1
|
Chauhan V, Rai U, Tripathy M, Kumar S. Neuropeptide Y at the crossroads of male reproductive functions in a seasonally breeding reptile, Hemidactylus flaviviridis. Comp Biochem Physiol A Mol Integr Physiol 2025; 303:111826. [PMID: 39971152 DOI: 10.1016/j.cbpa.2025.111826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/15/2025] [Accepted: 02/15/2025] [Indexed: 02/21/2025]
Abstract
Neuropeptide Y (NPY) is known to be a key regulator of reproductive functions across vertebrates. Its role in the modulation of gonadotropin releasing hormone (GnRH) has been extensively explored in mammals and fishes. However, no such report exists in aves, amphibians, or reptiles. Hence, the present study aimed to develop an insight into reproductive phase-dependent expression and role of NPY in the diencephalon region of male wall lizards. Expression of ligand, npy, and its receptor, npyr varied across the annual reproductive cycle of Hemidactylus flaviviridis with the highest expression of both observed during recrudescence. Further, the diencephalon region of recrudescent wall lizards treated with NPY demonstrated an increased mRNA level of gnrh and its receptor, gnrhr. In addition, the current study also elucidates hormonal regulation of diencephalonic npy and npyr wherein neuropeptides like kisspeptin and substance P, the gonadotropin, FSH, as well as sex steroids, dihydrotestosterone (DHT) and 17β-estradiol (E2) inhibited npy and npyr expression in the diencephalon region of wall lizards. With regard to adipokines, leptin stimulated while nesfatin-1 inhibited diencephalonic npy and npyr expression. In conclusion, the current study is the first to present a comprehensive picture of reproductive phase-specific expression pattern, role, and hormonal regulation of neuropeptide Y in the diencephalon region of the male wall lizard, H. flaviviridis.
Collapse
Affiliation(s)
- Vishesh Chauhan
- Zakir Husain Delhi College, University of Delhi, Delhi 110002, India; Department of Zoology, University of Delhi, Delhi 110007, India
| | - Umesh Rai
- University of Jammu, Jammu and Kashmir, 180006, India
| | - Mamta Tripathy
- Department of Zoology, University of Delhi, Delhi 110007, India.
| | - Sunil Kumar
- Zakir Husain Delhi College, University of Delhi, Delhi 110002, India.
| |
Collapse
|
2
|
Mansour MF, Behairy A, Mostafa M, Khamis T, Alsemeh AE, Ahmed NMQ, El-Emam MMA. Quercetin-loaded PEGylated liposomes alleviate testicular dysfunction in alloxan-induced diabetic rats: The role of Kisspeptin/Neurokinin B/Dynorphin pathway. Toxicol Appl Pharmacol 2025; 499:117337. [PMID: 40239742 DOI: 10.1016/j.taap.2025.117337] [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: 12/14/2024] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder that can lead to serious complications, including testicular dysfunction. This dysfunction is considered a significant cause of male infertility. Quercetin (Que), a naturally existing flavonoid with versatile biological functions, has limited water solubility and low bioavailability. The current study was designed to develop a bioavailable formulation of Que. via encapsulating it in PEGylated liposomes (Que-PEG-Lip) and determine whether this formulation is effective in the treatment of alloxan-induced testicular injury via targeting Kisspeptin/Neurokinin B/Dynorphin/steroidogenesis signaling pathway. Thirty-two male Sprague Dawley rats were randomly divided into four groups: Control, alloxan-induced diabetes with testicular dysfunction (ALX), ALX + metformin (MET) and ALX + Que-PEG-Lip. The results showed that treatment of ALX group with Que-PEG-Lip significantly improved the alteration of glycemic index, serum reproductive hormones, testicular antioxidant status, testicular Kiss-1, androgen receptor (AR), and proliferation marker protein (ki67) immunoexpression in compared to ALX group. Moreover, the treatment of ALX group with Que-PEG-Lip regulated the Kisspeptin/Neurokinin B/Dynorphin/steroidogenesis pathway gene expression. Interestingly, the outcomes of the molecular docking analysis revealed a strong agonistic effect of Que. on the kisspeptin, neurokinin, and dynorphin receptors. In conclusion, Que-PEG-Lip mitigated the testicular dysfunction in alloxan-induced diabetic rats via regulation of hypothalamic-pituitary-gonadal axis signaling pathway and alleviation the testicular oxidative stress.
Collapse
Affiliation(s)
- Mohamed Fouad Mansour
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt.
| | - Amany Behairy
- Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Mahmoud Mostafa
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Amira Ebrahim Alsemeh
- Department of Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | | | - Mahran Mohamed Abd El-Emam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
3
|
Dharani V, Nishu S, Hariprasath L. PCOS and genetics: Exploring the heterogeneous role of potential genes in ovarian dysfunction, a hallmark of PCOS - A review. Reprod Biol 2025; 25:101017. [PMID: 40222066 DOI: 10.1016/j.repbio.2025.101017] [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: 09/25/2024] [Revised: 03/08/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025]
Abstract
PCOS is an endocrine disorder that affects women of reproductive age. The root of PCOS is ovarian dysfunction, which presents as hormonal disturbances affecting normal ovarian function to cause the symptoms and complications of the disease. This dysfunction causes symptoms like impaired maturation of follicles and disorders of various origins with multiple treatment regimens that are not always clear. Therefore, the present review mainly concentrates on the genetic level of ovarian dysfunction of PCOS. The articles were identified through a vigorous literature search where search engines such as PubMed, Google Scholar, databases, and Science Direct were used, and the articles published from 2015 to 2025 were referred. We identified that the key genes involved in the ovarian dysfunctions in PCOS include CYP11A1, CYP17A1, CYP19A1, AR, FSHR, LHCGR, AMH, INSR, SHBG, IRS1, GATA4, ADIPOQ, YAP1, TCF7L2, and DENND1A, which play a role in gonadotropin action, steroidogenesis, and folliculogenesis. Furthermore, epigenetic factors and miRNAs miR-93, 222, 155, 146a, 132, 320, 27a, 483, 21, 378, 17-92 Cluster, and 375, 221 are also involved in it. Abnormal expression of these genes is known to play a critical role in the etiology and pathogenesis of PCOS. Present treatment includes the use of oral contraceptives, anti-androgen agents, insulin-sensitizing agents, and ovulation-inducing agents, and future treatment may consist of miRNA therapy, drug repositioning, and genetic markers that might be used for early identification and better management of ovarian dysfunction. Thus, the current review discusses ovarian dysfunction in PCOS, the involvement of potential genes and epigenetic factors, and miRNAs concerning ovulation and its therapeutic implications.
Collapse
Affiliation(s)
- V Dharani
- Department of Biotechnology, School of Life Sciences (Ooty Campus), JSS Academy of Higher Education & Research, Longwood, Ooty, Tamil Nadu 643001, India
| | - S Nishu
- Department of Biotechnology, School of Life Sciences (Ooty Campus), JSS Academy of Higher Education & Research, Longwood, Ooty, Tamil Nadu 643001, India.
| | - L Hariprasath
- Department of Biochemistry, School of Life Sciences (Ooty Campus), JSS Academy of Higher Education & Research, Longwood, Ooty, Tamil Nadu 643001, India
| |
Collapse
|
4
|
Martins FF, Amarante MDSM, Oliveira DS, Vasques-Monteiro IML, Souza-Mello V, Daleprane JB, Camillo CDS. Obesity, White Adipose Tissue, and Adipokines Signaling in Male Reproduction. Mol Nutr Food Res 2025:e70054. [PMID: 40195898 DOI: 10.1002/mnfr.70054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/19/2025] [Accepted: 03/17/2025] [Indexed: 04/09/2025]
Abstract
Currently, obesity is a global pandemic characterized by systemic metabolic complications that negatively impact several organs, including white adipose tissue (WAT) and the tissues of the male reproductive system. Since the discovery of leptin in 1994, WAT has been recognized as a dynamic endocrine organ for secreting a series of molecules with hormonal functions, collectively called adipokines. The link between obesity, WAT, adipokines, and the male reproductive system is direct and little explored. With changes in nutritional status, WAT undergoes morphofunctional changes, and the secretion of adipokines is altered, negatively impacting reproductive mechanisms, including steroidogenesis and spermatogenesis. In this review, we address in an updated way the structural and functional characteristics of WAT as well as the link between obesity and changes in the signaling pathways of the adipokines leptin, adiponectin, resistin, visfatin, apelin, chemerin, omentin-1, vaspin, and asprosin in male reproduction. Understanding the relationship between obesity, these adipokines, and reproductive dysfunction can contribute to new strategies for the treatment of subfertility and male infertility.
Collapse
Affiliation(s)
- Fabiane Ferreira Martins
- Department of Morphology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | | | - Daiana Santana Oliveira
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Isabela Macedo Lopes Vasques-Monteiro
- Department of Basic and Experimental Nutrition, Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Julio Beltrame Daleprane
- Department of Basic and Experimental Nutrition, Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | |
Collapse
|
5
|
Di M, Gong X, Zhu Y, Guo S, Pan Z, Li M, Wu Z, Zhang W, Liu X, Liu Y, Li Y, Li J, Fang F. Active immunization with a novel recombinant GnRH vaccine inhibits reproductive function in male goats. Domest Anim Endocrinol 2025; 91:106908. [PMID: 39708581 DOI: 10.1016/j.domaniend.2024.106908] [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: 08/13/2024] [Revised: 12/14/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
Gonadotropin-releasing hormone (GnRH) vaccines have been widely used to effectively inhibit gonadal development and reproductive function. To improve the immunogenicity of GnRH, we developed and evaluated the effects of GnRH6-kisspeptin-CRM197 immunization on the reproductive function in male goats. Thirty 3-month-old male goats (n = 30) were randomly assigned to control, surgical, and immunized groups. The immunized group received a 2 mL injection of the GnRH6-kisspeptin-CRM197 with a booster administered four weeks later. The control group was administered a white oil adjuvant. Blood samples were collected at regular intervals, and at week 20, the animals were euthanized for tissue collection. Serum antibody titers and testosterone levels were measured using ELISA and CLIA, respectively. Testicular parameters and histology were evaluated. The mRNA levels of reproductive-related genes in the HPG axis were measured using RT-qPCR. The results showed that the immunized goats had significantly increased serum GnRH and kisspeptin antibodies (P < 0.05) but decreased testosterone concentrations (P < 0.05) compared to the control group. Testicular size and histology were significantly affected in the immunized group, with notable reductions in testicular weight and dimensions (P < 0.01), and evidence of vacuolar degeneration and suppressed sperm production. The mRNA levels of FSHβ and LHβ in the pituitary, as well as FSHR, LHR, 3βHSD, and 17βHSD in the testis, were significantly lower in the immunized group compared to controls (P < 0.05). These findings suggest that GnRH6-kisspeptin-CRM197 is a safe antigen and a promising immunocastration vaccine with enhanced efficacy.
Collapse
Affiliation(s)
- Moyan Di
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China
| | - Xinbao Gong
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China
| | - Yanyun Zhu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China
| | - Shibao Guo
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China
| | - Zhihao Pan
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China
| | - Mengxian Li
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China
| | - Zhuoya Wu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China
| | - Wei Zhang
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China
| | - Xuelan Liu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ya Liu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yunsheng Li
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Jian Li
- Biological and Food Engineering College, Fuyang Normal University, 100 Qinghe West Road, Fuyang, Anhui 236037, China
| | - Fugui Fang
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China.
| |
Collapse
|
6
|
Chen D, Fang M, Huang E, Quan H, Zhang L, He Y, Zhou X, Ma B, Yuan X, Li J. DNA Methylation Mediates the Transcription of STAT4 to Regulate KISS1 During Follicular Development. Cells 2025; 14:523. [PMID: 40214477 PMCID: PMC11989168 DOI: 10.3390/cells14070523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Maturation of follicles is the primary condition for the initiation of puberty, and excessive apoptosis of granulosa cells (GCs) will hinder the normal development of follicles in pigs. Signal Transducer and Activator of Transcription 4 (STAT4) plays an important role in cell proliferation and apoptosis. However, the mechanism of DNA methylation regulating STAT4 transcription and affecting follicle development in pigs remains unclear. To resolve this problem, we constructed a STAT4 overexpression vector and interference fragment to explore the effects of STAT4 on GC function and investigate the effects of changes in methylation status of the STAT4 promoter region on cell function and kisspeptin-1 (KISS1) expression, as well as the STAT4 effects on the development of the follicles of pigs and mice in vitro. We found that the expression of STAT4 decreased, while DNA methylation of the STAT4 promoter region increased with the growth of the follicles. After overexpression of STAT4, the apoptosis of GCs was increased but the proliferation, cell cycle and estrogen secretion of GCs were inhibited. When GCs were treated with DNA methyltransferase inhibitor (5-Aza-CdR), the methylation of the STAT4 promoter region decreased, resulting in a significant increase in the expression of STAT4. Consequently, the expression of KISS1 was inhibited. At the same time, the expressions of genes related to cell proliferation, cell cycle and estrogen secretion signaling pathways decreased, while the expressions of genes related to the apoptosis signaling pathway increased. After infection with the STAT4 lentiviral vector (LV-STAT4) in follicles of mice, the expression of STAT4 in ovaries of mice significantly increased, and the expression of KISS1 was significantly decreased. The capillaries on the surface of follicles were constricted, the age of puberty onset in mice was delayed while the levels of GnRH, LH, FSH and E2 in serum were decreased. In conclusion, we found that reduced methylation status of the STAT4 promoter region promoted the transcription of STAT4 and then inhibited the expression of KISS1, as well as promoted the apoptosis of GCs and ultimately inhibited the normal development of follicles in mammals.
Collapse
Affiliation(s)
- Danxia Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (D.C.); (M.F.); (E.H.); (H.Q.); (L.Z.); (Y.H.); (X.Z.)
| | - Ming Fang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (D.C.); (M.F.); (E.H.); (H.Q.); (L.Z.); (Y.H.); (X.Z.)
| | - Enyuan Huang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (D.C.); (M.F.); (E.H.); (H.Q.); (L.Z.); (Y.H.); (X.Z.)
| | - Hongyan Quan
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (D.C.); (M.F.); (E.H.); (H.Q.); (L.Z.); (Y.H.); (X.Z.)
| | - Liuhong Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (D.C.); (M.F.); (E.H.); (H.Q.); (L.Z.); (Y.H.); (X.Z.)
| | - Yingting He
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (D.C.); (M.F.); (E.H.); (H.Q.); (L.Z.); (Y.H.); (X.Z.)
| | - Xiaofeng Zhou
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (D.C.); (M.F.); (E.H.); (H.Q.); (L.Z.); (Y.H.); (X.Z.)
| | - Bin Ma
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia;
| | - Xiaolong Yuan
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (D.C.); (M.F.); (E.H.); (H.Q.); (L.Z.); (Y.H.); (X.Z.)
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia;
- National Center of Technology Innovation for Pigs, Chongqing 402460, China
| | - Jiaqi Li
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (D.C.); (M.F.); (E.H.); (H.Q.); (L.Z.); (Y.H.); (X.Z.)
- National Center of Technology Innovation for Pigs, Chongqing 402460, China
| |
Collapse
|
7
|
Di Rienzi SC, Danhof HA, Forshee MD, Roberts A, Britton RA. Limosilactobacillus reuteri promotes the expression and secretion of enteroendocrine- and enterocyte-derived hormones. FASEB J 2025; 39:e70408. [PMID: 40098558 PMCID: PMC11914943 DOI: 10.1096/fj.202401669r] [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: 07/21/2024] [Revised: 01/20/2025] [Accepted: 02/10/2025] [Indexed: 03/19/2025]
Abstract
Intestinal microbes can beneficially impact host physiology, prompting investigations into the therapeutic usage of such microbes in a range of diseases. For example, human intestinal microbe Limosilactobacillus reuteri strains ATCC PTA 6475 and DSM 17938 are being considered for use for intestinal ailments, including colic, infection, and inflammation, as well as for non-intestinal ailments, including osteoporosis, wound healing, and autism spectrum disorder. While many of their beneficial properties are attributed to suppressing inflammatory responses, we postulated that L. reuteri may also regulate intestinal hormones to affect physiology within and outside of the gut. To determine if L. reuteri secreted factors impact the secretion of enteric hormones, we treated an engineered jejunal organoid line, NGN3-HIO, which can be induced to be enriched in enteroendocrine cells, with L. reuteri 6475 or 17938 conditioned medium and performed transcriptomics. Our data suggest that these L. reuteri strains affect the transcription of many gut hormones, including vasopressin and luteinizing hormone subunit beta, which have not been previously recognized as produced in the gut epithelium. Moreover, we find that these hormones appear to be produced in enterocytes, in contrast to canonical gut hormones produced in enteroendocrine cells. Finally, we show that L. reuteri conditioned media promote the secretion of enteric hormones, including serotonin, GIP, PYY, vasopressin, and luteinizing hormone subunit beta, and identify by metabolomics metabolites potentially mediating these effects on hormones. These results support L. reuteri affecting host physiology through intestinal hormone secretion, thereby expanding our understanding of the mechanistic actions of this microbe.
Collapse
Affiliation(s)
- Sara C. Di Rienzi
- Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTexasUSA
- Alkek Center for Metagenomics and Microbiome ResearchBaylor College of MedicineHoustonTexasUSA
| | - Heather A. Danhof
- Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTexasUSA
- Alkek Center for Metagenomics and Microbiome ResearchBaylor College of MedicineHoustonTexasUSA
| | - Micah D. Forshee
- Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTexasUSA
- Alkek Center for Metagenomics and Microbiome ResearchBaylor College of MedicineHoustonTexasUSA
| | - Ari Roberts
- Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTexasUSA
- Alkek Center for Metagenomics and Microbiome ResearchBaylor College of MedicineHoustonTexasUSA
| | - Robert A. Britton
- Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTexasUSA
- Alkek Center for Metagenomics and Microbiome ResearchBaylor College of MedicineHoustonTexasUSA
| |
Collapse
|
8
|
Shen Y, Fan J, Li C, Wu F, Wu X, Tao L, Yang Q, Shen X. Restorative mechanisms of Shugan Yiyang capsule on male infertility through 'pharmaco-metabo-net' tripartite correlation analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156706. [PMID: 40220430 DOI: 10.1016/j.phymed.2025.156706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 03/21/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Shugan Yiyang capsule (SGYY), a commonly used traditional Chinese medicine formulation, is primarily indicated for the treatment of erectile dysfunction, yet existing studies on the therapeutic effects on male infertility (MI) are insufficient and the specific mechanisms remain poorly understood. Given the close relationship between MI, sperm quality, and erectile function, this study aims to investigate the role of SGYY in the restoration of MI and explore the underlying mechanisms. METHODS The efficacy of SGYY is comprehensively evaluated through pharmacodynamic, metabolomic, and network pharmacology. Sperm parameters, reproductive hormones, sexual behavior, neural enzymes, oxidative stress markers, pro-inflammatory cytokines, and testicular histopathology are measured to reveal the restorative effects of MI. Furthermore, urine and serum metabolomics, along with network pharmacology and surface plasmon resonance, are employed to explore the molecular mechanisms and predict core targets. RESULTS SGYY significantly improved overall health parameters, including body weight, water intake, urine output, food consumption, and spontaneous activity. Specifically, SGYY prominently recovered sexual behavior, ameliorated sperm quality, increased mitochondrial membrane potential, normalized reproductive hormones, upregulated endothelial nitric oxide synthase, attenuated oxidative stress markers, and pro-inflammatory cytokines, and repaired testicular pathological damage. Metabolomic analysis identified 47 candidate biomarkers, among which SGYY significantly modulated 39 potential biomarkers, encompassing 8 main metabolic pathways such as histidine metabolism, cysteine and methionine metabolism, propanoate metabolism, and taurine and hypotaurine metabolism. Additionally, network pharmacology predicted 8 core targets, comprising HSP90AA1, ESR1, MAPK1, CASP3, IL6, TNF, BCL2, and MAPK8. CONCLUSION SGYY improves sperm quality and erectile function by regulating oxidative stress, energy metabolism, and neurological function, thereby exerting a restorative effect on MI, as evidenced by the modulation of 8 main metabolic pathways, 39 potential biomarkers, and 8 core targets. Pharmacodynamic provides foundational validation, metabolomic uncovers intrinsic metabolic changes, and network pharmacology predicts therapeutic targets, with findings from the 'Pharmaco-Metabo-Net' tripartite correlation analysis providing a solid theoretical strategy and scientific evidence to support the clinical application of SGYY in restoring MI.
Collapse
Affiliation(s)
- Ying Shen
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 561113, China; High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China; National Engineering Research Center of Miao's Medicines, Guizhou Yibai Pharmaceutical Co., Ltd., Guiyang 550008, China
| | - Jian Fan
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 561113, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Chunmei Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 561113, China; High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Fuli Wu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 561113, China; High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Xiangli Wu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 561113, China; High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Ling Tao
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 561113, China; High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China.
| | - Qingbo Yang
- High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China; National Engineering Research Center of Miao's Medicines, Guizhou Yibai Pharmaceutical Co., Ltd., Guiyang 550008, China.
| | - Xiangchun Shen
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 561113, China; High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China.
| |
Collapse
|
9
|
Paganos P, Wolff C, Voronov D, Swartz SZ. Molecular evidence for pre-chordate origins of ovarian cell types and neuroendocrine control of reproduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.644836. [PMID: 40196654 PMCID: PMC11974710 DOI: 10.1101/2025.03.24.644836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Sexual reproduction in animals requires the development of oocytes, or egg cells. This process, termed oogenesis, requires complex interactions amongst germline and somatic cell types in the ovary. How did these cell types and their signaling interactions evolve? Here we use the sea star Patiria miniata as a non-chordate deuterostome representative to define the ovarian cell type toolkit in echinoderms. Sea stars continuously produce millions of new oocytes throughout their lifespan, making them a practical system to understand the mechanisms that drive oogenesis from a biomedical and evolutionary perspective. We performed scRNA-seq combined with high-resolution 3D-imaging to reveal the ovarian cell types and their spatial organization. Our data support the presence of actively dividing oogonial stem cells and granulosa-like and theca-like cells, which display similarities and possible homology with their mammalian counterparts. Lastly, our data support the existence of an endocrine signaling system between oogonial stem cells and intrinsic ovarian neurons with striking similarities to the vertebrate hypothalamic-pituitary-gonadal axis. Overall, this study provides molecular evidence supporting the possible pre-chordate origins of conserved ovarian cell types, and the presence of an intrinsic neuroendocrine system which potentially controls oogenesis and predates the formation of the hypothalamic-pituitary-gonadal axis in vertebrates.
Collapse
Affiliation(s)
- Periklis Paganos
- Marine Biological Laboratory, 7 MBL Street, Woods Hole, Massachusetts, 02543, United States of America
| | - Carsten Wolff
- Marine Biological Laboratory, 7 MBL Street, Woods Hole, Massachusetts, 02543, United States of America
| | - Danila Voronov
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Straße 2, 24306 Plön, Germany
| | - S. Zachary Swartz
- Marine Biological Laboratory, 7 MBL Street, Woods Hole, Massachusetts, 02543, United States of America
| |
Collapse
|
10
|
Feng X, Xu M, Liu Y, Wang X, Duan Y, Zheng X, Yin W, Cai Y, Zhang W, Jiang Q, Pang J, Li J. The sperm quality in DIO male mice is linked to the NF-κB signaling and Ppp2ca expression in the hypothalamus. iScience 2025; 28:112110. [PMID: 40160428 PMCID: PMC11951025 DOI: 10.1016/j.isci.2025.112110] [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: 07/29/2024] [Revised: 11/24/2024] [Accepted: 02/21/2025] [Indexed: 04/02/2025] Open
Abstract
Recent studies show obesity correlated with reduced sperm quality in males, but the mechanism is unclear. In this study, diet-induced obese (DIO) male mice exhibited disrupted luteinizing hormone (LH) pulse release due to altered function of the hypothalamic-pituitary-gonadal (HPG) axis. This alteration was caused by activation of nuclear factor kappa B (NF-κB) signaling in the hypothalamus, which led to decreased sperm quality. RNA sequencing (RNA-seq) analysis of the hypothalamic arcuate nucleus (ARC) revealed a signaling network involving protein phosphatase 2 catalytic subunit alpha (Ppp2ca). This network disrupted LH pulse secretion by inhibiting Akt kinase (AKT) and cAMP responsive element-binding protein 1 (CREB1) activities, thereby reducing KiSS-1 metastasis-suppressor (Kiss1) expression. Furthermore, overexpression of the Ppp2ca gene in the ARC led to disrupted LH patterns and reduced sperm quality. These findings offer new insights into the molecular mechanisms underlying sperm quality decline in DIO male mice.
Collapse
Affiliation(s)
- Xu Feng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Maoxing Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ying Liu
- Clinical Center of Reproductive Medicine, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Xiaoyu Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yiman Duan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaoyan Zheng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Wen Yin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yafei Cai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Wei Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jing Pang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Juxue Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, China
| |
Collapse
|
11
|
Kawada T, Aoyama M, Matsubara S, Osugi T, Sakai T, Kirimoto S, Nakaoka S, Sugiura Y, Yasuda K, Satake H. Ovarian tachykinin signaling system induces the growth of secondary follicles during the gonadotropin-independent process. J Biol Chem 2025; 301:108375. [PMID: 40043951 PMCID: PMC11999619 DOI: 10.1016/j.jbc.2025.108375] [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: 04/18/2024] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Mammalian follicle growth development is mainly regulated by the hypothalamus-pituitary-gonadal axis after puberty. Although pituitary hormones, gonadotropins, are involved in hypothalamus-pituitary-gonadal axis signaling, they are not responsible for the growth of early stage follicles, namely, primordial follicles, primary follicles, and secondary follicles, in both sexually immature and mature individuals. Unlike those of gonadotropin-dependent follicle growth, the specific regulatory factors of gonadotropin-independent follicle growth have yet to be identified. Here, we identified tachykinins (TKs) as inducers of gonadotropin-independent secondary follicle growth. TKs play various roles as neuropeptides or hormones in a wide variety of biological events both in the central nervous system and in peripheral tissues, but a direct effect of TKs on ovarian follicles has yet to investigated. Follicle development was suppressed in sexually immature 3-week-old KO mice of Tac1 gene encoding TKs (substance P and neurokinin A), which is independent of gonadotropins. TKs and their receptors are specifically localized to granulosa cells in mouse secondary follicles. Furthermore, TKs upregulate the prostaglandin (PG) synthase cyclooxygenase 2 via the Janus kinase 1-signal transducers and activators of transcription protein 3 signaling cascade. We also demonstrated that PGE2 and PGF2α are major PGs in the immature ovary, and the secondary follicle growth was enhanced by interaction between PGE2-PGF2α and their receptors, PGE2 receptor localized in the oocyte membrane and PGF2α receptor localized in the oocyte membrane, granulosa cells, and theca cells. Consequently, this study paves the way for exploring gonadotropin-independent early stage follicle growth systems and relevant dysfunctions, including pediatric endocrinological diseases.
Collapse
Affiliation(s)
- Tsuyoshi Kawada
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Masato Aoyama
- Department of Chemistry, Biology, and Environmental Science, Faculty of Science, Nara Women's University, Nara, Japan
| | - Shin Matsubara
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Tomohiro Osugi
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Tsubasa Sakai
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Shinji Kirimoto
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Satsuki Nakaoka
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Yuki Sugiura
- Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Keiko Yasuda
- Department of Chemistry, Biology, and Environmental Science, Faculty of Science, Nara Women's University, Nara, Japan
| | - Honoo Satake
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan.
| |
Collapse
|
12
|
Wang X, Liu Q, Li J. Dynamic Transcriptional Regulation of the Hypothalamic-Pituitary-Testis Axis in the Seasonally Breeding Teleost Sebastes schlegelii. Int J Mol Sci 2025; 26:2048. [PMID: 40076678 PMCID: PMC11899799 DOI: 10.3390/ijms26052048] [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: 01/08/2025] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Spermatogenesis, the process of male germ cell development, is tightly regulated by the hypothalamic-pituitary-testis (HPT) axis in seasonally breeding teleosts. Despite its importance, our understanding of how the brain and male germ cells coordinate key transitions-such as testis initiation and maturation-remains limited, particularly in species with distinct seasonal reproductive cycles. Black rockfish (Sebastes schlegelii), a marine viviparous teleost, exhibits a prolonged testis quiescent phase lasting three-quarters of the year, with testis initiation occurring in September and maturation concluding in November and December. The mechanisms underlying these transitions are poorly characterized, leaving a critical gap in our knowledge of seasonal spermatogenesis and its regulation. Addressing this gap is crucial for advancing artificial breeding technologies, which could significantly benefit the aquaculture industry. RNA-seq was used to explore the gene regulatory networks involved in testis initiation in S. schlegelii. Transcriptomic analyses of brain and testis were conducted across key developmental phases. In the brain, upregulated genes were notably involved in neuroactive ligand-receptor interactions, whereas in the testis, differentially expressed genes were linked to cell cycle processes and ATP-dependent chromatin remodeling. Our findings reveal the molecular mechanisms underlying testis initiation in S. schlegelii, providing omics evidence for the role of the HPT axis in regulating this process. By elucidating the gene regulatory networks of the brain and testis during critical transitions, this study advances our understanding of spermatogenesis in seasonally breeding teleosts. These insights pave the way for developing year-round artificial breeding technologies, contributing to the sustainable management of commercially valuable fish species.
Collapse
Affiliation(s)
- Xueying Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266000, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Qingdao 266000, China
| | - Qinghua Liu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266000, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Qingdao 266000, China
| | - Jun Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266000, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Qingdao 266000, China
| |
Collapse
|
13
|
Denizli I, Monteiro A, Elmer KR, Stevenson TJ. Photoperiod-driven testicular DNA methylation in gonadotropin and sex steroid receptor promoters in Siberian hamsters. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2025:10.1007/s00359-025-01733-w. [PMID: 39954063 DOI: 10.1007/s00359-025-01733-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/17/2025]
Abstract
Seasonal cycles in breeding, often orchestrated by annual changes in photoperiod, are common in nature. Here, we studied how change in photoperiod affects DNA methylation in the testes of a highly seasonal breeder: the Siberian hamster (Phodopus sungorus). We hypothesized that DNA methylation in promoter regions associated with key reproductive genes such as follicle-stimulating hormone receptor in the testes is linked to breeding and non-breeding states. Using Oxford Nanopore sequencing, we identified more than 10 million (10,151,742) differentially methylated cytosine-guanine (CpG) sites in the genome between breeding long photoperiod and non-breeding short photoperiod conditions. ShinyGo enrichment analyses identified biological pathways consisting of reproductive system, hormone-mediated signalling and gonad development. We found that short photoperiod induced DNA methylation in the promoter regions for androgen receptor (Ar), estrogen receptors (Esr1, Esr2), kisspeptin1 receptor (kiss1r) and follicle-stimulating hormone receptor (Fshr). Long photoperiods were observed to have higher DNA methylation in promoters for basic helix-loop-helix ARNT-like 1 (Bmal1), progesterone receptor (Pgr) and thyroid-stimulating hormone receptor (Tshr). Our findings provide insights into the epigenetic mechanisms underlying seasonal adaptations in timing reproduction in Siberian hamsters and could be informative for understanding male fertility and reproductive disorders in mammals.
Collapse
Affiliation(s)
- Irem Denizli
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK.
| | - Ana Monteiro
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Kathryn R Elmer
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Tyler J Stevenson
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| |
Collapse
|
14
|
Wang Y, Wang Y, Chen Y, Lu W, Tong X, Li J, Gao W, Huang R, Hu W, Feng Y. Distinct mechanisms of electroacupuncture and manual acupuncture in modulating hypothalamic GnRH-tanycyte unit function of polycystic ovary syndrome. Chin Med 2025; 20:18. [PMID: 39910658 DOI: 10.1186/s13020-025-01068-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a complex neuroendocrine disorder characterized by dysregulation of the hypothalamus. Both electroacupuncture (EA) and manual acupuncture (MA) have demonstrated therapeutic efficacy in the treatment of PCOS through improvements in hypothalamic function. However, the underlying mechanisms remain poorly understood. Gonadotropin-releasing hormone (GnRH) neurons are pivotal in regulating hypothalamic endocrine function, whereas tanycyte, a specialized glial cell type, potentially contribute to this process. METHODS A dihydrotestosterone (DHT)-induced PCOS-like mouse model was used to investigate the effects of acupuncture. Tissue clearing and three-dimensional (3D) imaging were employed to visualize the hypothalamic GnRH neuronal network and assess postacupuncture modifications. Transcriptome sequencing was performed to identify changes in the gene profiles associated with EA and MA. Rax-CreERT2 transgenic mice were utilized to investigate the molecular targets of EA in tanycytes. RESULTS EA significantly alleviated neuroendocrine dysfunction in PCOS-like mice by restoring the density and coverage of GnRH axonal projections. MA displayed similar therapeutic effects but had less pronounced effects on GnRH axons. Transcriptome analysis revealed distinct mechanisms for these two approaches: EA primarily regulates neuroglial plasticity, whereas MA predominantly targets neurotransmitter regulation. Both EA and MA share a common therapeutic target in the integrin family. Functional studies in Rax-CreERT2 transgenic mice confirmed that Itgb1 plays a critical role in maintaining the balance of hypothalamic GnRH-tanycyte unit during EA treatment. CONCLUSIONS EA exerts therapeutic effects on PCOS by targeting hypothalamic GnRH-tanycyte unit, with Itgb1 identified as a key factor. MA primarily functions through neurotransmitter regulation. These findings highlight potential hypothalamic targets and provide new insights into the distinct mechanisms of EA and MA.
Collapse
Affiliation(s)
- Yu Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Yicong Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Yuning Chen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Wenhan Lu
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoyu Tong
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Jiajia Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Wenhao Gao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Rui Huang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Wei Hu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China.
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
15
|
Li X, Hou M, Zhang F, Ji Z, Cai Y, Shi Y. Per- and Polyfluoroalkyl Substances and Female Health Concern: Gender-based Accumulation Differences, Adverse Outcomes, and Mechanisms. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:1469-1486. [PMID: 39803974 DOI: 10.1021/acs.est.4c08701] [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: 01/29/2025]
Abstract
The deleterious health implications of perfluoroalkyl and polyfluoroalkyl substances (PFAS) are widely recognized. Females, in contrast to males, exhibit unique pathways for PFAS exposure and excretion, leading to complex health outcomes. The health status of females is largely influenced by hormone-related processes. PFAS have been reported to be associated with various aspects of female health, including reproductive system disorders and pregnancy-related diseases. In this article, we provide insights into the correlations between PFAS and female-prevalent diseases. Current epidemiological and toxicological evidence has demonstrated that the adverse effects of PFAS on the health of the female reproductive system are primarily attributed to the disruption of the hypothalamic-pituitary-gonadal (HPG) axis and hormonal homeostasis. However, these findings do not sufficiently elucidate the intricate associations between PFAS and specific diseases. Furthermore, autoimmune disorders, another category that is more prevalent in women compared to men, require additional investigation. Immune biomarkers pertinent to autoimmune disorders have been observed to be influenced by PFAS exposure, although epidemiological evidence is insufficient to substantiate these relations. Further thorough exploration encompassing epidemiological and toxicological studies is essential to elucidating the inherent influence of PFAS on human pathologies. Additionally, comprehensive investigations into female health issues beyond their reproductive functions is essential.
Collapse
Affiliation(s)
- Xin Li
- 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, UCAS, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Minmin Hou
- 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, UCAS, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feng Zhang
- Environmental Science Research & Design Institute of Zhejiang Province and Key Laboratory of Environmental Pollution Control Technology of Zhejiang Province, HangzhouZhejiang310007, China
| | - Zhengquan Ji
- Environmental Science Research & Design Institute of Zhejiang Province and Key Laboratory of Environmental Pollution Control Technology of Zhejiang Province, HangzhouZhejiang310007, China
| | - Yaqi Cai
- 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, UCAS, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yali Shi
- 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, UCAS, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
16
|
Xie Y, Li X, Wang M, Chu M, Cao G. Lin28b-let-7 Modulates mRNA Expression of GnRH1 Through Multiple Signaling Pathways Related to Glycolysis in GT1-7 Cells. Animals (Basel) 2025; 15:120. [PMID: 39858120 PMCID: PMC11758636 DOI: 10.3390/ani15020120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Lin28b and let-7 miRNA regulate mammalian pubertal initiation and Gonadotropin-releasing hormone (GnRH) production. However, it remains unclear which signaling pathways Lin28b regulates to modulate GnRH production. In this study, the mRNA expression levels of Lin28b and let-7 in the pubertal and juvenile goat hypothalamus and pituitary gland were detected, and Lin28b expression in the pubertal hypothalamus decreased significantly compared with that in juvenile tissues. It was predicted that Lin28b might inhibit GnRH1 expression, which was verified in the GnRH-producing cell model GT1-7 cells. Lin28b inhibited GnRH1 expression and promoted Kiss1/Gpr54 signaling. The pyruvate content and the expression of Hif1a and Hk2, which were related to glycolysis, were also promoted by Lin28b overexpression. Additionally, 77 differentially expressed miRNAs (DEMIs) in Lin28b-overexpressed GT1-7 cells were identified. Bioinformatics analysis and mRNA expression of the target genes of DEMIs revealed that the MAPK and PI3K-AKT-mTOR signaling pathways were key pathways that involved the regulatory effect of Lin28b on GnRH. In GT1-7 cells, GnRH1 expression was suppressed by blocking mTOR signaling with rapamycin, which was rescued by Lin28b overexpression. These results indicate that Lin28b-let-7 regulates GnRH1 expression through several pathways, including the Kiss1/Gpr54, MAPK, and mTOR signaling pathways, which are all related to glucose metabolism and provide new insights into the molecular mechanism of the regulatory role of Lin28b on GnRH production.
Collapse
Affiliation(s)
- Yujing Xie
- School of Agriculture and Biology, Liaocheng University, Liaocheng 252059, China (X.L.)
| | - Xin Li
- School of Agriculture and Biology, Liaocheng University, Liaocheng 252059, China (X.L.)
| | - Meng Wang
- School of Agriculture and Biology, Liaocheng University, Liaocheng 252059, China (X.L.)
| | - Mingxing Chu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Guiling Cao
- School of Agriculture and Biology, Liaocheng University, Liaocheng 252059, China (X.L.)
| |
Collapse
|
17
|
Ghasemzadeh Hasankolaei M, Evans NP, Elcombe CS, Lea RG, Sinclair KD, Padmanabhan V, Bellingham M. In-utero exposure to real-life environmental chemicals disrupts gene expression within the hypothalamo-pituitary-gonadal axis of prepubertal and adult rams. ENVIRONMENTAL RESEARCH 2025; 264:120303. [PMID: 39510237 DOI: 10.1016/j.envres.2024.120303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/20/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
Environmental chemicals (ECs) have been associated with a broad range of disorders and diseases. Daily exposure to various ECs in the environment, or real-life exposure, has raised significant public health concerns. Utilizing the biosolids-treated pasture (BTP) sheep model, this study demonstrates that in-utero exposure to a real-life EC mixture disrupts hypothalamo-pituitary-gonadal (HPG) axis gene expression and reproductive traits in prepubertal (8-week-old, 8w) and adult (11-month-old) male sheep. Ewes were maintained on either BTP or pastures fertilized with inorganic fertilizer [control (C)] from approximately one month prior to insemination until around parturition. Thereafter, all animals were kept under control conditions. Effects on reproductive parameters including testosterone concentrations and the expression of key genes in the HPG axis were evaluated in eight-week-old and adult male offspring from both C and biosolids-exposed (B) groups. Results showed that, at 8w, relative to C (n = 11), B males (n = 11) had lower body weight, and altered testicular expression of HSD3B1, LHR and HSD17B3, BMP4, ABP, P27kip and CELF1. Principal component analysis (PCA) identified two 8w B subgroups, based on hypothalamic expression of GnRH, ESR1, and AR, and pituitary expression of KISSR. The two subgroups also exhibited different serum testosterone concentrations. The largest biosolids effects were observed in the hypothalamus of adult rams with NKB, ESR1, KISS1, AR, DLK1 and GNRH1 mRNA expression differing between B (n = 10) and C (n = 11) rams. Testicular steroidogenic enzymes CYP11A1 and HSD3B1 mRNA expression also differed between exposure groups. PCA identified two adult B subgroups, with BS1 (n = 6) displaying hypothalamic effects and BS2 (n = 4) both hypothalamic and testicular effects. The subgroups also differed in circulating testosterone concentrations. These findings demonstrate that exposure to a real-life EC mixture may predispose some males to infertility, by disrupting key functional HPG markers before puberty with consequent downstream effects on steroid hormones and spermatogenesis.
Collapse
Affiliation(s)
- Mohammad Ghasemzadeh Hasankolaei
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK.
| | - Neil P Evans
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Chris S Elcombe
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Richard G Lea
- University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Kevin D Sinclair
- University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | | | - Michelle Bellingham
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| |
Collapse
|
18
|
Scandlan OLM, Favetta LA. Do Delta-9-tetrahydrocannabinol and Cannabidiol have opposed effects on male fertility? Toxicol Lett 2025; 403:94-104. [PMID: 39657895 DOI: 10.1016/j.toxlet.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Cannabis sativa is a complex plant, renowned for its diverse array of bioactive compounds, the most prominent of which are delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD). These compounds exhibit markedly opposing pharmacological effects, with THC being primarily psychoactive and CBD known for its non-psychoactive properties. In recent years, there has been growing interest in the potential health implications of these compounds, particularly concerning male reproductive health. Accumulating evidence over the past decade has alluded to the potential negative effects of THC, including its association with reduced sperm quality, altered hormone levels, changes in genetic and epigenetic profiles, and potential impacts on fertility. Conversely, emerging studies suggest that CBD may exert protective and beneficial effects on male reproductive health, possibly through its anti-inflammatory and antioxidant properties. This review aims to provide a comprehensive analysis of the current scientific literature, delineating the mechanisms by which THC and CBD influence male reproductive health, highlighting the disparities in their effects, and discussing the clinical and therapeutic implications of these findings.
Collapse
Affiliation(s)
- Olivia L M Scandlan
- Reproductive Health and Biotechnology Laboratory, Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Laura A Favetta
- Reproductive Health and Biotechnology Laboratory, Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
19
|
Forrester-Fronstin Z, Barrett AR, Mondschein AS, Johnson JM, Cordes CN, Lawton-Stone TS, Schatz KC, Paul MJ. Exogenous estradiol impacts anxiety-like behavior of juvenile male and female Siberian hamsters in a dose-dependent manner. Horm Behav 2025; 167:105674. [PMID: 39731972 DOI: 10.1016/j.yhbeh.2024.105674] [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: 07/19/2024] [Revised: 11/27/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024]
Abstract
Anxiety is among the most prevalent mental health issues in children. While it is well established that gonadal steroids influence anxiety-like behavior in adulthood, a potential role in prepubertal juveniles has been overlooked because it is commonly thought that the gonads are quiescent during the juvenile period. However, the juvenile gonads secrete measurable amounts of steroids, and we have recently found that prepubertal ovariectomy decreases anxiety-like behavior of juvenile Siberian hamsters in the light/dark box test. The present study tested whether an injection of estradiol benzoate (1 μg or 10 μg, SC) to gonadectomized hamsters (Exp. 1) or chronic suppression of endogenous estradiol with the aromatase inhibitor, letrozole (2 mg/kg, PO), to intact hamsters (Exp. 2) affects anxiety-like behavior in the light/dark box test during the juvenile phase. Estradiol benzoate altered anxiety-like behavior of both male and female juveniles in a dose-dependent manner, with anxiolytic actions at the low dose, but no effect at the high dose. Similar effects were seen for activity measures, albeit only in females. Letrozole suppressed uterine weights demonstrating an active role for endogenous estradiol during the juvenile phase. Anxiety-like behavior, however, was impacted by the administration procedure itself, preventing conclusions on letrozole's actions on behavior. While the role for endogenous estradiol in juvenile anxiety-like behavior remains unresolved, the present findings indicate that the neural centers regulating affective behavior are responsive to exogenous estradiol prior to puberty. These findings highlight the potential impact of exogenous estrogen exposures on juvenile affective behavior.
Collapse
Affiliation(s)
| | - Abigal R Barrett
- Department of Psychology, University at Buffalo, SUNY, Buffalo, NY, USA
| | | | - Jordan M Johnson
- Department of Psychology, University at Buffalo, SUNY, Buffalo, NY, USA
| | - Chloe N Cordes
- Department of Psychology, University at Buffalo, SUNY, Buffalo, NY, USA
| | | | - Kelcie C Schatz
- Department of Psychology, University at Buffalo, SUNY, Buffalo, NY, USA
| | - Matthew J Paul
- Department of Psychology, University at Buffalo, SUNY, Buffalo, NY, USA; Neuroscience Program, University at Buffalo, SUNY, Buffalo, NY, USA; Evolution, Ecology, and Behavior Program, University at Buffalo, SUNY, NY, USA
| |
Collapse
|
20
|
Meczekalski B, Nowicka A, Bochynska S, Szczesnowicz A, Bala G, Szeliga A. Kisspeptin and Endometriosis-Is There a Link? J Clin Med 2024; 13:7683. [PMID: 39768606 PMCID: PMC11678265 DOI: 10.3390/jcm13247683] [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: 10/01/2024] [Revised: 11/23/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
This article presents a narrative review that explores the potential link between kisspeptin-a key regulator of the hypothalamic-pituitary-gonadal axis-and the pathogenesis of endometriosis. Kisspeptin plays a significant role in regulating reproductive functions by modulating the release of gonadotropin-releasing hormone (GnRH), which in turn stimulates the secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH). Recent studies suggest that kisspeptin may also impact peripheral reproductive tissues and influence inflammatory processes involved in the development of endometriosis. Altered kisspeptin signaling has been associated with the abnormal hormonal environment observed in endometriosis, which affects menstrual cycles and ovarian function. Research indicates that women with endometriosis exhibit altered levels of kisspeptin and its receptor, KISS1R, in both eutopic and ectopic endometrial tissues, suggesting a role in disease progression, particularly in tissue invasion and lesion formation. Kisspeptin's role in regulating matrix metalloproteinases (MMPs), enzymes essential for tissue remodeling, further supports its potential contribution to the pathophysiology of endometriosis. Moreover, kisspeptin-based therapeutic strategies are currently under investigation, with the aim of providing targeted treatments that reduce the side effects commonly associated with existing therapies. Despite promising findings, further research is needed to fully understand the mechanisms by which kisspeptin influences endometriosis.
Collapse
Affiliation(s)
- Blazej Meczekalski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Agata Nowicka
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Stefania Bochynska
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Aleksandra Szczesnowicz
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Gregory Bala
- UCD School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Anna Szeliga
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| |
Collapse
|
21
|
Ye S, Zhou L. Role of serum kisspeptin as a biomarker to detect miscarriage: a systematic review and meta-analysis. HUM FERTIL 2024; 27:2417934. [PMID: 39468787 DOI: 10.1080/14647273.2024.2417934] [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: 04/24/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024]
Abstract
Miscarriage is a common adverse pregnancy outcome with physical and emotional effects. Identifying predictive miscarriage biomarkers should improve early detection and management. Serum kisspeptin, known for its critical role in reproductive biology, has emerged as a potential biomarker for miscarriage. With this systematic review and meta-analysis, we aimed to assess the association between serum kisspeptin levels and the miscarriage risk. We systematically searched PubMed, Embase, and Cochrane Library databases for studies published up to February 2024, examining the association between serum kisspeptin levels and miscarriage. Eligible studies were observational designs that reported kisspeptin levels in women with and without miscarriage. We included 12 studies involving 2,050 participants. Pooled analysis demonstrated that low serum kisspeptin levels were significantly associated with an increased risk of miscarriage (standardized mean difference = -2.750; 95%CI: -4.357 to -1.143), with substantial heterogeneity (I2 = 98.7%). The pooled area under the curve from three studies indicated high diagnostic accuracy (AUC = 0.903; 95%CI: 0.860-0.946). Low serum kisspeptin levels are significantly associated with an increased miscarriage risk, suggesting that kisspeptin could serve as an effective biomarker for early detection. However, the significant heterogeneity among studies and the lack of standardized measurement protocols emphasize the necessity of further research before clinical implementation. SYSTEMATIC REVIEW REGISTRATION: (Registration ID: CRD42024520639).
Collapse
Affiliation(s)
- Sisi Ye
- Department of Laboratory Department, Huzhou Maternity & Child Health Care Hospital, Huzhou, China
| | - Liping Zhou
- Department of Gynaecology and Obstetrics, Huzhou Maternity & Child Health Care Hospital, Huzhou, China
| |
Collapse
|
22
|
Mate NA, Wadhwa G, Taliyan R, Banerjee A. Impact of polyamine supplementation on GnRH expression, folliculogenesis, and puberty onset in young mice. Theriogenology 2024; 229:202-213. [PMID: 39217649 DOI: 10.1016/j.theriogenology.2024.07.028] [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: 04/16/2024] [Revised: 07/10/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The hypothalamic-pituitary-gonadal (HPG) axis is pivotal in regulating reproductive functions, with gonadotropin-releasing hormone (GnRH) acting as a central regulator. Recently, polyamines have been shown to regulate the HPG axis, including GnRH expression and ovarian biology in old and adult rodents. The present study firstly highlights the age-specific variation in the polyamine and their corresponding biosynthetic enzymes in the ovary during aging, and further, the study focuses on the effect of polyamines, putrescine, and agmatine, in young female mice. METHOD AND RESULT Immunofluorescence analysis revealed age-related differences in the expression of ornithine decarboxylase 1 (ODC1), spermine (SPM), and spermidine (SPD) in the ovaries, with adult mice exhibiting significantly higher expression levels compared to young and old mice. Likewise, qPCR analysis showed the mRNA levels of Odc1, Spermidine synthase (Srm), and Spermine synthase (Sms) show a significant increase in adult ovaries, which is then followed by a significant decline in old age. Histological examination demonstrated morphological alterations in the ovaries with age, including decreased follicle numbers and increased stromal cells in old mice. Furthermore, treatment with putrescine, a polyamine, in young mice resulted in larger ovaries and increased follicle numbers compared to controls. Additionally, serum levels of gonadotropin-releasing hormone (GnRH) and progesterone (P4) were measured, showing elevated levels in polyamine-treated mice. GnRH mRNA expression also increased significantly. Gene expression analysis revealed upregulation of genes associated with folliculogenesis such as Fshr, Bmp15, Gdf9, Amh, Star, Hsdb3, and Plaur in the ovaries and onset of puberty such as Tac2, and Kiss1, and a decrease in Mkrn3 in the hypothalamus of polyamine-treated mice. CONCLUSION This study investigates the effect of polyamines in young immature female mice, shedding light on their role in upregulating GnRH, and enhancing folliculogenesis. Overall, these findings suggest that polyamines play a crucial role in ovarian aging and HPG axis regulation, offering potential therapeutics to reinstate fertility in reproductively challenged individuals.
Collapse
Affiliation(s)
- Nayan Anand Mate
- Department of Biological Sciences, Birla Institute of Technology and Science, K K Birla Goa Campus, Zuarinagar, Goa, India
| | - Geetika Wadhwa
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, 333031, India
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, 333031, India
| | - Arnab Banerjee
- Department of Biological Sciences, Birla Institute of Technology and Science, K K Birla Goa Campus, Zuarinagar, Goa, India.
| |
Collapse
|
23
|
Zhu J, Wang JX, Jin ZY, Li D, Qi S, Han SZ, Chang SY, Yan J, Kang JD, Quan LH. Eicosatrienoic acid inhibits estradiol synthesis through the CD36/FOXO1/CYP19A1 signaling pathway to improve PCOS in mice. Biochem Pharmacol 2024; 229:116517. [PMID: 39236935 DOI: 10.1016/j.bcp.2024.116517] [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: 04/25/2024] [Revised: 08/11/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a common metabolic and endocrine disorder characterized by abnormal elevation in hormone levels, with currently lacking effective treatment options. N-3 polyunsaturated fatty acids (PUFA) have broad pharmacological activity and play a beneficial role in the development of PCOS. In this study, we observed that n-3 PUFA-eicosatrienoic acid (ETA) improves the estrous cycle and ovarian morphology in dehydroepiandrosterone (DHEA)-induced PCOS mice, particularly serum hormone levels. Additionally, it suppresses the expression of CYP19A1 and E2 synthesis in human granulosa-like tumor cell line (KGN) cells. Further investigation revealed that ETA significantly upregulates the expression of CD36, cAMP, P-PKA, and FOXO1 in KGN cells and mouse ovaries to lower E2 levels. This conclusion was supported by inhibiting CD36 and FOXO1 at both the mouse and cellular levels. Additionally, ETA treatment decreased the expression of ESR1, Kiss1, Gnrh in the hypothalamus, and GnRHR, Lhβ, Egr1, Pitx1, Sf1 in the pituitary of PCOS mice. No differences were observed after ETA treatment in the CD36 and FOXO1 inhibitor groups, indicating that ETA improves PCOS mice by regulating the hypothalamic-pituitary axis through E2 synthesis inhibition. In summary, we have elucidated for the first time the mechanism by which CD36 regulates E2 synthesis in ovarian granulosa cells and demonstrated that ETA activates the CD36 receptor to inhibit E2 synthesis through the cAMP/PKA/FOXO1/CYP19A1 signaling pathway, thereby improving hormonal imbalance and treating PCOS. This provides a new strategy for the effective prevention and treatment of PCOS.
Collapse
Affiliation(s)
- Jun Zhu
- College of Integration Science, Yanbian University, Yanji 133002, China
| | - Jun-Xia Wang
- Department of Animal Science, College of Agricultural, Yanbian University, Yanji 133002, China
| | - Zheng-Yun Jin
- Department of Animal Science, College of Agricultural, Yanbian University, Yanji 133002, China
| | - Dongxu Li
- College of Integration Science, Yanbian University, Yanji 133002, China
| | - Shaobo Qi
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Sheng-Zhong Han
- Department of Animal Science, College of Agricultural, Yanbian University, Yanji 133002, China
| | - Shuang-Yan Chang
- Department of Animal Science, College of Agricultural, Yanbian University, Yanji 133002, China
| | - Jin Yan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Jin-Dan Kang
- Department of Animal Science, College of Agricultural, Yanbian University, Yanji 133002, China.
| | - Lin-Hu Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China.
| |
Collapse
|
24
|
Han S, Yu C, Qiu M, Xiong X, Peng H, Song X, Hu C, Zhang Z, Xia B, Yang L, Chen J, Zhu S, Li W, Yang C. USP13 regulates ferroptosis in chicken follicle granulosa cells by deubiquitinating ATG7. Poult Sci 2024; 103:104209. [PMID: 39214053 PMCID: PMC11402030 DOI: 10.1016/j.psj.2024.104209] [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: 06/27/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
The development and maturation of follicles are intricately linked to egg production and reproductive performance of chickens. Granulosa cells death directly affects the development and maturation of follicles, thereby impacting the reproductive performance of hens. Ferroptosis is a new type of cell death, it is unknown how it affects the growth and development of chicken follicles. In this study, RNA-seq analysis revealed significant differences in the expression of ferroptosis-related genes between normal follicles and atretic follicles, suggesting a potential role for ferroptosis in follicle growth and development. In addition, we found that ubiquitin-specific protease 13 (USP13) was significantly upregulated in atrophic follicles. Overexpression of USP13 results in depletion of glutathione (GSH), peroxidation of lipids, accumulation of iron, and activation of ferroptosis in chicken granulosa cells. In contrast, USP13 knockdown significantly inhibited ferroptosis events. Mechanistically, USP13 prevents the degradation of autophagy related 7 (ATG7) by deubiquitinating it, thereby enhancing the stability of ATG7 protein and ultimately promoting ferroptosis. In conclusion, this study elucidates the crucial role of the USP13-ATG7 axis in regulating ferroptosis in chicken follicle granulosa cells, thereby presenting a novel avenue for molecular breeding research in chickens.
Collapse
Affiliation(s)
- Shunshun Han
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Chunlin Yu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Mohan Qiu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Xia Xiong
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Han Peng
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Xiaoyan Song
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Chenming Hu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Zengrong Zhang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Bo Xia
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Li Yang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Jialei Chen
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Shiliang Zhu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Wen Li
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Chaowu Yang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China.
| |
Collapse
|
25
|
Salvio G, Balercia G, Kadioglu A. Hypogonadotropic hypogonadism as a cause of NOA and its treatment. Asian J Androl 2024:00129336-990000000-00258. [PMID: 39513636 DOI: 10.4103/aja202483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 08/19/2024] [Indexed: 11/15/2024] Open
Abstract
ABSTRACT Hypogonadotropic hypogonadism (HH) represents a relatively rare cause of nonobstructive azoospermia (NOA), but its knowledge is crucial for the clinical andrologists, as it represents a condition that can be corrected with medical therapy in 3 quarters of cases. There are forms of congenital HH, whether or not associated with an absent sense of smell (anosmic HH or Kallmann syndrome, and normosmic HH, respectively), and forms of acquired HH. In congenital HH, complete absence of pubertal development is characteristic. On the other hand, if the deficit occurs after the time of pubertal development, as in acquired HH patients, infertility and typical symptoms of late-onset hypogonadism are the main reasons for seeking medical assistance. Gonadotropin-releasing hormone (GnRH) or gonadotropin replacement therapy is the mainstay of drug therapy and offers excellent results, although a small but significant proportion of patients do not achieve sufficient responses.
Collapse
Affiliation(s)
- Gianmaria Salvio
- Endocrinology Clinic, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona 60126, Italy
| | - Giancarlo Balercia
- Endocrinology Clinic, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona 60126, Italy
| | - Ates Kadioglu
- Section of Andrology, Department of Urology, Istanbul School of Medicine, Istanbul 34093, Türkiye
| |
Collapse
|
26
|
Liang Y, Yuan Y, Yang J. Acupoint catgut embedding: a potential intervention strategy for obesity-related precocious puberty. Front Endocrinol (Lausanne) 2024; 15:1448111. [PMID: 39469573 PMCID: PMC11513323 DOI: 10.3389/fendo.2024.1448111] [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: 06/12/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Abstract
Introduction Obesity-related precocious puberty is induced by obesity, and acupoint catgut embedding (ACE) therapy is known to treat obesity. This study aims to validate the hypothesis that ACE can delay the onset of obesity-related precocious puberty. Methods Female Sprague-Dawley rats, 21 days old, were randomly divided into three groups: the high-fat diet combined with ACE treatment group (ACE), the high-fat diet group (HFD), and the normal control diet group (NCD), with 8 rats in each group. The vaginal opening (VO) time was monitored, and serum levels of luteinizing hormone (LH), follicle-stimulating hormone (FSH), and total estradiol (E2) were measured, followed by statistical analysis. Results Kaplan-Meier survival curves, with VO as the endpoint, showed that vaginal opening was delayed in the ACE group compared to the HFD group, with a statistically significant difference (p < 0.05). The changes in levels of FSH, LH, and E2 indicated that sexual development was delayed in the ACE group compared to the HFD group and was more similar to the NCD group. Discussion Combining the vaginal opening time and changes in hormone levels, this study confirms the potential role of ACE in delaying the onset of obesity-related precocious puberty.
Collapse
Affiliation(s)
- Yun Liang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yuan Yuan
- Department of Pediatric Surgery, Affiliated Hospital, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jie Yang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
27
|
Ding Q, Hao T, Gao Y, Jiang S, Zhu Y, Huang Y, Liang Y. Associations of Co-Exposure to Polycyclic Aromatic Hydrocarbons and Heavy Metals with Sex Steroid Hormones among Children Aged 6-19 Years. Horm Res Paediatr 2024:1-11. [PMID: 39396497 DOI: 10.1159/000541875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024] Open
Abstract
INTRODUCTION Polycyclic aromatic hydrocarbons (PAHs) and heavy metals (HMs) are endocrine-disrupting chemicals (EDCs) that may have a combined effect on sex hormone levels in children. This study investigated the correlations between co-exposure to PAHs and HMs and levels of sex steroid hormones in children. METHODS We employed the data from the National Health and Nutrition Examination Survey (NHANES) from 2013 to 2016, including 1,167 participants aged 6-19 years. Sex hormone indicators include testosterone (TT), estradiol (E2), sex hormone-binding globulin (SHBG), free androgen index (FAI), and the TT/E2 ratio. Weighted multivariate linear regression, weighted quantile sum (WQS) regression, and Bayesian kernel machine regression (BKMR) were used to analyze the associations between co-exposure to PAHs and HMs and sex steroid hormone levels. RESULTS Co-exposure to PAHs and HMs was associated with a 16.2% reduction (95% CI [-0.321, -0.004]) in SHBG level among prepubertal males and a 16% reduction (95% CI [-0.30, -0.03]) in E2 level among pubertal males by the WQS regression, and cadmium (Cd) and mercury (Hg) contributed the highest weight, respectively. In the Bayesian kernel machine regression (BKMR) model, co-exposure to PAHs and HMs was positively associated with TT/E2 in pubertal males and negatively correlated with FAI in pubertal females, and 1-hydroxypyrene (1-PYR) and Cd were the most important components, respectively. CONCLUSIONS Co-exposure to PAHs and HMs was associated with sex hormone levels in children. These findings highlight the necessity for preventing the effects of these chemicals on sex hormones.
Collapse
Affiliation(s)
- Qi Ding
- School of Public Health, Wannan Medical College, Wuhu, China
| | - Tingting Hao
- School of Public Health, Wannan Medical College, Wuhu, China
| | - Yuan Gao
- School of Public Health, Wannan Medical College, Wuhu, China
| | | | - Yu Zhu
- School of Public Health, Wannan Medical College, Wuhu, China
| | - Yue'e Huang
- School of Public Health, Wannan Medical College, Wuhu, China
| | - Yali Liang
- School of Public Health, Wannan Medical College, Wuhu, China
| |
Collapse
|
28
|
Gong J, Lv Y, Meng Y, Zhang W, Jiang X, Xiao M. Effects of prenatal stress on reproductive function of male offspring through the KISS1 system. Endocr Connect 2024; 13:e240027. [PMID: 39140811 PMCID: PMC11466243 DOI: 10.1530/ec-24-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 08/14/2024] [Indexed: 08/15/2024]
Abstract
Prenatal stress can lead to the programming of the neuroendocrine system in male offspring, disrupting the hypothalamic testicular axis and adversely affecting the reproductive health of male offspring. This study aimed to determine the long-term effects of prenatal stress on the KISS1 system in male offspring and the effects on reproductive function in male offspring. Sixteen pregnant females were divided into a prenatal control group (PC, n = 8) and a prenatal stress group (PS, n = 8). The PS group was modeled with chronic unpredictable mild stress (CUMS) from day 1 of gestation to full-term delivery. Differences between the two groups in various maternal parameters, including glucocorticoid secretion, litter size, and the effects of male offspring birth weight, the KISS1 system, and reproductive function, were determined. Male offspring of PS dams had lower birth weights compared to prenatal controls.KISS1 gene expression is reduced at birth and in adult PS offspring, and its receptor KISS1-R protein is similarly reduced in PS offspring at birth and adulthood. In adulthood, PS male offspring show significantly reduced sex hormone production, altered testicular morphology, reduced maturation of their supporting cells, and decreased expression of connexin 43 (CX43), leading to an altered sperm microenvironment and reduced sperm quality. In conclusion, prenatal stress leads to adverse changes in the KISS1 system in male offspring and decreased reproductive function.
Collapse
Affiliation(s)
- Jian Gong
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Yinjuan Lv
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Yuhao Meng
- Hubei University of Chinese Medicine, Wuhan, China
| | | | | | - Min Xiao
- Laboratory Animal Center, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
29
|
Hu R, Geng Y, Huang Y, Liu Z, Li F, Song K, Ma W, Dong H, Zhang M, Lei T, Song Y, Zhang Z. Jiawei Buzhong Yiqi Decoction attenuates polycystic ovary syndrome through regulating kisspeptin-GPR54-AKT-SHBG system. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155931. [PMID: 39116604 DOI: 10.1016/j.phymed.2024.155931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 07/27/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is one of the most common reproductive endocrine disorders. Accumulated evidence has suggested the indispensable role of kisspeptin-G protein-coupled receptor (GPR54) system and SHBG in development of PCOS. However, potential mechanisms and their relationship are unclear. Jiawei Buzhong Yiqi Decoction (JWBZYQ) has been reported to ameliorate obese PCOS. Whereas, potential mechanisms remain elusive. PURPOSE To determine whether JWBZYQ attenuates PCOS by regulating the kisspeptin-GPR54 system and SHBG production. And to explore potential mechanisms. METHODS An overweight PCOS rat model was developed with testosterone propionate (TP) and high-fat diet (HFD). The efficacy of JWBZYQ was assessed by tracking changes in weight, estrous cycle, ovarian morphology, and serum sex hormone levels. Additionally, kisspeptin-GPR54 system expression in multiple organs and PI3K-AKT pathway activity in liver of different rats were detected. Modifications in SHBG production were also measured. Kisspeptin54 was administered to establish a cellular model. The levels of AKT phosphorylation and SHBG protein within HepG2 cells were analyzed. Finally, confirmatory studies were performed using AKT phosphorylation activator and inhibitor. RESULTS JWBZYQ effectively attenuated the overweight, disrupted estrous cycle, altered sex hormone levels, and aberrant ovarian morphology in PCOS rats. Meanwhile, PCOS rats exhibited elevated levels of kisspeptin and GPR54, along with reduced SHBG levels, which could be reversed by JWBZYQ. These alterations might be connected with the activation of AKT phosphorylation. In vitro experiment identified that JWBZYQ could rectify the hyperactivated AKT phosphorylation and deficient production of SHBG caused by kisspeptin54. CONCLUSIONS Overexpressed kisspeptin-GPR54 system inhibited SHBG synthesis in PCOS. JWBZYQ curtailed the exorbitant expression of kisspeptin and GPR54, which moderated the rise in AKT phosphorylation and subsequently promoted the production of SHBG.
Collapse
Affiliation(s)
- Runan Hu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuli Geng
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanjing Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhuo Liu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fan Li
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kunkun Song
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenwen Ma
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Haoxu Dong
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mingmin Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ting Lei
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yufan Song
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhuo Zhang
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
30
|
Mohamed ZI, Sivalingam M, Radhakrishnan AK, Jaafar F, Zainal Abidin SA. Chronic unpredictable stress (CUS) reduced phoenixin expression, induced abnormal sperm and testis morphology in male rats. Neuropeptides 2024; 107:102447. [PMID: 38870753 DOI: 10.1016/j.npep.2024.102447] [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: 04/16/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Chronic stress caused by prolonged emotional pressure can lead to various physiological issues, including reproductive dysfunction. Although reproductive problems can also induce chronic stress, the impact of chronic stress-induced reproductive dysfunction remains contentious. This study investigates the effects of chronic unpredictable stress (CUS) on reproductive neuropeptides, sperm quality, and testicular morphology. Sixteen twelve-week-old Sprague Dawley rats were divided into two groups: a non-stress control group and a CUS-induced group. The CUS regimen involved various stressors over 28 days, with both groups undergoing behavioural assessments through sucrose-preference and forced-swim tests. Hypothalamic gene expression levels of CRH, PNX, GPR173, kisspeptin, GnRH, GnIH, and spexin neuropeptides were measured via qPCR, while plasma cortisol, luteinizing hormone (LH), and testosterone concentrations were quantified using ELISA. Seminal fluid and testis samples were collected for sperm analysis and histopathological evaluation, respectively. Results showed altered behaviours in CUS-induced rats, reflecting stress impacts. Hypothalamic corticotropin-releasing hormone (CRH) expression and plasma cortisol levels were significantly higher in CUS-induced rats compared to controls (p < 0.05). Conversely, phoenixin (PNX) expression decreased in the CUS group (p < 0.05), while kisspeptin, spexin, and gonadotropin-inhibitory hormone (GnIH) levels showed no significant differences between groups. Despite a significant increase in GnRH expression (p < 0.05), plasma LH and testosterone concentrations were significantly lower (p < 0.05) in CUS-induced rats. Histopathological analysis revealed abnormal testis morphology in CUS-induced rats, including disrupted architecture, visible interstitial spaces between seminiferous tubules, and absence of spermatogenesis. In conclusion, CUS affects reproductive function by modulating PNX and GnRH expression, influencing cortisol levels, and subsequently reducing plasma LH and testosterone concentrations. This study highlights the complex interplay between chronic stress and reproductive health, emphasizing the significant impact of stress on reproductive functions.
Collapse
Affiliation(s)
- Zahra Isnaini Mohamed
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 46150 Bandar Sunway, Selangor, Malaysia
| | - Mageswary Sivalingam
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 46150 Bandar Sunway, Selangor, Malaysia
| | - Ammu K Radhakrishnan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 46150 Bandar Sunway, Selangor, Malaysia
| | - Faizul Jaafar
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 46150 Bandar Sunway, Selangor, Malaysia
| | - Syafiq Asnawi Zainal Abidin
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 46150 Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
31
|
Fidecicchi T, Giannini A, Chedraui P, Luisi S, Battipaglia C, Genazzani AR, Genazzani AD, Simoncini T. Neuroendocrine mechanisms of mood disorders during menopause transition: A narrative review and future perspectives. Maturitas 2024; 188:108087. [PMID: 39111089 DOI: 10.1016/j.maturitas.2024.108087] [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: 04/10/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
The menopause transition is an important period in a woman's life, during which she is at an increased risk of mood disorders. Estrogen and progesterone fluctuations during the menopausal transition and very low levels of estradiol after menopause have a profound effect on the central nervous system (CNS), causing an imbalance between excitatory and inhibitory inputs. Changes in neurotransmission and neuronal interactions that occur with estradiol withdrawal disrupt the normal neurological balance and may be associated with menopausal symptoms. Hot flushes, depressed mood and anxiety are all symptoms of menopause that are a consequence of the complex changes that occur in the CNS, involving many signaling pathways and neurotransmitters (i.e. γ-aminobutyric acid, serotonin, dopamine), neurosteroids (i.e. allopregnanolone), and neuropeptides (i.e. kisspeptin, neurokinin B). All these pathways are closely linked, and the complex interactions that exist are not yet fully understood. This review summarizes the neuroendocrine changes in the CNS during the menopausal transition, with particular emphasis on those that underlie mood changes.
Collapse
Affiliation(s)
- Tiziana Fidecicchi
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 67, 56126 Pisa, PI, Italy
| | - Andrea Giannini
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 67, 56126 Pisa, PI, Italy.
| | - Peter Chedraui
- Escuela de Posgrado en Salud, Universidad Espíritu Santo, Av. Samborondón 5, Samborondón 092301, Ecuador
| | - Stefano Luisi
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 67, 56126 Pisa, PI, Italy.
| | - Christian Battipaglia
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Largo del Pozzo, 71, 41125 Modena, MO, Italy.
| | - Andrea R Genazzani
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 67, 56126 Pisa, PI, Italy
| | - Alessandro D Genazzani
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Largo del Pozzo, 71, 41125 Modena, MO, Italy
| | - Tommaso Simoncini
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 67, 56126 Pisa, PI, Italy.
| |
Collapse
|
32
|
Kokori E, Olatunji G, Komolafe R, Ogieuhi IJ, Ukoaka B, Ajayi I, Aderinto N. Serum kisspeptin as a promising biomarker for PCOS: a mini review of current evidence and future prospects. Clin Diabetes Endocrinol 2024; 10:27. [PMID: 39343941 PMCID: PMC11440685 DOI: 10.1186/s40842-024-00190-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/06/2024] [Indexed: 10/01/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder affecting women of reproductive age, characterised by its multifactorial nature and intricate interplay of genetic, hormonal, and environmental factors. As the search for reliable biomarkers intensifies, serum kisspeptin emerges as a promising candidate due to its central role in regulating the hypothalamic-pituitary-gonadal (HPG) axis. This review aims to consolidate the evolving understanding of kisspeptin as a potential PCOS biomarker, comprehensively exploring its physiological basis, diagnostic challenges in PCOS, and clinical implications. Diagnostic challenges in PCOS are addressed, underscoring the limitations of current criteria and the need for objective and standardised biomarkers. Kisspeptin's introduction as a potential biomarker brings forth both promises and challenges in terms of its diagnostic utility. The review recognises the importance of standardisation in research methodologies and emphasises the exploration of genetic polymorphisms to enhance kisspeptin's robustness as a diagnostic tool.
Collapse
Affiliation(s)
- Emmanuel Kokori
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Rosemary Komolafe
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | | | - Bonaventure Ukoaka
- Department of Internal Medicine, Asokoro District Hospital, Abuja, Nigeria
| | - Irene Ajayi
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Old Oyo/ Ilorin Rd, P.M.B 4000, Ogbomosho, Oyo-State, 210214, Nigeria.
| |
Collapse
|
33
|
Liu J, Song J, Li Y, Gao D, Ma Q, Song X, Jiang J, Zhang Y, Wang R, Dong Z, Chen L, Qin Y, Yuan W, Guo T, Song Z, Dong Y, Zou Z, Ma J. Geneenvironment interaction between phthalate exposure and pubertal genetic polymorphisms on blood pressure variability in children: Exploring the moderating effects of lifestyle behaviours. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116966. [PMID: 39216218 DOI: 10.1016/j.ecoenv.2024.116966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Phthalates (PAEs) are synthetic compounds extensively employed in consumer products. Blood pressure (BP) in children can vary, the degree of visit-to-visit BP variability (VVV) is at least partially independent of BP. The interactions between PAEs exposure, pubertal-related genetic susceptibility and lifestyles on childhood VVV are not investigated. This study utilized data from a cohort collected from Oct 2017-2020 in Xiamen, China. Seven urine PAE metabolites were measured. The long-term VVV was characterized employing the standard deviation (SD) and average real variability. We constructed a genetic risk score (GRS) of pubertal-related genes and healthy lifestyle scores. Exposed to high levels of mono-2-ethyl-5-hydroxyhexyl phthalate (MEHHP) (OR=1.43, 95 %CI=1.07, 1.92) and mono-2-ethyl-5-oxohexyl phthalate (OR=1.36, 95 % CI=1.01, 1.83) was related to increased SBP-SD, and the OR for high SBP-SD related to high GRS was 1.38 (95 % CI=1.02, 1.85). Compared to participants who had low GRS and low MEHHP exposure, participants exhibiting high GRS and MEHHP levels were more likely to experience high SBP-SD (OR=2.00, P<0.05). Individuals exhibiting low GRS, low MEHHP levels, and adhering to healthy lifestyles were associated with the least probability of experiencing high SBP-SD (OR=0.31, P<0.05). Increased PAEs exposure could elevate childhood systolic VVV, and exacerbated the adverse impact of pubertal-related genetic susceptibility on the high VVV of SBP; however, healthy lifestyles might alleviate these adverse effects. Promoting healthy lifestyles and reducing PAEs exposure for preventing elevated BP variability among children is important, especially for individuals with greater genetic susceptibility to early pubertal onset. ENVIRONMENTAL IMPLICATION: Blood pressure (BP) in children can vary, as a noninvasive, inexpensive and applicable method, the extent of visit-to-visit variability (VVV) is at least partially independent of BP. The interactions between phthalates (PAEs) exposure, variants of puberty-related genes and lifestyles on VVV are not investigated. Increased childhood systolic VVV might be associated with PAEs exposure, with the associations more pronounced combined with pubertal genetic susceptibility. Yet, healthy habits could partly eliminate such adverse effects. Our study underscores the importance of advocating for healthy lifestyles and reducing exposure to PAEs, especially among individuals with high genetic susceptibility to early puberty onset.
Collapse
Affiliation(s)
- Jieyu Liu
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Jieyun Song
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Yanhui Li
- School of Nursing, Peking University, Beijing, China
| | - Di Gao
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Qi Ma
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Xinli Song
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Jianuo Jiang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Yi Zhang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Ruolin Wang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Ziqi Dong
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Li Chen
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Yang Qin
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Wen Yuan
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Tongjun Guo
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Zhiying Song
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Yanhui Dong
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China.
| | - Zhiyong Zou
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China.
| | - Jun Ma
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| |
Collapse
|
34
|
Plagens-Rotman K, Merks P, Pisarska-Krawczyk M, Jakubek E, Mizgier M, Jaskulska J, Wójcik M, Religioni U, Jarząbek-Bielecka G, Kędzia W. Oncosexology - selected issues taking into account the problem of sexological care of patients with cancer. PRZEGLAD MENOPAUZALNY = MENOPAUSE REVIEW 2024; 23:152-158. [PMID: 39464499 PMCID: PMC11504478 DOI: 10.5114/pm.2024.143481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 06/15/2024] [Indexed: 10/29/2024]
Abstract
As can be seen from research, 44% of oncological problems disclose problems in relationships with a partner. About 80% of oncological patients report deterioration of the quality of their sexual life. Although the situation improves, a significant majority of ill persons do not obtain assistance in the scope of sexuality during oncological treatment. Intimacy is an important sphere of life and can support the process of recovery. Intimacy plays special role in the difficult period of the fight against disease. Independently of the applied method of oncological treatment, side effects of therapy can influence one's sexual life. This means that an increasing number of persons who are healed from cancerous disease are exposed to the long-term undesirable influence of treatment. The process of oncological treatment has an essential impact on the intimate life of those who suffer from oncological diseases. To a great extent, this process and the disease itself contribute to a decrease of the quality of life due to the appearance of symptoms caused by earlier menopause, they negatively influence the feeling desire and sexual performance. Unfortunately, despite the development of oncosexology, sexual problems of oncologically ill persons are not noticed, and sometimes they are played down or overlooked due to everyday problems.
Collapse
Affiliation(s)
- Katarzyna Plagens-Rotman
- Centre for Paediatric, Adolescent Gynaecology and Sexology Division of Gynaecology, Department of Gynaecology, Poznań University of Medical Sciences, Poznań, Poland
| | - Piotr Merks
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszyński University, Warszawa, Poland
| | | | - Ewa Jakubek
- Department of Public Health, Faculty of Medicine and Health Sciences, Calisia University, Kalisz, Poland
| | - Małgorzata Mizgier
- Department of Sports Dietetics, Chair of Dietetics, Faculty of Health Sciences, Poznań University of Physical Education, Poznań, Poland
| | | | - Małgorzata Wójcik
- Department of Physiotherapy, Poznań University of Physical Education, Faculty of Sport Sciences in Gorzów Wielkopolski, Poland
| | - Urszula Religioni
- School of Public Health, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Grażyna Jarząbek-Bielecka
- Centre for Paediatric, Adolescent Gynaecology and Sexology Division of Gynaecology, Department of Gynaecology, Poznań University of Medical Sciences, Poznań, Poland
- Division of Gynaecology, Department of Gynaecology, Poznań University of Medical Sciences, Poznań, Poland
| | - Witold Kędzia
- Division of Gynaecology, Department of Gynaecology, Poznań University of Medical Sciences, Poznań, Poland
| |
Collapse
|
35
|
Rienzi SCD, Danhof HA, Forshee MD, Roberts A, Britton RA. Limosilactobacillus reuteri promotes the expression and secretion of enteroendocrine- and enterocyte-derived hormones. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610555. [PMID: 39257733 PMCID: PMC11384013 DOI: 10.1101/2024.08.30.610555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Observations that intestinal microbes can beneficially impact host physiology have prompted investigations into the therapeutic usage of such microbes in a range of diseases. For example, the human intestinal microbe Limosilactobacillus reuteri strains ATCC PTA 6475 and DSM 17938 are being considered for use for intestinal ailments including colic, infection, and inflammation as well as non-intestinal ailments including osteoporosis, wound healing, and autism spectrum disorder. While many of their beneficial properties are attributed to suppressing inflammatory responses in the gut, we postulated that L. reuteri may also regulate hormones of the gastrointestinal tract to affect physiology within and outside of the gut. To determine if L. reuteri secreted factors impact the secretion of enteric hormones, we treated an engineered jejunal organoid line, NGN3-HIO, which can be induced to be enriched in enteroendocrine cells, with L. reuteri 6475 or 17938 conditioned medium and performed transcriptomics. Our data suggest that these L. reuteri strains affect the transcription of many gut hormones, including vasopressin and luteinizing hormone subunit beta, which have not been previously recognized as being produced in the gut epithelium. Moreover, we find that these hormones appear to be produced in enterocytes, in contrast to canonical gut hormones which are produced in enteroendocrine cells. Finally, we show that L. reuteri conditioned media promotes the secretion of several enteric hormones including serotonin, GIP, PYY, vasopressin, and luteinizing hormone subunit beta. These results support L. reuteri affecting host physiology through intestinal hormone secretion, thereby expanding our understanding of the mechanistic actions of this microbe.
Collapse
Affiliation(s)
- Sara C. Di Rienzi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Heather A. Danhof
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Micah D. Forshee
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Ari Roberts
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Robert A. Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
36
|
Mączka K, Stasiak O, Przybysz P, Grymowicz M, Smolarczyk R. The Impact of the Endocrine and Immunological Function of Adipose Tissue on Reproduction in Women with Obesity. Int J Mol Sci 2024; 25:9391. [PMID: 39273337 PMCID: PMC11395521 DOI: 10.3390/ijms25179391] [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: 07/15/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Obesity, which leads to metabolic dysregulation and body function impairment, emerges as one of the pressing health challenges worldwide. Excessive body fat deposits comprise a dynamic and biologically active organ possessing its own endocrine function. One of the mechanisms underlying the pathophysiology of obesity is low-grade systemic inflammation mediated by pro-inflammatory factors such as free fatty acids, lipopolysaccharides, adipokines (including leptin, resistin and visfatin) and cytokines (TNF-α, IL-1β, Il-6), which are secreted by adipose tissue. Together with obesity-induced insulin resistance and hyperandrogenism, the exacerbated immune response has a negative impact on the hypothalamic-pituitary-gonadal axis at all levels and directly affects reproduction. In women, it results in disrupted ovarian function, irregular menstrual cycles and anovulation, contributing to infertility. This review focuses on the abnormal intracellular communication, altered gene expression and signaling pathways activated in obesity, underscoring its multifactorial character and consequences at a molecular level. Extensive presentation of the complex interplay between adipokines, cytokines, immune cells and neurons may serve as a foundation for future studies in search of potential sites for more targeted treatment of reproductive disorders related to obesity.
Collapse
Affiliation(s)
- Katarzyna Mączka
- Department of Gynecological Endocrinology, Medical University of Warsaw, 00-315 Warsaw, Poland
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Olga Stasiak
- Department of Gynecological Endocrinology, Medical University of Warsaw, 00-315 Warsaw, Poland
| | - Paulina Przybysz
- Department of Gynecological Endocrinology, Medical University of Warsaw, 00-315 Warsaw, Poland
| | - Monika Grymowicz
- Department of Gynecological Endocrinology, Medical University of Warsaw, 00-315 Warsaw, Poland
| | - Roman Smolarczyk
- Department of Gynecological Endocrinology, Medical University of Warsaw, 00-315 Warsaw, Poland
| |
Collapse
|
37
|
Barbagallo F, Bosoni D, Perone V, Cucinella L, Dealberti D, Cannarella R, Calogero AE, Nappi RE. Gene-environment interaction in functional hypothalamic amenorrhea. Front Endocrinol (Lausanne) 2024; 15:1423898. [PMID: 39268244 PMCID: PMC11390525 DOI: 10.3389/fendo.2024.1423898] [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: 04/26/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Functional hypothalamic amenorrhea (FHA) is a common cause of amenorrhea and chronic anovulation in adolescent girls and young women, diagnosed after excluding other organic causes. It is commonly associated with calorie restriction, excessive physical exercise, and psychosocial stress. These stressors alter the pulsatile secretion of gonadotropin-releasing hormone, leading to a chronic condition of hypoestrogenism and significant health consequences. Recent evidence has highlighted a genetic predisposition to FHA that could explain interindividual variability in stress response. Indeed, not all women experience FHA in response to stress. Rare variants in genes associated with idiopathic hypogonadotropic hypogonadism have been identified in women with FHA, suggesting that these mutations may contribute to an increased susceptibility of women to the trigger of stress exposure. FHA appears today as a complex disease resulting from the combination of genetic predisposition, environmental factors, and epigenetic changes. Furthermore, the genetic background of FHA allows for the hypothesis of a male counterpart. Despite the paucity of data, preliminary findings indicate that an equivalent condition of FHA exists in men, warranting further investigation. This narrative review aims to summarize the recent genetic evidence contributing to the pathophysiology of FHA and to raise awareness on a possible male counterpart.
Collapse
Affiliation(s)
- Federica Barbagallo
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - David Bosoni
- Department of Obstetrics and Gynecology, Azienda Ospedaliera Nazionale SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Valeria Perone
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, IRCCS San Matteo Foundation, Pavia, Italy
| | - Laura Cucinella
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, IRCCS San Matteo Foundation, Pavia, Italy
| | - Davide Dealberti
- Department of Obstetrics and Gynecology, Azienda Ospedaliera Nazionale SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rossella E Nappi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, IRCCS San Matteo Foundation, Pavia, Italy
| |
Collapse
|
38
|
Chen J, Yang X, He Z, Chen W, Zhao Y, Li J, Zong Y, Du R. Cynomorium songaricum Rupr. flavonoids improve cyclophosphamide-induced reproductive function damage by regulating the testosterone synthesis pathway. Front Pharmacol 2024; 15:1457780. [PMID: 39239657 PMCID: PMC11374658 DOI: 10.3389/fphar.2024.1457780] [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: 07/01/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024] Open
Abstract
Introduction The prevalence of male infertility has been increasing globally, necessitating the search for safe and nontoxic active compounds to alleviate reproductive dysfunction. Although the precise mechanism remains unknown, Cynomorium songaricum Rupr. (CS) extract has protective effects on the reproductive system. The effect of C. songaricum Rupr. flavonoids (CSF) on reproductive injury and testicular mesenchymal stem cell viability in male mice and TM3 cells was investigated. Methods We explored the possible association between these effects and the testosterone (T) synthesis pathway. Mice were administered cyclophosphamide to induce reproductive damage, followed by CSF administration. Body mass and organ index were recorded. Pathological changes in T and the epididymis were observed using hematoxylin-eosin staining. ELISA measured the serum levels of T, luteinizing hormone (LH), gonadotropin-releasing hormone (GnRH), follicle-stimulating hormone (FSH), and estradiol (E2) in mice. Fructose and zinc ion levels in the seminal plasma were measured. TM3 cells were treated with Bisphenol A (BPA) and different concentrations of CSF, followed by proliferative evaluations using the CCK-8 assay and T and LH level assessments using ELISA. Furthermore, the expression of steroidogenic enzyme genes and proteins was investigated using western blotting and RT-PCR. Results CSF exhibited a notable reduction in reproductive damage and improved pathological changes in testicular and epididymal tissues. CSF group demonstrated substantially higher levels of seminal plasma fructose and zinc ions; markedly elevated serum levels of T, LH, GnRH, and FSH; and lower levels of E2 than those of the model group. Intracellular T content and secretion of T and LH increase with CSF while effectively mitigating BPA-induced damage to TM3 cells. CSF group exhibited substantially higher gene and protein expression of steroidogenic enzymes than those of the model group, both in vivo and in vitro. CSF ameliorates reproductive impairment by enhancing the expression of pivotal enzymes involved in synthesizing T. Discussion CSF ameliorates cyclophosphamide-induced reproductive impairment and bisphenol A-induced TM3 cell damage in mice by regulating sex hormone levels in the Hypothalamic-Pituitary-Gonadal Axis (HPG axis) and upregulating the expression of steroidogenic enzymes. Therefore, CS is a potential treatment for male reproductive impairment.
Collapse
Affiliation(s)
- Jiarong Chen
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Xiaoyue Yang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Zhongmei He
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer, Jilin Agricultural University, Changchun, China
| | - Weijia Chen
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer, Jilin Agricultural University, Changchun, China
| | - Yan Zhao
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer, Jilin Agricultural University, Changchun, China
| | - Jianming Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer, Jilin Agricultural University, Changchun, China
| | - Ying Zong
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer, Jilin Agricultural University, Changchun, China
| | - Rui Du
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality and Safety, Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
39
|
Wang D, Tan L, Zhi Y, Bu L, Wang Y, Wang Z, Guo Y, Tian W, Xu C, Li D, Li Z, Jiang R, Han R, Li G, Wang Y, Xia D, Tian Y, Dunn IC, Hu X, Li H, Zhao Y, Kang X, Liu X. Genome-wide variation study and inter-tissue communication analysis unveil regulatory mechanisms of egg-laying performance in chickens. Nat Commun 2024; 15:7069. [PMID: 39152103 PMCID: PMC11329784 DOI: 10.1038/s41467-024-50809-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 07/22/2024] [Indexed: 08/19/2024] Open
Abstract
Egg-laying performance is of great economic importance in poultry, but the underlying genetic mechanisms are still elusive. In this work, we conduct a multi-omics and multi-tissue integrative study in hens with distinct egg production, to detect the hub candidate genes and construct hub molecular networks contributing to egg-laying phenotypic differences. We identifiy three hub candidate genes as egg-laying facilitators: TFPI2, which promotes the GnRH secretion in hypothalamic neuron cells; CAMK2D, which promotes the FSHβ and LHβ secretion in pituitary cells; and OSTN, which promotes granulosa cell proliferation and the synthesis of sex steroid hormones. We reveal key endocrine factors involving egg production by inter-tissue crosstalk analysis, and demonstrate that both a hepatokine, APOA4, and an adipokine, ANGPTL2, could increase egg production by inter-tissue communication with hypothalamic-pituitary-ovarian axis. Together, These results reveal the molecular mechanisms of multi-tissue coordinative regulation of chicken egg-laying performance and provide key insights to avian reproductive regulation.
Collapse
Affiliation(s)
- Dandan Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Lizhi Tan
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yihao Zhi
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Lina Bu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yangyang Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Zhang Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Yulong Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Weihua Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Chunlin Xu
- Henan Sangao Agriculture and Animal Husbandry Co, Ltd, Gushi, China
| | - Donghua Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, China
| | - Zhuanjian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, China
| | - Ruirui Jiang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, China
| | - Ruili Han
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, China
| | - Guoxi Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, China
| | - Yongqiang Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Dong Xia
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, London, UK
| | - Yadong Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, China
| | - Ian C Dunn
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Xiaoxiang Hu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Hong Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
- International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, China.
| | - Yiqiang Zhao
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China.
| | - Xiangtao Kang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
- International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, China.
| | - Xiaojun Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
- International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, China.
| |
Collapse
|
40
|
Abdalqader MM, Hussein SS, Jadi HF, Nori W. Kisspeptin-10: A Predictor for Fetal Growth Restriction among Preeclamptic Women that Discriminated Early Onset Cases. CLIN EXP OBSTET GYN 2024; 51. [DOI: 10.31083/j.ceog5108177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Background: Preeclampsia (PE) is a major cause of maternal and neonatal morbidity. Fetal growth restriction (FGR) shares many pathophysiological roles with PE. Kisspeptin-10 is a peptide secreted by placental syncytium. It was linked to many adverse pregnancy events. The current study aimed to examine Kisspeptin’s-10 role in predicting FGR in PE pregnancies and to verify whether it can predict its onset as early or late FGR. Methods: An observational case-control study enrolled 120 eligible cases at matched gestational age (28–40 weeks) and body mass index (BMI); they were divided into 2-groups: (60) healthy controls and (60) PE cases. PE cases were subdivided into early onset FGR (28/60), who had a gestational age less than 34 weeks, and late-onset FGR (32/60) with a gestational age equal to 34 weeks. A collection was made of the following data: first: pregnant primary criteria [age, BMI, systolic and diastolic blood pressure (BP), and urine for albumin], second: serum Kisspetein-10 was evaluated via enzyme-linked immunosorbent assay (ELISA), and third: ultrasonic criteria [estimated fetal weight, resistance, and pulsatility index (RI, PI)] were recorded for all. Results: Serum Kisspeptin-10 was significantly higher among the controls (309.56 ± 67.72) followed by late-onset FGR and early onset FGR (235.46 ± 68.97) vs. (212.09 ± 58.44) ng/dL; p = 0.0001 respectively. It was negatively linked to systolic, diastolic BP, and urine for albumin; Pearson correlation coefficient (r) was (–0.29, –0.48, –0.28) respectively; p < 0.0001, 0.0018, 0.028 respectively. Kisspeptin-10 was positively linked to estimated fetal weight (r = 0.27; p = 0.034); it had an odds ratio (OR) of 3.04; 95% confidence interval of (1.37–4.765); p = 0.0001 in discriminating healthy pregnancies from FGR cases. Conclusions: The significant correlation of Kisspeptin-10 with PE parameters and estimated fetal weight with high sensitivity, specificity and reliable area under the curve in predicting early onset FGR cases make it recommended for practice in predicting FGR onset.
Collapse
Affiliation(s)
- Manal Madany Abdalqader
- Department of Obstetrics and Gynecology, College of Medicine, Mustansiriyah University, 10052 Baghdad, Iraq
| | - Shatha Sami Hussein
- Department of Obstetrics and Gynecology, College of Medicine, Mustansiriyah University, 10052 Baghdad, Iraq
| | - Huda Fadhil Jadi
- Department of Obstetrics and Gynecology, Fatima Al Zahraa Maternity Hospital, 10052 Baghdad, Iraq
| | - Wassan Nori
- Department of Obstetrics and Gynecology, College of Medicine, Mustansiriyah University, 10052 Baghdad, Iraq
| |
Collapse
|
41
|
Singh A, Lal B, Kumar P, Parhar IS, Millar RP. Nitric oxide mediated kisspeptin regulation of steroidogenesis and gametogenesis in the catfish, Clarias batrachus. Cell Tissue Res 2024; 397:111-124. [PMID: 38829397 DOI: 10.1007/s00441-024-03899-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 05/22/2024] [Indexed: 06/05/2024]
Abstract
Nitric oxide (NO) is a gaseous molecule that regulates various reproductive functions. It is a well-recognized regulator of GnRH-FSH/LH-sex steroid secretion in vertebrates including fish. Kisspeptin is a recently discovered neuropeptide which also regulates GnRH secretion. Nitrergic and kisspeptin neurons are reported in close physical contact in the mammalian brain suggesting their interactive role in the release of GnRH. The existence of kisspeptin and NOS is also demonstrated in vertebrate gonads, but information on their reciprocal relation in gonads, if any, is obscure. Therefore, attempts were made to evaluate the functional reciprocal relation between nitric oxide and kisspeptin in the catfish gonads, if any, by administering the nitric oxide synthase (NOS) inhibitor, L-NAME {N(G)-nitro-L-arginine methyl ester}, which reduces NO production, and kisspeptin agonist (KP-10) and assessing their impacts on the expressions of kisspeptin1, different NOS isoforms, NO and steroid production in the gonadal tissue. The results revealed that L-NAME suppressed the expression of kiss1 in gonads of the catfish establishing the role of NO in kisspeptin expression. However, KP-10 increased the expression of all the isoforms of NOSs (iNOS, eNOS, nNOS) and concurrently NO and steroids in the ovary and testis. In vitro studies also indicate that kisspeptin stimulates the production of NO and estradiol and testosterone levels in the gonadal explants and medium. Thus, in vivo results clearly suggest a reciprocal interaction between kisspeptin and NO to regulate the gonadal activity of the catfish. The in vitro findings further substantiate our contention regarding the interactive role of kisspeptin and NO in gonadal steroidogenesis.
Collapse
Affiliation(s)
- Ankur Singh
- Fish Endocrinology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Bechan Lal
- Fish Endocrinology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| | - Pankaj Kumar
- Department of Zoology, University of Jammu, Jammu, UT of Jammu and Kashmir, India
| | - Ishwar S Parhar
- Center Initiative for Training International Researches, University of Toyama, Toyama, Japan
| | - Robert P Millar
- Centre for Neuroendocrinology, Department of Immunology, University of Pretoria, Pretoria, South Africa
- Department of Integrative Biomedical Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
42
|
Yi Y, Feng Y, Shi Y, Xiao J, Liu M, Wang K. Per- and Polyfluoroalkyl Substances (PFASs) and Their Potential Effects on Female Reproductive Diseases. TOXICS 2024; 12:539. [PMID: 39195641 PMCID: PMC11358978 DOI: 10.3390/toxics12080539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Per- and polyfluoroalkyl substances (PFASs) are a class of anthropogenic organic compounds widely present in the natural and human living environments. These emerging persistent pollutants can enter the human body through multiple channels, posing risks to human health. In particular, exposure to PFASs in women may cause a series of reproductive health hazards and infertility. Based on a review of the existing literature, this study preliminarily summarizes the effects of PFAS exposure on the occurrence and development of female reproductive endocrine diseases, such as polycystic ovary syndrome (PCOS), endometriosis, primary ovarian insufficiency (POI), and diminished ovarian reserve (DOR). Furthermore, we outline the relevant mechanisms through which PFASs interfere with the physiological function of the female ovary and finally highlight the role played by nutrients in reducing the reproductive health hazards caused by PFASs. It is worth noting that the physiological mechanisms of PFASs in the above diseases are still unclear. Therefore, it is necessary to further study the molecular mechanisms of PFASs in female reproductive diseases and the role of nutrients in this process.
Collapse
Affiliation(s)
- Yuqing Yi
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China; (Y.Y.); (Y.F.); (Y.S.); (J.X.); (M.L.)
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Yang Feng
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China; (Y.Y.); (Y.F.); (Y.S.); (J.X.); (M.L.)
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Yuechen Shi
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China; (Y.Y.); (Y.F.); (Y.S.); (J.X.); (M.L.)
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Jiaming Xiao
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China; (Y.Y.); (Y.F.); (Y.S.); (J.X.); (M.L.)
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Ming Liu
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China; (Y.Y.); (Y.F.); (Y.S.); (J.X.); (M.L.)
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Ke Wang
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China; (Y.Y.); (Y.F.); (Y.S.); (J.X.); (M.L.)
| |
Collapse
|
43
|
Shen S, Wang D, Liu H, He X, Cao Y, Chen J, Li S, Cheng X, Xu HE, Duan J. Structural basis for hormone recognition and distinctive Gq protein coupling by the kisspeptin receptor. Cell Rep 2024; 43:114389. [PMID: 38935498 DOI: 10.1016/j.celrep.2024.114389] [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: 12/13/2023] [Revised: 04/25/2024] [Accepted: 06/06/2024] [Indexed: 06/29/2024] Open
Abstract
Kisspeptin signaling through its G protein-coupled receptor, KISS1R, plays an indispensable role in regulating reproduction via the hypothalamic-pituitary-gonadal axis. Dysregulation of this pathway underlies severe disorders like infertility and precocious puberty. Here, we present cryo-EM structures of KISS1R bound to the endogenous agonist kisspeptin-10 and a synthetic analog TAK-448. These structures reveal pivotal interactions between peptide ligands and KISS1R extracellular loops for receptor activation. Both peptides exhibit a conserved binding mode, unveiling their common activation mechanism. Intriguingly, KISS1R displays a distinct 40° angular deviation in its intracellular TM6 region compared to other Gq-coupled receptors, enabling distinct interactions with Gq. This study reveals the molecular intricacies governing ligand binding and activation of KISS1R, while highlighting its exceptional ability to couple with Gq. Our findings pave the way for structure-guided design of therapeutics targeting this physiologically indispensable receptor.
Collapse
Affiliation(s)
- Shiyi Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dongxue Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Heng Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xinheng He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yinglong Cao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Juanhua Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shujie Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xi Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - H Eric Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Jia Duan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
44
|
Zhang A, Li S, Huang L, Jiang Y, Chen Y, Zhu S, Xiong F, Luo Z, Ou M, Ying J, Wang S, Mu D, Qu Y. Bmal1 regulates female reproduction in mice via the hypothalamic-pituitary-ovarian axis. FASEB J 2024; 38:e23744. [PMID: 38885031 DOI: 10.1096/fj.202400391r] [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: 02/21/2024] [Revised: 05/22/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024]
Abstract
The hypothalamic-pituitary-gonadal axis (HPG) is the key neuroendocrine axis involved in reproductive regulation. Brain and muscle ARNT-like protein 1 (Bmal1) participates in regulating the metabolism of various endocrine hormones. However, the regulation of Bmal1 on HPG and female fertility is unclear. This study aims to explore the regulation of female reproduction by Bmal1 via the HPG axis in mice. Bmal1-knockout (Ko) mice were generated using the CRISPR/Cas9 technology. The structure, function, and estrous cycle of ovarian in Bmal1 Ko female mice were measured. The key genes and proteins of the HPG axis involved in regulating female reproduction were examined through transcriptome analysis and then verified by RT-PCR, immunohistochemistry, and western blot. Furthermore, the fertility of female mice was detected after intervening prolactin (PRL) and progesterone (Pg) in Bmal1 ko mice. The number of offspring and ovarian weight were significantly lower in Bmal1-Ko mice than in wild-type (Wt) mice. In Bmal1-Ko mice, ovarian cells were arranged loosely and irregularly, and the total number of follicles was significantly reduced. No corpus luteum was found in the ovaries. Vaginal smears revealed that Bmal1-Ko mice had an irregular estrus cycle. In Bmal1-Ko mice, Star expression was decreased, PRL and luteinizing hormone (LH) levels were increased, and dopamine (DA) and Pg levels were decreased. Inhibition of PRL partially recovered the estrous cycle, corpus luteum formation, and Star expression in the ovaries. Pg supplementation promoted embryo implantation in Bmal1-Ko female mice. Bmal1 Ko increases serum PRL levels in female mice likely by reducing DA levels, thus affecting luteal formation, resulting in decreased Star expression and Pg production, hindering female reproduction. Inhibition of PRL or restoration of Pg can partially restore reproductive capacity in female Bmal1-Ko mice. Thus, Bmal1 may regulate female reproduction via the HPG axis in mice, suggesting that Bmal1 is a potential target to treat female infertility.
Collapse
Affiliation(s)
- Ayuan Zhang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Pediatric Genetic Metabolism Endocrinology, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, China
| | - Shiping Li
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lingyi Huang
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China College of Stomatology, Sichuan University, Chengdu, China
| | - Yin Jiang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yan Chen
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shuyao Zhu
- Department of Pediatric Genetic Metabolism Endocrinology, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, China
| | - Fu Xiong
- Department of Pediatric Genetic Metabolism Endocrinology, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, China
| | - Zemin Luo
- Department of Pediatric Genetic Metabolism Endocrinology, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, China
| | - Mingcai Ou
- Department of Pediatric Genetic Metabolism Endocrinology, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shaopu Wang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
45
|
Mineiro R, Cardoso MR, Pinheiro JV, Cipolla-Neto J, do Amaral FG, Quintela T. Overlapping action of melatonin and female reproductive hormones-Understand the impact in pregnancy and menopause. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 142:163-190. [PMID: 39059985 DOI: 10.1016/bs.apcsb.2024.06.005] [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: 07/28/2024]
Abstract
Melatonin is an indolamine secreted to circulation by the pineal gland according to a circadian rhythm. Melatonin levels are higher during nighttime, and the principal function of this hormone is to organize the temporal night and day distribution of physiological adaptive processes. Besides hormonal pineal production, melatonin is synthesized in various organs and tissues like the ovaries or the placenta for local utilization. In addition to its function as a circadian messenger, melatonin is also associated with many physiological functions. For example, melatonin has antioxidant properties and is involved in the regulation of energy and bone metabolism, and reproduction. Melatonin impacts several stages of reproduction and the action across the hypothalamus-pituitary-gonadal axis is well described. However, it is not well understood how those actions impact the female reproductive hormones secretion nor the consequent physiological outcomes. Thus, the first part of this chapter describes the regulation of female reproductive hormone synthesis by melatonin. Moreover, melatonin and female reproductive hormones have coincident physiological functions. Life stages like pregnancy or menopause are characterized by alterations in the reproductive hormones secretion that may be associated with certain physiological stages. Therefore, the second part discusses whether melatonin fluctuations could have an overlapping role with reproductive hormones in contributing to clinical outcomes associated with pregnancy and menopause.
Collapse
Affiliation(s)
- Rafael Mineiro
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| | | | - João Vieira Pinheiro
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| | - José Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Telma Quintela
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal; Instituto Politécnico da Guarda, Guarda, Portugal.
| |
Collapse
|
46
|
Kovács A, Szabó E, László K, Kertes E, Zagorácz O, Mintál K, Tóth A, Gálosi R, Berta B, Lénárd L, Hormay E, László B, Zelena D, Tóth ZE. Brain RFamide Neuropeptides in Stress-Related Psychopathologies. Cells 2024; 13:1097. [PMID: 38994950 PMCID: PMC11240450 DOI: 10.3390/cells13131097] [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: 04/29/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/13/2024] Open
Abstract
The RFamide peptide family is a group of proteins that share a common C-terminal arginine-phenylalanine-amide motif. To date, the family comprises five groups in mammals: neuropeptide FF, LPXRFamides/RFamide-related peptides, prolactin releasing peptide, QRFP, and kisspeptins. Different RFamide peptides have their own cognate receptors and are produced by different cell populations, although they all can also bind to neuropeptide FF receptors with different affinities. RFamide peptides function in the brain as neuropeptides regulating key aspects of homeostasis such as energy balance, reproduction, and cardiovascular function. Furthermore, they are involved in the organization of the stress response including modulation of pain. Considering the interaction between stress and various parameters of homeostasis, the role of RFamide peptides may be critical in the development of stress-related neuropathologies. This review will therefore focus on the role of RFamide peptides as possible key hubs in stress and stress-related psychopathologies. The neurotransmitter coexpression profile of RFamide-producing cells is also discussed, highlighting its potential functional significance. The development of novel pharmaceutical agents for the treatment of stress-related disorders is an ongoing need. Thus, the importance of RFamide research is underlined by the emergence of peptidergic and G-protein coupled receptor-based therapeutic targets in the pharmaceutical industry.
Collapse
Affiliation(s)
- Anita Kovács
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Evelin Szabó
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Kristóf László
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Erika Kertes
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Olga Zagorácz
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Kitti Mintál
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Attila Tóth
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Rita Gálosi
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Bea Berta
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - László Lénárd
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Edina Hormay
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Bettina László
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Dóra Zelena
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Zsuzsanna E. Tóth
- Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary
| |
Collapse
|
47
|
Ruiz A, Ramos L. Genetic Variants in KNDy Pathway Lack Association with Premature Ovarian Insufficiency in Mexican Women: A Sequencing-Based Cohort Study. Genes (Basel) 2024; 15:788. [PMID: 38927724 PMCID: PMC11203232 DOI: 10.3390/genes15060788] [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: 05/21/2024] [Revised: 06/12/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Previous studies have demonstrated the essential role of the Kisspeptin/Neurokinin B/Dynorphin A (KNDy) pathway in female reproductive biology by regulating the activity of the hypothalamic-pituitary-gonadal axis. Identified loss-of-function mutations in these genes are linked to various reproductive disorders. This study investigated genetic disorders linked to mutations in the KNDy genes related to premature ovarian insufficiency (POI). A cohort of 14 Mexican POI patients underwent genetic screening using PCR-SSCP and Sanger sequencing, assessing the genetic variations' impact on protein function thereafter using multiple in silico tools. The PCR excluded extensive deletions, insertions, and duplications, while SSCP detected five genetic variants. Variations occurred in the KISS1 (c.58G>A and c.242C>G), KISS1R (c.1091A>T), PDYN (c.600C>T), and OPRK1 (c.36G>T) genes, whereas no genetic anomalies were found in NK3/NK3R genes. Each single-nucleotide variant underwent genotyping using PCR-SSCP in 100 POI-free subjects. Their allelic frequencies paralleled the patient group. These observations indicate that allelic variations in the KNDy genes may not contribute to POI etiology. Hence, screening for mutations in KNDy genes should not be a part of the diagnostic protocol for POI.
Collapse
Affiliation(s)
| | - Luis Ramos
- Department of Reproductive Biology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga #15, Tlalpan, México City C.P. 14080, Mexico
| |
Collapse
|
48
|
Chen X, Feng Y, Dai S, Guo B, Yan L, Liu J, Zhu H. Advances in research on spexin-mediated regulation of reproductive function in vertebrates. Front Endocrinol (Lausanne) 2024; 15:1422711. [PMID: 38915898 PMCID: PMC11194384 DOI: 10.3389/fendo.2024.1422711] [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: 04/24/2024] [Accepted: 05/30/2024] [Indexed: 06/26/2024] Open
Abstract
Spexin (SPX, NPQ) is a 14-amino acid neuroactive peptide identified using bioinformatics. This amino acid sequence of the mature spexin peptide has been highly conserved during species evolution and is widely distributed in the central nervous system and peripheral tissues and organs. Therefore, spexin may play a role in various biological functions. Spexin, the cognate ligand for GALR2/3, acting as a neuromodulator or endocrine signaling factor, can inhibit reproductive performance. However, controversies and gaps in knowledge persist regarding spexin-mediated regulation of animal reproductive functions. This review focuses on the hypothalamic-pituitary-gonadal axis and provides a comprehensive overview of the impact of spexin on reproduction. Through this review, we aim to enhance understanding and obtain in-depth insights into the regulation of reproduction by spexin peptides, thereby providing a scientific basis for future investigations into the molecular mechanisms underlying the influence of spexin on reproductive function. Such investigations hold potential benefits for optimizing farming practices in livestock, poultry, and fish industries.
Collapse
Affiliation(s)
- Xiaojing Chen
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yuyan Feng
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Shudi Dai
- School of Life Science, Jiangsu University, Zhenjiang, China
| | - Binbin Guo
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Jiangsu Province Engineering Research Center of Precision Animal Breeding, Nanjing, China
| | - Leyan Yan
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Jiangsu Province Engineering Research Center of Precision Animal Breeding, Nanjing, China
| | - Jie Liu
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Jiangsu Province Engineering Research Center of Precision Animal Breeding, Nanjing, China
| | - Huanxi Zhu
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Jiangsu Province Engineering Research Center of Precision Animal Breeding, Nanjing, China
| |
Collapse
|
49
|
Jiang J, Shu Z, Qiu L. Adverse effects and potential mechanisms of polystyrene microplastics (PS-MPs) on the blood-testis barrier. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2024; 46:238. [PMID: 38849627 DOI: 10.1007/s10653-024-02033-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/13/2024] [Indexed: 06/09/2024]
Abstract
Microplastics (MPs) are defined as plastic particles or fragments with a diameter of less than 5 mm. These particles have been identified as causing male reproductive toxicity, although the precise mechanism behind this association is yet to be fully understood. Recent research has found that exposure to polystyrene microplastics (PS-MPs) can disrupt spermatogenesis by impacting the integrity of the blood-testis barrier (BTB), a formidable barrier within mammalian blood tissues. The BTB safeguards germ cells from harmful substances and infiltration by immune cells. However, the disruption of the BTB leads to the entry of environmental pollutants and immune cells into the seminiferous tubules, resulting in adverse reproductive effects. Additionally, PS-MPs induce reproductive damage by generating oxidative stress, inflammation, autophagy, and alterations in the composition of intestinal flora. Despite these findings, the precise mechanism by which PS-MPs disrupt the BTB remains inconclusive, necessitating further investigation into the underlying processes. This review aims to enhance our understanding of the pernicious effects of PS-MP exposure on the BTB and explore potential mechanisms to offer novel perspectives on BTB damage caused by PS-MPs.
Collapse
Affiliation(s)
- Jinchen Jiang
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, People's Republic of China
| | - Zhenhao Shu
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, People's Republic of China
| | - Lianglin Qiu
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, People's Republic of China.
| |
Collapse
|
50
|
Ahmad I, Kaur M, Tyagi D, Singh TB, Kaur G, Afzal SM, Jauhar M. Exploring novel insights into the molecular mechanisms underlying Bisphenol A-induced toxicity: A persistent threat to human health. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 108:104467. [PMID: 38763439 DOI: 10.1016/j.etap.2024.104467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/09/2024] [Accepted: 05/11/2024] [Indexed: 05/21/2024]
Abstract
Bisphenol A (BPA) is a ubiquitous industrial chemical used in the production of polycarbonate plastics and epoxy resins, found in numerous consumer products. Despite its widespread use, its potential adverse health effects have raised significant concerns. This review explores the molecular mechanisms and evidence-based literature underlying BPA-induced toxicities and its implications for human health. BPA is an endocrine-disrupting chemical (EDC) which exhibits carcinogenic properties by influencing various receptors, such as ER, AhR, PPARs, LXRs, and RARs. It induces oxidative stress and contributes to cellular dysfunction, inflammation, and DNA damage, ultimately leading to various toxicities including but not limited to reproductive, cardiotoxicity, neurotoxicity, and endocrine toxicity. Moreover, BPA can modify DNA methylation patterns, histone modifications, and non-coding RNA expression, leading to epigenetic changes and contribute to carcinogenesis. Overall, understanding molecular mechanisms of BPA-induced toxicity is crucial for developing effective strategies and policies to mitigate its adverse effects on human health.
Collapse
Affiliation(s)
- Israel Ahmad
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road, Phagwara, Punjab, India.
| | - Mandeep Kaur
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road, Phagwara, Punjab, India.
| | - Devansh Tyagi
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road, Phagwara, Punjab, India.
| | - Tejinder Bir Singh
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road, Phagwara, Punjab, India.
| | - Gurpreet Kaur
- School of Business Studies, Punjab Agricultural University, Ludhiana, Punjab, India.
| | - Shaikh Mohammad Afzal
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road, Phagwara, Punjab, India.
| | - Mohsin Jauhar
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road, Phagwara, Punjab, India.
| |
Collapse
|