of Cnr1. CONCLUSION Increased CB1 signaling in adipose tissue might contribute to higher adiposity programmed by maternal HF diet because endocannabinoids stimulate the accumulation of fat in the adipose tissue. Our findings provide molecular insights into sex-specific targets for anti-obesity therapies based on the endocannabinoid system.
Collapse
Affiliation(s)
- Mariana Macedo de Almeida
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil. .,Laboratory of Molecular Endocrinology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro, 21941-902, Brazil.
| | - Camilla P Dias-Rocha
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Clara F Reis-Gomes
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Haimei Wang
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Aline Cordeiro
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Carmen C Pazos-Moura
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Lisa Joss-Moore
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Isis H Trevenzoli
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
46
|
Effects of letrozole administration on growth and reproductive performance in Markhoz goat bucklings. Theriogenology 2020; 147:183-191. [PMID: 31785859 DOI: 10.1016/j.theriogenology.2019.11.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 08/14/2019] [Accepted: 11/17/2019] [Indexed: 11/22/2022]
Abstract
This study evaluated the growth performance, testicular and semen characteristics, and hormonal profile of Markhoz (Iranian Angora) bucklings injected with letrozole (LTZ). Twenty-eight 4-4.5 month old bucks were randomly assigned into four groups and received either 0.25 mg/kg body weight (BW) LTZ subcutaneously (sc LTZ) or intramuscularly (im LTZ), and also sc (sc CONT) or im (im CONT) controls every week for 3 months. The study was performed at the beginning of the breeding season in Sanandaj Animal Husbandry Research Station (46.99 °E, 35.31 °N). The results showed that LTZ causes increased final body weight (25.78 ± 1.61 kg), higher average daily gain (104 ± 0.03 g/days), and decreased feed conversion ratio (7.81 ± 2.57) (P < 0.05). The pre-slaughter, hot, and cold carcass weights (27.56 ± 2.40, 11.45 ± 1.07 and 11.11 ± 1.05 kg, respectively) were (P < 0.05) heavier in LTZ groups while other carcass characteristics did not differ between groups. No differences occurred between the groups in biochemical parameters, except high-density lipoprotein levels (35.47 ± 2.43 mg/dL) which was higher in LTZ treatments (P < 0.05). LTZ-treated bucks had larger scrotal circumference (20.12 ± 5.75 cm), higher relative testicular weight (560.91 ± 78.59 mg/100 g BW) and volume (175.5 ± 29.71 cm3), greater diameter of seminiferous tubules (224.5 ± 5.21 μm), and number of Sertoli cells (8.39 ± 0.77) (P < 0.05). Semen volume (0.74 ± 0.16 mL), sperm concentration (2.64 ± 0.19 × 10-9/mL), total sperm per ejaculate (1.95 ± 0.49 × 10-9), and semen index (1248 ± 323) increased (P < 0.05) by LTZ treatments, while semen pH (6.77), motility (80.91%), progressive motility (76.75%), viability (83.35%), abnormality (13.70%), acrosome integrity (78.06%), and membrane integrity (80.05%) of sperm remained unaffected. Intratesticular and serum testosterone (T) levels (7.97 ± 0.89 ng/mg protein and 2.47 ± 0.59 ng/mL, respectively), serum luteinizing hormone (LH), growth hormone (GH) levels (1.71 ± 0.24 and 3.62 ± 0.33 ng/mL, respectively) of LTZ groups were elevated, whereas intratesticular and serum estradiol (E2) levels (84.14 ± 8.15 pg/mg protein and 32.33 ± 2.16 pg/mL, respectively) decreased (P < 0.05). No differences were recorded between the sc and im routes of LTZ administration in the measured parameters. To conclude, we have found that LTZ treatment improves growth and reproductive functions of goat bucklings associated with increased serum LH and GH, elevated T and reduced E2 levels in both serum and testis.
Collapse
|
47
|
Gurvich C, Warren AM, Worsley R, Hudaib AR, Thomas N, Kulkarni J. Effects of Oral Contraceptive Androgenicity on Visuospatial and Social-Emotional Cognition: A Prospective Observational Trial. Brain Sci 2020; 10:brainsci10040194. [PMID: 32218215 PMCID: PMC7226060 DOI: 10.3390/brainsci10040194] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/14/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022] Open
Abstract
Oral contraceptives (OCs) containing estrogen and progesterone analogues are widely used amongst reproductive-aged women, but their neurocognitive impact is poorly understood. Preliminary studies suggest that OCs improve verbal memory and that OCs with greater androgenic activity may improve visuospatial ability. We sought to explore the cognitive impact of OCs by assessing performance of OC users at different stages of the OC cycle, and comparing this performance between users of different OC formulations according to known androgenic activity. We conducted a prospective, observational trial of OC users, evaluating cognitive performance with CogState software on two occasions: days 7-10 of active hormonal pill phase, and days 3-5 of the inactive pill phase (coinciding with the withdrawal bleed resembling menstruation). Thirty-five OC users (18 taking androgenic formulations, 17 taking anti-androgenic) were assessed. Analysis by androgenic activity showed superior performance by users of androgenic OCs, as compared to anti-androgenic OCs, in visuospatial ability and facial affect discrimination tasks. A growing understanding of cognitive effects of OC progestin androgenicity may have implications in choice of OC formulation for individuals and in future OC development.
Collapse
|
48
|
Pronsato L, Milanesi L, Vasconsuelo A. Testosterone induces up-regulation of mitochondrial gene expression in murine C2C12 skeletal muscle cells accompanied by an increase of nuclear respiratory factor-1 and its downstream effectors. Mol Cell Endocrinol 2020; 500:110631. [PMID: 31676390 DOI: 10.1016/j.mce.2019.110631] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 10/25/2019] [Accepted: 10/25/2019] [Indexed: 01/03/2023]
Abstract
The reduction in muscle mass and strength with age, sarcopenia, is a prevalent condition among the elderly, linked to skeletal muscle dysfunction and cell apoptosis. We demonstrated that testosterone protects against H2O2-induced apoptosis in C2C12 muscle cells. Here, we analyzed the effect of testosterone on mitochondrial gene expression in C2C12 skeletal muscle cells. We found that testosterone increases mRNA expression of genes encoded by mitochondrial DNA, such as NADPH dehydrogenase subunit 1 (ND1), subunit 4 (ND4), cytochrome b (CytB), cytochrome c oxidase subunit 1 (Cox1) and subunit 2 (Cox2) in C2C12. Additionally, the hormone induced the expression of the nuclear respiratory factors 1 and 2 (Nrf-1 and Nrf-2), the mitochondrial transcription factors A (Tfam) and B2 (TFB2M), and the optic atrophy 1 (OPA1). The simultaneous treatment with testosterone and the androgen receptor antagonist, Flutamide, reduced these effects. H2O2-oxidative stress induced treatment, significantly decreased mitochondrial gene expression. Computational analysis revealed that mitochondrial DNA contains specific sequences, which the androgen receptor could recognize and bind, probably taking place a direct regulation of mitochondrial transcription by the receptor. These findings indicate that androgen plays an important role in the regulation of mitochondrial transcription and biogenesis in skeletal muscle.
Collapse
Affiliation(s)
- Lucía Pronsato
- Instituto de Investigaciones Biológicas y Biomédicas del Sur (INBIOSUR-CONICET), 8000, Bahía Blanca, Argentina.
| | - Lorena Milanesi
- Instituto de Investigaciones Biológicas y Biomédicas del Sur (INBIOSUR-CONICET), 8000, Bahía Blanca, Argentina.
| | - Andrea Vasconsuelo
- Instituto de Investigaciones Biológicas y Biomédicas del Sur (INBIOSUR-CONICET), 8000, Bahía Blanca, Argentina
| |
Collapse
|
49
|
Koundourakis NE, Margioris AN. The complex and bidirectional interaction between sex hormones and exercise performance in team sports with emphasis on soccer. Hormones (Athens) 2019; 18:151-172. [PMID: 31256350 DOI: 10.1007/s42000-019-00115-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 01/07/2019] [Indexed: 10/26/2022]
Abstract
A constant topic reported in the lay press is the effect of sex hormones on athletic performance and their abuse by athletes in their effort to enhance their performance or to either boost or sidestep their hard, protracted, and demanding training regimens. However, an issue that it is almost never mentioned is that the athletic training itself affects the endogenous production of androgens and estrogens, while also being affected by them. Among sports, soccer is a particularly demanding activity, soccer players needing to possess high levels of endurance, strength, and both aerobic and anaerobic capacity, with the very great physiological, metabolic, physical, and psychological exertion required of the players being both influenced by sex steroids and, reciprocally, affecting sex steroid levels. This review focuses on the currently available knowledge regarding the complex relationship between athletic training and competition and sex steroid hormone adaptation to the demands of the exercise effort. In the first part of the review, we will examine the effects of endogenous testosterone, estrogen, and adrenal androgens on athletic performance both during training and in competition. In the second part, we will explore the reciprocal effects of exercise on the endogenous sex hormones while briefly discussing the recent data on anabolic androgenic steroid abuse.
Collapse
Affiliation(s)
- Nikolaos E Koundourakis
- Lab of Clinical Chemistry-Biochemistry, Department of Laboratory Medicine, School of Medicine, University of Crete, Heraklion, Crete, Greece.
| | - Andrew N Margioris
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Crete, Greece
| |
Collapse
|
50
|
Flöter VL, Bauersachs S, Fürst RW, Krebs S, Blum H, Reichenbach M, Ulbrich SE. Exposure of pregnant sows to low doses of estradiol-17β impacts on the transcriptome of the endometrium and the female preimplantation embryos†. Biol Reprod 2019; 100:624-640. [PMID: 30260370 DOI: 10.1093/biolre/ioy206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 07/30/2018] [Accepted: 09/25/2018] [Indexed: 11/14/2022] Open
Abstract
Maternal exposure to estrogens can induce long-term adverse effects in the offspring. The epigenetic programming may start as early as the period of preimplantation development. We analyzed the effects of gestational estradiol-17β (E2) exposure with two distinct low doses, corresponding to the acceptable daily intake "ADI" and close to the no-observed-effect level "NOEL", and a high dose (0.05, 10, and 1000 μg E2/kg body weight daily, respectively). The E2 doses were orally applied to sows from insemination until sampling at day 10 of pregnancy and compared to carrier-treated controls leading to a significant increase in E2 in plasma, bile and selected somatic tissues including the endometrium in the high-dose group. Conjugated and unconjugated E2 metabolites were as well elevated in the NOEL group. Although RNA-sequencing revealed a dose-dependent effect of 14, 17, and 27 differentially expressed genes (DEG) in the endometrium, single embryos were much more affected with 982 DEG in female blastocysts of the high-dose group, while none were present in the corresponding male embryos. Moreover, the NOEL treatment caused 62 and 3 DEG in female and male embryos, respectively. Thus, we detected a perturbed sex-specific gene expression profile leading to a leveling of the transcriptome profiles of female and male embryos. The preimplantation period therefore demonstrates a vulnerable time window for estrogen exposure, potentially constituting the cause for lasting consequences. The molecular fingerprint of low-dose estrogen exposure on developing embryos warrants a careful revisit of effect level thresholds.
Collapse
Affiliation(s)
- Veronika L Flöter
- ETH Zurich, Animal Physiology, Institute of Agricultural Sciences, Zurich, Switzerland.,Physiology Weihenstephan, Technische Universität München, Freising, Germany
| | - Stefan Bauersachs
- ETH Zurich, Animal Physiology, Institute of Agricultural Sciences, Zurich, Switzerland
| | - Rainer W Fürst
- Physiology Weihenstephan, Technische Universität München, Freising, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center of the Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center of the Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Myriam Reichenbach
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center of the Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Susanne E Ulbrich
- ETH Zurich, Animal Physiology, Institute of Agricultural Sciences, Zurich, Switzerland.,Physiology Weihenstephan, Technische Universität München, Freising, Germany
| |
Collapse
|