1
|
Hornberger T, Reese S, Perbandt K, Meyer-Lindenberg A, Walter B. Correlation of Anti-Müllerian hormone serum concentration measured in proestrus and estrus with the litter size as a fertility marker in bitches. Domest Anim Endocrinol 2024; 91:106897. [PMID: 39647379 DOI: 10.1016/j.domaniend.2024.106897] [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: 05/21/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 12/10/2024]
Abstract
The use of the anti-Müllerian hormone (AMH) serum concentration as a fertility marker has been shown in cows, sheep and mares and has been indicated in one study in female dogs. The aim of this study was to investigate the connection between the AMH serum concentration, taken at two defined time points during the bitch`s heat, and the litter size to investigate whether AMH can be used as a practical measurement for the individual breeding bitch to predict litter size. The study was carried out on 27 healthy female dogs presented for pre-breeding examination, considering all previously known influencing factors on AMH in the bitch such as age, body weight and estrous cycle phase at the time the sample was taken. Due to the AMH increase in early proestrus and its drop around ovulation, AMH was measured in blood samples taken within the first three days of heat (AMH1) and near ovulation (AMH2) with AMH1 being significantly higher than AMH 2 (p < 0.001). There was a highly significant negative correlation of body weight and AMH at both sampling times (p < 0.001). There were no significant results when the dogs were paired and grouped according to high or low AMH concentrations, but a significant effect of AMH serum concentration on litter size was found in the multifactorial analysis when the dogs were matched according to their body weight (AMH1: p = 0.022; AMH2: p = 0.030). In conclusion, a significant effect of the AMH concentration and the litter size among female dogs with matching body weight could be found, but a much larger sample collection is needed to evaluate reference intervals for AMH for bitches of different weight to predict the fertility of an individual bitch in the future.
Collapse
Affiliation(s)
- Theresa Hornberger
- Clinic of Small Animal Surgery and Reproduction at the Centre for Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Ludwig-Maximilians-University, Veterinaerstr. 13, 80539 Munich, Germany.
| | - Sven Reese
- Chair of Anatomy, Histology and Embryology, Department of Veterinary Sciences, Faculty of Veterinary Medicine, Ludwig-Maximilians-University, Veterinaerstr. 13, 80539 Munich, Germany
| | - Klaus Perbandt
- Freelance Mathematician, Rotenhöfer Weg 4, 24768 Rendsburg, Germany
| | - Andrea Meyer-Lindenberg
- Clinic of Small Animal Surgery and Reproduction at the Centre for Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Ludwig-Maximilians-University, Veterinaerstr. 13, 80539 Munich, Germany
| | - Beate Walter
- Clinic of Small Animal Surgery and Reproduction at the Centre for Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Ludwig-Maximilians-University, Veterinaerstr. 13, 80539 Munich, Germany.
| |
Collapse
|
2
|
Li L, Lin W, Wang Z, Huang R, Xia H, Li Z, Deng J, Ye T, Huang Y, Yang Y. Hormone Regulation in Testicular Development and Function. Int J Mol Sci 2024; 25:5805. [PMID: 38891991 PMCID: PMC11172568 DOI: 10.3390/ijms25115805] [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: 04/07/2024] [Revised: 05/01/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
The testes serve as the primary source of androgens and the site of spermatogenesis, with their development and function governed by hormonal actions via endocrine and paracrine pathways. Male fertility hinges on the availability of testosterone, a cornerstone of spermatogenesis, while follicle-stimulating hormone (FSH) signaling is indispensable for the proliferation, differentiation, and proper functioning of Sertoli and germ cells. This review covers the research on how androgens, FSH, and other hormones support processes crucial for male fertility in the testis and reproductive tract. These hormones are regulated by the hypothalamic-pituitary-gonad (HPG) axis, which is either quiescent or activated at different stages of the life course, and the regulation of the axis is crucial for the development and normal function of the male reproductive system. Hormonal imbalances, whether due to genetic predispositions or environmental influences, leading to hypogonadism or hypergonadism, can precipitate reproductive disorders. Investigating the regulatory network and molecular mechanisms involved in testicular development and spermatogenesis is instrumental in developing new therapeutic methods, drugs, and male hormonal contraceptives.
Collapse
Affiliation(s)
- Lu Li
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Wanqing Lin
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Zhaoyang Wang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Rufei Huang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Huan Xia
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Ziyi Li
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Jingxian Deng
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Tao Ye
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Yadong Huang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China
| | - Yan Yang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China
| |
Collapse
|
3
|
Rey RA, Grinspon RP. Anti-Müllerian hormone, testicular descent and cryptorchidism. Front Endocrinol (Lausanne) 2024; 15:1361032. [PMID: 38501100 PMCID: PMC10944898 DOI: 10.3389/fendo.2024.1361032] [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: 12/24/2023] [Accepted: 02/22/2024] [Indexed: 03/20/2024] Open
Abstract
Anti-Müllerian hormone (AMH) is a Sertoli cell-secreted glycoprotein involved in male fetal sex differentiation: it provokes the regression of Müllerian ducts, which otherwise give rise to the Fallopian tubes, the uterus and the upper part of the vagina. In the first trimester of fetal life, AMH is expressed independently of gonadotropins, whereas from the second trimester onwards AMH testicular production is stimulated by FSH and oestrogens; at puberty, AMH expression is inhibited by androgens. AMH has also been suggested to participate in testicular descent during fetal life, but its role remains unclear. Serum AMH is a well-recognized biomarker of testicular function from birth to the first stages of puberty. Especially in boys with nonpalpable gonads, serum AMH is the most useful marker of the existence of testicular tissue. In boys with cryptorchidism, serum AMH levels reflect the mass of functional Sertoli cells: they are lower in patients with bilateral than in those with unilateral cryptorchidism. Interestingly, serum AMH increases after testis relocation to the scrotum, suggesting that the ectopic position result in testicular dysfunction, which may be at least partially reversible. In boys with cryptorchidism associated with micropenis, low AMH and FSH are indicative of central hypogonadism, and serum AMH is a good marker of effective FSH treatment. In patients with cryptorchidism in the context of disorders of sex development, low serum AMH is suggestive of gonadal dysgenesis, whereas normal or high AMH is found in patients with isolated androgen synthesis defects or with androgen insensitivity. In syndromic disorders, assessment of serum AMH has shown that Sertoli cell function is preserved in boys with Klinefelter syndrome until mid-puberty, while it is affected in patients with Noonan, Prader-Willi or Down syndromes.
Collapse
Affiliation(s)
- Rodolfo A. Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular, Histología, Embriología y Genética, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas, Santa Fe, Argentina
| | - Romina P. Grinspon
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| |
Collapse
|
4
|
Prapaiwan N, Manee-In S, Thanawongnuwech R, Srisuwatanasagul S. Anti-Müllerian hormone levels in serum and testes of male dogs: relations with neuter status and bilateral abdominal cryptorchidism. Theriogenology 2023; 208:171-177. [PMID: 37352559 DOI: 10.1016/j.theriogenology.2023.06.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/11/2023] [Accepted: 06/11/2023] [Indexed: 06/25/2023]
Abstract
Anti-Müllerian hormone (AMH) analysis has contributed to new information in the reproductive endocrinology of domestic animals, due to clinically available diagnostic tools. An accurate and rapid diagnostic method to distinguish between neutered and bilateral abdominal cryptorchid dogs is needed in veterinary practice. Therefore, this study uses an enzyme-linked immunosorbent assay to evaluate the clinical relevance of AMH analysis in peripheral blood as a diagnostic tool for dogs with suspected bilateral abdominal cryptorchidism. The possible alteration of the AMH localization in testicular tissue caused by this pathologic condition was also investigated using immunohistochemistry. Male dogs were divided into three groups of healthy intact (n = 14), healthy castrated (n = 14), and bilateral abdominal cryptorchid (n = 14) dogs. The results demonstrated a higher level of serum AMH in the cryptorchid group compared to the intact group (P < 0.01), while serum AMH levels of all castrated dogs were below the limit of detection (<0.05 ng/mL). Moreover, the percentage of positive AMH immunostaining of the intact group was less than that of the cryptorchid group (P < 0.01). A significantly positive correlation was found between serum AMH concentration and localization in testicular tissues (r = 0.93, P < 0.01). Our findings suggest that AMH levels in the peripheral blood could be used as an alternative and rapid screening method for detecting dogs with abdominal cryptorchidism.
Collapse
Affiliation(s)
- N Prapaiwan
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - S Manee-In
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - R Thanawongnuwech
- Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - S Srisuwatanasagul
- Department of Anatomy, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
5
|
Holterhus PM, Kulle A, Busch H, Spielmann M. Classic genetic and hormonal switches during fetal sex development and beyond. MED GENET-BERLIN 2023; 35:163-171. [PMID: 38840820 PMCID: PMC10842585 DOI: 10.1515/medgen-2023-2036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Critical genetic and hormonal switches characterize fetal sex development in humans. They are decisive for gonadal sex determination and subsequent differentiation of the genital and somatic sex phenotype. Only at the first glace these switches seem to behave like the dual 0 and 1 system in computer sciences and lead invariably to either typically male or female phenotypes. More recent data indicate that this model is insufficient. In addition, in case of distinct mutations, many of these switches may act variably, causing a functional continuum of alterations of gene functions and -dosages, enzymatic activities, sex hormone levels, and sex hormone sensitivity, giving rise to a broad clinical spectrum of biological differences of sex development (DSD) and potentially diversity of genital and somatic sex phenotypes. The gonadal anlage is initially a bipotential organ that can develop either into a testis or an ovary. Sex-determining region Y (SRY) is the most important upstream switch of gonadal sex determination inducing SOX9 further downstream, leading to testicular Sertoli cell differentiation and the repression of ovarian pathways. If SRY is absent (virtually "switched off"), e. g., in 46,XX females, RSPO1, WNT4, FOXL2, and other factors repress the male pathway and promote ovarian development. Testosterone and its more potent derivative, dihydrotestosterone (DHT) as well as AMH, are the most important upstream hormonal switches in phenotypic sex differentiation. Masculinization of the genitalia, i. e., external genital midline fusion forming the scrotum, growth of the genital tubercle, and Wolffian duct development, occurs in response to testosterone synthesized by steroidogenic cells in the testis. Müllerian ducts will not develop into a uterus and fallopian tubes in males due to Anti-Müllerian-Hormone (AMH) produced by the Sertoli cells. The functionality of these two hormone-dependent switches is ensured by their corresponding receptors, the intracellular androgen receptor (AR) and the transmembrane AMH type II receptor. The absence of high testosterone and high AMH is crucial for anatomically female genital development during fetal life. Recent technological advances, including single-cell and spatial transcriptomics, will likely shed more light on the nature of these molecular switches.
Collapse
Affiliation(s)
- Paul-Martin Holterhus
- Christian-Albrechts University of Kiel (CAU)Pediatric Endocrinology and Diabetes, Department of Pediatrics IKielGermany
| | - Alexandra Kulle
- Christian-Albrechts University of Kiel (CAU)Pediatric Endocrinology and Diabetes, Department of Pediatrics IKielGermany
| | - Hauke Busch
- University of LübeckMedical Systems Biology Group, Lübeck Institute of Experimental Dermatology (LIED)Ratzeburger Allee 16023562LübeckGermany
| | - Malte Spielmann
- University of LübeckInstitute of Human GeneticsLübeckGermany
| |
Collapse
|
6
|
Rey RA. Steroid receptors in the testis: implications in the physiology of prenatal and postnatal development and translation to clinical application. Histol Histopathol 2023; 38:373-389. [PMID: 36218320 DOI: 10.14670/hh-18-533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2023]
Abstract
The testes are the main source of sex steroids in the male, especially androgens and to a lesser extent estrogens. In target cells, steroid hormones typically signal after binding to intracellular receptors, which act as transcription factors. Androgens and estrogens have ubiquitous functions in peripheral organs, but also have paracrine actions within the gonads where they are far more concentrated. The levels of steroid production by the testes vary throughout fetal and postnatal development: they are high in intrauterine life and in the first months after birth, then they decline and are almost undetectable in childhood and increase again during puberty to attain adult levels. The expression of the androgen and estrogen receptors also depict specific ontogenies in the various testicular cell types. The combination of intratesticular steroid concentration with the pattern of expression of the steroid hormone receptors defines androgen and estrogen action on Sertoli, germ and Leydig cells. Here, we review the ontogeny of expression of the androgen and estrogen receptors in the testis, its impact on testicular physiology during prenatal and postnatal development, as well as its implication on the pathophysiology of different disorders affecting gonadal function throughout life.
Collapse
Affiliation(s)
- Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina.
- Unidad de Medicina Traslacional, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
- Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
7
|
Arato I, Giovagnoli S, Di Michele A, Bellucci C, Lilli C, Aglietti MC, Bartolini D, Gambelunghe A, Muzi G, Calvitti M, Eugeni E, Gaggia F, Baroni T, Mancuso F, Luca G. Nickel oxide nanoparticles exposure as a risk factor for male infertility: " In vitro" effects on porcine pre-pubertal Sertoli cells. Front Endocrinol (Lausanne) 2023; 14:1063916. [PMID: 37065743 PMCID: PMC10098343 DOI: 10.3389/fendo.2023.1063916] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 03/17/2023] [Indexed: 04/18/2023] Open
Abstract
Lately, nickel oxide nanoparticles (NiO NPs) have been employed in different industrial and biomedical fields. Several studies have reported that NiO NPs may affect the development of reproductive organs inducing oxidative stress and, resulting in male infertility. We investigated the in vitro effects of NiO NPs on porcine pre-pubertal Sertoli cells (SCs) which undergone acute (24 h) and chronic (from 1 up to 3 weeks) exposure at two subtoxic doses of NiO NPs of 1 μg/ml and 5 μg/ml. After NiO NPs exposure we performed the following analysis: (a) SCs morphological analysis (Light Microscopy); (b) ROS production and oxidative DNA damage, gene expression of antioxidant enzymes (c) SCs functionality (AMH, inhibin B Real-time PCR analysis and ELISA test); (d) apoptosis (WB analysis); (e) pro-inflammatory cytokines (Real-time PCR analysis), and (f) MAPK kinase signaling pathway (WB analysis). We found that the SCs exposed to both subtoxic doses of NiO NPs didn't sustain substantial morphological changes. NiO NPs exposure, at each concentration, reported a marked increase of intracellular ROS at the third week of treatment and DNA damage at all exposure times. We demonstrated, un up-regulation of SOD and HO-1 gene expression, at both concentrations tested. The both subtoxic doses of NiO NPs detected a down-regulation of AMH and inhibin B gene expression and secreted proteins. Only the 5 μg/ml dose induced the activation of caspase-3 at the third week. At the two subtoxic doses of NiO NPs a clear pro-inflammatory response was resulted in an up-regulation of TNF-α and IL-6 in terms of mRNA. Finally, an increased phosphorylation ratio of p-ERK1/2, p-38 and p-AKT was observed up to the third week, at both concentrations. Our results show the negative impact of subtoxic doses NiO NPs chronic exposure on porcine SCs functionality and viability.
Collapse
Affiliation(s)
- Iva Arato
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- *Correspondence: Iva Arato,
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | | | - Catia Bellucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cinzia Lilli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Desirée Bartolini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Angela Gambelunghe
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giacomo Muzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Mario Calvitti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elena Eugeni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesco Gaggia
- Internal Medicine Endocrine and Metabolic Sciences Unit, Santa Maria della Misericordia Hospital of Perugia, Perugia, Italy
| | - Tiziano Baroni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesca Mancuso
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giovanni Luca
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- International Biotechnological Center for Endocrine, Metabolic and Embryo-Reproductive Translational Research (CIRTEMER), Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Division of Medical Andrology and Endocrinology of Reproduction, Saint Mary Hospital, Terni, Italy
| |
Collapse
|
8
|
Mancuso F, Arato I, Bellucci C, Lilli C, Eugeni E, Aglietti MC, Stabile AM, Pistilli A, Brancorsini S, Gaggia F, Calvitti M, Baroni T, Luca G. Zinc restores functionality in porcine prepubertal Sertoli cells exposed to subtoxic cadmium concentration via regulating the Nrf2 signaling pathway. Front Endocrinol (Lausanne) 2023; 14:962519. [PMID: 36843583 PMCID: PMC9950629 DOI: 10.3389/fendo.2023.962519] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 01/24/2023] [Indexed: 02/12/2023] Open
Abstract
INTRODUCTION Among substances released into the environment by anthropogenic activities, the heavy metal cadmium (Cd) is known to induce severe testicular injury causing male subfertility/infertility. Zinc (Zn) is another heavy metal that, unlike Cd, is physiologically present in the testis, being essential for spermatogenesis. We aimed to examine the possibility that 50 µM ZnCl2 could counteract the toxic effects induced by Cd in an in vitro model of porcine prepubertal Sertoli cells (SCs) exposed to both subtoxic (5 μM) and toxic (10 μM) concentrations of CdCl2 for 48 h. MATERIALS AND METHODS Apoptosis, cell cycle, and cell functionality were assessed. The gene expression of Nrf2 and its downstream antioxidant enzymes, ERK1/2, and AKT kinase signaling pathways were evaluated. MATERIALS AND RESULTS We found that Zn, in co-treatment with subtoxic and toxic Cd concentration, increased the number of metabolically active SCs compared to Cd exposure alone but restored SC functionality only in co-treatment with subtoxic Cd concentration with respect to subtoxic Cd alone. Exposure of Cd disrupted cell cycle in SCs, and Zn co-treatment was not able to counteract this effect. Cd alone induced SC death through apoptosis and necrosis in a dose-dependent manner, and co-treatment with Zn increased the pro-apoptotic effect of Cd. Subtoxic and toxic Cd exposures activated the Nrf2 signaling pathway by increasing gene expression of Nrf2 and its downstream genes (SOD, HO-1, and GSHPx). Zn co-treatment with subtoxic Cd attenuated upregulation on the Nrf2 system, while with toxic Cd, the effect was more erratic. Studying ERK1/2 and AKT pathways as a target, we found that the phosphorylation ratio of p-ERK1/2 and p-AKT was upregulated by both subtoxic and toxic Cd exposure alone and in co-treatment with Zn. DISCUSSION Our results suggest that Zn could counteract Cd effects by increasing the number of metabolically active SCs, fully or partially restoring their functionality by modulating Nrf2, ERK1/2, and AKT pathways. Our SC model could be useful to study the effects of early Cd exposure on immature testis, evaluating the possible protective effects of Zn.
Collapse
Affiliation(s)
- Francesca Mancuso
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Iva Arato
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Catia Bellucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cinzia Lilli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elena Eugeni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Division of Medical Andrology and Endocrinology of Reproduction, Saint Mary Hospital, Terni, Italy
| | | | - Anna Maria Stabile
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Stefano Brancorsini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- International Biotechnological Center for Endocrine, Metabolic and Embryo-Reproductive Translational Research (CIRTEMER), Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesco Gaggia
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Mario Calvitti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Tiziano Baroni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- International Biotechnological Center for Endocrine, Metabolic and Embryo-Reproductive Translational Research (CIRTEMER), Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- *Correspondence: Tiziano Baroni,
| | - Giovanni Luca
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Division of Medical Andrology and Endocrinology of Reproduction, Saint Mary Hospital, Terni, Italy
- International Biotechnological Center for Endocrine, Metabolic and Embryo-Reproductive Translational Research (CIRTEMER), Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
9
|
Ramzan R, Khan NA, Khalique A, Aziz MA. A rare form of male pseudohermaphroditism-Persistent Mullerian Duct Syndrome. J Surg Case Rep 2022; 2022:rjac596. [PMID: 36601094 PMCID: PMC9803974 DOI: 10.1093/jscr/rjac596] [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: 11/07/2022] [Accepted: 12/04/2022] [Indexed: 01/01/2023] Open
Abstract
Persistent Mullerian Duct Syndrome (PMDS) is a rare disorder of defective sexual development in males. It is characterized by aberrant synthesis or inadequate action of Mullerian inhibiting factor resulting in derivatives of Mullerian duct, i.e. uterus, fallopian tube and upper vagina, to persist in a phenotypic male with 46XY karyotype. Here, we report a 19-year-old male with bilateral undescended testes. Further evaluation revealed that the patient had both his testes placed intra-abdominally along with a rudimentary uterus.
Collapse
Affiliation(s)
- Raazia Ramzan
- Correspondence address. Dow University of Health Sciences, Karachi 74200, Pakistan. E-mail:
| | | | - Abdul Khalique
- Dow University of Health Sciences, Karachi 74200, Pakistan
| | | |
Collapse
|
10
|
Dapas M, Dunaif A. Deconstructing a Syndrome: Genomic Insights Into PCOS Causal Mechanisms and Classification. Endocr Rev 2022; 43:927-965. [PMID: 35026001 PMCID: PMC9695127 DOI: 10.1210/endrev/bnac001] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Indexed: 01/16/2023]
Abstract
Polycystic ovary syndrome (PCOS) is among the most common disorders in women of reproductive age, affecting up to 15% worldwide, depending on the diagnostic criteria. PCOS is characterized by a constellation of interrelated reproductive abnormalities, including disordered gonadotropin secretion, increased androgen production, chronic anovulation, and polycystic ovarian morphology. It is frequently associated with insulin resistance and obesity. These reproductive and metabolic derangements cause major morbidities across the lifespan, including anovulatory infertility and type 2 diabetes (T2D). Despite decades of investigative effort, the etiology of PCOS remains unknown. Familial clustering of PCOS cases has indicated a genetic contribution to PCOS. There are rare Mendelian forms of PCOS associated with extreme phenotypes, but PCOS typically follows a non-Mendelian pattern of inheritance consistent with a complex genetic architecture, analogous to T2D and obesity, that reflects the interaction of susceptibility genes and environmental factors. Genomic studies of PCOS have provided important insights into disease pathways and have indicated that current diagnostic criteria do not capture underlying differences in biology associated with different forms of PCOS. We provide a state-of-the-science review of genetic analyses of PCOS, including an overview of genomic methodologies aimed at a general audience of non-geneticists and clinicians. Applications in PCOS will be discussed, including strengths and limitations of each study. The contributions of environmental factors, including developmental origins, will be reviewed. Insights into the pathogenesis and genetic architecture of PCOS will be summarized. Future directions for PCOS genetic studies will be outlined.
Collapse
Affiliation(s)
- Matthew Dapas
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Andrea Dunaif
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
11
|
Bartolini D, Arato I, Mancuso F, Giustarini D, Bellucci C, Vacca C, Aglietti MC, Stabile AM, Rossi R, Cruciani G, Rende M, Calafiore R, Luca G, Galli F. Melatonin modulates Nrf2 activity to protect porcine pre-pubertal Sertoli cells from the abnormal H 2 O 2 generation and reductive stress effects of cadmium. J Pineal Res 2022; 73:e12806. [PMID: 35524288 PMCID: PMC9539639 DOI: 10.1111/jpi.12806] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 11/27/2022]
Abstract
Melatonin (MLT) is a cytoprotective agent holding potential to prevent cadmium (Cd) toxicity and its impact in testicular function and fertility. In this study, we explored such potential in porcine pre-pubertal Sertoli cells (SCs). Cd toxicity resulted in impaired SC viability and function, abnormal cellular H2 O2 generation and efflux, and induction of reductive stress by the upregulation of Nrf2 expression and activity, cystine uptake and glutathione biosynthesis, glutathione-S-transferase P (GSTP) expression, and protein glutathionylation inhibition. Cd toxicity also stimulated the activity of cellular kinases (MAPK-ERK1/2 and Akt) and NFkB transcription factor, and cJun expression was increased. MLT produced a potent cytoprotective effect when co-administered with Cd to SCs; its efficacy and the molecular mechanism behind its cytoprotective function varied according to Cd concentrations. However, a significant restoration of cell viability and function, and of H2 O2 levels, was observed both at 5 and 10 μM Cd. Mechanistically, these effects of MLT were associated with a significant reduction of the Cd-induced activation of Nrf2 and GSTP expression at all Cd concentrations. CAT and MAPK-ERK1/2 activity upregulation was associated with these effects at 5 μM Cd, whereas glutathione biosynthesis and efflux were involved at 10 μM Cd together with an increased expression of the cystine transporter xCT, of cJun and Akt and NFkB activity. MLT protects SCs from Cd toxicity reducing its H2 O2 generation and reductive stress effects. A reduced activity of Nrf2 and the modulation of other molecular players of MLT signaling, provide a mechanistic rational for the cytoprotective effect of this molecule in SCs.
Collapse
Affiliation(s)
- Desirée Bartolini
- Department of Pharmaceutical SciencesUniversity of PerugiaPerugiaItaly
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic AnatomyUniversity of PerugiaPerugiaItaly
| | - Iva Arato
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | | | - Daniela Giustarini
- Department of Biotechnology, Chemistry and PharmacyUniversity of SienaSienaItaly
| | - Catia Bellucci
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Carmine Vacca
- Department of Chemistry, Biology and BiotechnologyUniversity of PerugiaPerugiaItaly
| | | | - Anna Maria Stabile
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic AnatomyUniversity of PerugiaPerugiaItaly
| | - Ranieri Rossi
- Department of Biotechnology, Chemistry and PharmacyUniversity of SienaSienaItaly
| | - Gabriele Cruciani
- Department of Chemistry, Biology and BiotechnologyUniversity of PerugiaPerugiaItaly
| | - Mario Rende
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic AnatomyUniversity of PerugiaPerugiaItaly
| | - Riccardo Calafiore
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
- Department of Medicine and Surgery, International Biotechnological Center for Endocrine, Metabolic and Embryo‐Reproductive Translational Research (CIRTEMER)University of PerugiaPerugiaItaly
| | - Giovanni Luca
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
- Division of Medical Andrology and Endocrinology of ReproductionSaint Mary HospitalTerniItaly
- Department of Medicine and Surgery, International Biotechnological Center for Endocrine, Metabolic and Embryo‐Reproductive Translational Research (CIRTEMER)University of PerugiaPerugiaItaly
| | - Francesco Galli
- Department of Pharmaceutical SciencesUniversity of PerugiaPerugiaItaly
| |
Collapse
|
12
|
Edelsztein NY, Valeri C, Lovaisa MM, Schteingart HF, Rey RA. AMH Regulation by Steroids in the Mammalian Testis: Underlying Mechanisms and Clinical Implications. Front Endocrinol (Lausanne) 2022; 13:906381. [PMID: 35712256 PMCID: PMC9195137 DOI: 10.3389/fendo.2022.906381] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/11/2022] [Indexed: 11/26/2022] Open
Abstract
Anti-Müllerian hormone (AMH) is a distinctive biomarker of the immature Sertoli cell. AMH expression, triggered by specific transcription factors upon fetal Sertoli cells differentiation independently of gonadotropins or sex steroids, drives Müllerian duct regression in the male, preventing the development of the uterus and Fallopian tubes. AMH continues to be highly expressed by Sertoli until the onset of puberty, when it is downregulated to low adult levels. FSH increases testicular AMH output by promoting immature Sertoli cell proliferation and individual cell expression. AMH secretion also showcases a differential regulation exerted by intratesticular levels of androgens and estrogens. In the fetus and the newborn, Sertoli cells do not express the androgen receptor, and the high androgen concentrations do not affect AMH expression. Conversely, estrogens can stimulate AMH production because estrogen receptors are present in Sertoli cells and aromatase is stimulated by FSH. During childhood, sex steroids levels are very low and do not play a physiological role on AMH production. However, hyperestrogenic states upregulate AMH expression. During puberty, testosterone inhibition of AMH expression overrides stimulation by estrogens and FSH. The direct effects of sex steroids on AMH transcription are mediated by androgen receptor and estrogen receptor α action on AMH promoter sequences. A modest estrogen action is also mediated by the membrane G-coupled estrogen receptor GPER. The understanding of these complex regulatory mechanisms helps in the interpretation of serum AMH levels found in physiological or pathological conditions, which underscores the importance of serum AMH as a biomarker of intratesticular steroid concentrations.
Collapse
Affiliation(s)
- Nadia Y. Edelsztein
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Clara Valeri
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - María M. Lovaisa
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Helena F. Schteingart
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Rodolfo A. Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
- Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
13
|
Concepción-Zavaleta M, Paz Ibarra JL, Ramos-Yataco A, Coronado-Arroyo J, Concepción-Urteaga L, Roseboom PJ, Williams CA. Assessment of hormonal status in male infertility. An update. Diabetes Metab Syndr 2022; 16:102447. [PMID: 35272174 DOI: 10.1016/j.dsx.2022.102447] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/26/2022] [Accepted: 03/01/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS The prevalence and incidence of infertility are increasing worldwide; they are associated with a significant economic and social impact. Infertility is defined as the inability to achieve pregnancy after 12 months or more of regular unprotected sex. In recent times, the male factor has gained importance and currently it contributes to approximately 50% of infertility cases. Multiple etiologies are stated, such as metabolic, anatomical, genetic or even idiopathic causes; however, the main cause is semen abnormalities. The aim of this manuscript is to provide a complete review of hormonal assessment of male infertility, as well as to review the physiology and pathophysiology related to the male gonadal axis. METHODS This study is a narrative abstract carried out on basis of systematic bibliographic review, using articles indexed in PubMed/Medline, Scopus, Embase and Scielo, which were published during the last 20 years. RESULTS The cornerstone of the evaluation of the hormonal status is semen analysis. Clinicians must rule out hypogonadism in those patients who present oligospermia and azoospermia, by determining levels of testosterone and gonadotropins, which provide the functionality status of the hypothalamic-pituitary-testicular axis. Evaluation of the adrenal, thyroid, and lactotroph axis are indicated in those patients with central hypogonadism. CONCLUSIONS Despite advances in the diagnosis of male infertility, some causes are not fully understood, therefore, it is crucial to perform a timely hormonal evaluation of the male factor in infertile couples, in order to provide adequate treatment and improve fertility rates.
Collapse
Affiliation(s)
| | - José Luis Paz Ibarra
- Division of Endocrinology, National Hospital Edgardo Rebagliati Martins, Lima, Peru
| | | | - Julia Coronado-Arroyo
- Division of Obstetrics and Gynecology, National Hospital Edgardo Rebagliati Martins, Lima, Peru
| | | | - Pela J Roseboom
- Division of Emergency Medicine, Regional Academic Hospital of Trujillo, Trujillo, Peru
| | | |
Collapse
|
14
|
Domain G, Buczkowska J, Kalak P, Wydooghe E, Banchi P, Pascottini OB, Niżański W, Van Soom A. Serum Anti-Müllerian Hormone: A Potential Semen Quality Biomarker in Stud Dogs? Animals (Basel) 2022; 12:ani12030323. [PMID: 35158647 PMCID: PMC8833318 DOI: 10.3390/ani12030323] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 01/03/2023] Open
Abstract
Anti-Müllerian hormone (AMH) has been suggested to be involved in spermatogenesis. The aim of this study was to investigate the relationship between blood serum AMH concentration and semen quality in dogs. Moreover, this study sought to find the optimal cut-off point value of serum AMH with the greatest sensitivity and specificity to predict semen quality. Forty-five clinically healthy dogs were included in the study and their age as well as the following semen parameters were determined and correlated to serum AMH concentration: total sperm output, normal morphology, plasma membrane integrity, total motility, progressive motility, and velocity parameters. Statistical analysis for correlations were performed using Spearman’s correlation coefficients. Moderate negative associations were found between serum AMH and semen total motility (r = −0.38, p = 0.01), progressive motility (r = −0.36, p = 0.01), and normal morphology (r = −0.36, p= 0.02). Based on these associations, an AMH concentration of 5.54 µg/L was found to be the optimal cut-off point value to obtain the greatest summation of sensitivity (86%) and specificity (63%) to predict semen quality. The serum AMH assay may therefore be a potential hormonal marker to predict which dogs would require further semen analysis. Future research is however needed to confirm these preliminary results.
Collapse
Affiliation(s)
- Guillaume Domain
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; (E.W.); (P.B.); (O.B.P.); (A.V.S.)
- Correspondence:
| | - Justyna Buczkowska
- Department of Reproduction and Clinic of Farm Animals, University of Environmental Science, Grundwaldzki Square 49, 50-357 Wroclaw, Poland; (J.B.); (P.K.); (W.N.)
| | - Patrycja Kalak
- Department of Reproduction and Clinic of Farm Animals, University of Environmental Science, Grundwaldzki Square 49, 50-357 Wroclaw, Poland; (J.B.); (P.K.); (W.N.)
| | - Eline Wydooghe
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; (E.W.); (P.B.); (O.B.P.); (A.V.S.)
| | - Penelope Banchi
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; (E.W.); (P.B.); (O.B.P.); (A.V.S.)
| | - Osvaldo Bogado Pascottini
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; (E.W.); (P.B.); (O.B.P.); (A.V.S.)
- Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Wojciech Niżański
- Department of Reproduction and Clinic of Farm Animals, University of Environmental Science, Grundwaldzki Square 49, 50-357 Wroclaw, Poland; (J.B.); (P.K.); (W.N.)
| | - Ann Van Soom
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; (E.W.); (P.B.); (O.B.P.); (A.V.S.)
| |
Collapse
|
15
|
Kanakatti Shankar R, Dowlut-McElroy T, Dauber A, Gomez-Lobo V. Clinical Utility of Anti-Mullerian Hormone in Pediatrics. J Clin Endocrinol Metab 2022; 107:309-323. [PMID: 34537849 PMCID: PMC8764360 DOI: 10.1210/clinem/dgab687] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Indexed: 12/15/2022]
Abstract
CONTEXT Anti-Mullerian hormone (AMH) was originally described in the context of sexual differentiation in the male fetus but has gained prominence now as a marker of ovarian reserve and fertility in females. In this mini-review, we offer an updated synopsis on AMH and its clinical utility in pediatric patients. DESIGN AND RESULTS A systematic search was undertaken for studies related to the physiology of AMH, normative data, and clinical role in pediatrics. In males, AMH, secreted by Sertoli cells, is found at high levels prenatally and throughout childhood and declines with progression through puberty to overlap with levels in females. Thus, serum AMH has clinical utility as a marker of testicular tissue in males with differences in sexual development and cryptorchidism and in the evaluation of persistent Mullerian duct syndrome. In females, serum AMH has been used as a predictive marker of ovarian reserve and fertility, but prepubertal and adolescent AMH assessments need to be interpreted cautiously. AMH is also a marker of tumor burden, progression, and recurrence in germ cell tumors of the ovary. CONCLUSIONS AMH has widespread clinical diagnostic utility in pediatrics but interpretation is often challenging and should be undertaken in the context of not only age and sex but also developmental and pubertal stage of the child. Nonstandardized assays necessitate the need for assay-specific normative data. The recognition of the role of AMH beyond gonadal development and maturation may usher in novel diagnostic and therapeutic applications that would further expand its utility in pediatric care.
Collapse
Affiliation(s)
- Roopa Kanakatti Shankar
- Division of Endocrinology, Children’s National Hospital, Washington DC, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Correspondence: Roopa Kanakatti Shankar, MBBS, MS, George Washington University School of Medicine, Endocrinologist, Children’s National Hospital, 111 Michigan Ave NW, Washington DC, 20010, USA.
| | - Tazim Dowlut-McElroy
- Pediatric and Adolescent Gynecology Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Andrew Dauber
- Division of Endocrinology, Children’s National Hospital, Washington DC, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Veronica Gomez-Lobo
- Pediatric and Adolescent Gynecology Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| |
Collapse
|
16
|
Identification of AMH and AMHR2 Variants Led to the Diagnosis of Persistent Müllerian Duct Syndrome in Three Cases. Genes (Basel) 2022; 13:genes13010159. [PMID: 35052499 PMCID: PMC8774887 DOI: 10.3390/genes13010159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 11/18/2022] Open
Abstract
Persistent Müllerian duct syndrome (PMDS) is a rare autosomal recessive disorder of sexual development in males, defined by the presence of Müllerian remnants with otherwise normal sexual differentiation. Mutations in anti-Müllerian hormone (AMH) and AMH receptor type 2 (AMHR2) genes are the main causes of PMDS. In this study, we performed molecular genetic analysis of 11 unrelated cryptorchidism patients using whole-exome sequencing and classified the variants. Three of the 11 patients had biallelic mutations in AMH or AMHR2. Case 1 carried a homozygous 4-bp deletion; c.321_324del:p.Q109Lfs*29 in exon 1 of AMH (NM_000479 transcript), which is a frameshift mutation, leading to the loss of function of AMH. Case 2 carried compound heterozygous mutations; c.494_502del (p.I165_A168delinsT) in exon 4 and g.6147C>A of AMHR2 (NM_001164690 transcript). Case 3 carried compound heterozygous mutations; c.G1168A (p.E390K) in exon 9 and c.A1315G (p.M439V) in exon 10 of AMHR2 (NM_001164690 transcript). All three patients were admitted due to azoospermia- and oligospermia-caused infertility. They were furtherly diagnosed with PMDS, as pelvic magnetic resonance imaging revealed the presence of Müllerian remnants. Our study suggests that PMDS and genetic analysis should be considered during the differential diagnosis of cryptorchidism.
Collapse
|
17
|
Mancuso F, Arato I, Di Michele A, Antognelli C, Angelini L, Bellucci C, Lilli C, Boncompagni S, Fusella A, Bartolini D, Russo C, Moretti M, Nocchetti M, Gambelunghe A, Muzi G, Baroni T, Giovagnoli S, Luca G. Effects of Titanium Dioxide Nanoparticles on Porcine Prepubertal Sertoli Cells: An " In Vitro" Study. Front Endocrinol (Lausanne) 2022; 12:751915. [PMID: 35046890 PMCID: PMC8762334 DOI: 10.3389/fendo.2021.751915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022] Open
Abstract
The increasing use of nanomaterials in a variety of industrial, commercial, medical products, and their environmental spreading has raised concerns regarding their potential toxicity on human health. Titanium dioxide nanoparticles (TiO2 NPs) represent one of the most commonly used nanoparticles. Emerging evidence suggested that exposure to TiO2 NPs induced reproductive toxicity in male animals. In this in vitro study, porcine prepubertal Sertoli cells (SCs) have undergone acute (24 h) and chronic (from 1 up to 3 weeks) exposures at both subtoxic (5 µg/ml) and toxic (100 µg/ml) doses of TiO2 NPs. After performing synthesis and characterization of nanoparticles, we focused on SCs morphological/ultrastructural analysis, apoptosis, and functionality (AMH, inhibin B), ROS production and oxidative DNA damage, gene expression of antioxidant enzymes, proinflammatory/immunomodulatory cytokines, and MAPK kinase signaling pathway. We found that 5 µg/ml TiO2 NPs did not induce substantial morphological changes overtime, but ultrastructural alterations appeared at the third week. Conversely, SCs exposed to 100 µg/ml TiO2 NPs throughout the whole experiment showed morphological and ultrastructural modifications. TiO2 NPs exposure, at each concentration, induced the activation of caspase-3 at the first and second week. AMH and inhibin B gene expression significantly decreased up to the third week at both concentrations of nanoparticles. The toxic dose of TiO2 NPs induced a marked increase of intracellular ROS and DNA damage at all exposure times. At both concentrations, the increased gene expression of antioxidant enzymes such as SOD and HO-1 was observed whereas, at the toxic dose, a clear proinflammatory stress was evaluated along with the steady increase in the gene expression of IL-1α and IL-6. At both concentrations, an increased phosphorylation ratio of p-ERK1/2 was observed up to the second week followed by the increased phosphorylation ratio of p-NF-kB in the chronic exposure. Although in vitro, this pilot study highlights the adverse effects even of subtoxic dose of TiO2 NPs on porcine prepubertal SCs functionality and viability and, more importantly, set the basis for further in vivo studies, especially in chronic exposure at subtoxic dose of TiO2 NPs, a condition closer to the human exposure to this nanoagent.
Collapse
Affiliation(s)
- Francesca Mancuso
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Iva Arato
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Cinzia Antognelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- International Biotechnological Center for Endocrine, Metabolic and Embryo-Reproductive Translational Research (CIRTEMER), Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Luca Angelini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Catia Bellucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cinzia Lilli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Simona Boncompagni
- Center for Advanced Studies and Technology (CAST) and Department of Neuroscience, Imaging and Clinical Sciences (DNICS), University G. d’Annunzio (Ud’A) of Chieti-Pescara, Chieti, Italy
| | - Aurora Fusella
- Center for Advanced Studies and Technology (CAST) and Department of Neuroscience, Imaging and Clinical Sciences (DNICS), University G. d’Annunzio (Ud’A) of Chieti-Pescara, Chieti, Italy
| | - Desirée Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Carla Russo
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Massimo Moretti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Morena Nocchetti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Angela Gambelunghe
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giacomo Muzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Tiziano Baroni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- International Biotechnological Center for Endocrine, Metabolic and Embryo-Reproductive Translational Research (CIRTEMER), Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Giovanni Luca
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- International Biotechnological Center for Endocrine, Metabolic and Embryo-Reproductive Translational Research (CIRTEMER), Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Division of Medical Andrology and Endocrinology of Reproduction, Saint Mary Hospital, Terni, Italy
| |
Collapse
|
18
|
Rey RA. Recent advancement in the treatment of boys and adolescents with hypogonadism. Ther Adv Endocrinol Metab 2022; 13:20420188211065660. [PMID: 35035874 PMCID: PMC8753232 DOI: 10.1177/20420188211065660] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/22/2021] [Indexed: 12/02/2022] Open
Abstract
Clinical manifestations and the need for treatment varies according to age in males with hypogonadism. Early foetal-onset hypogonadism results in disorders of sex development (DSD) presenting with undervirilised genitalia whereas hypogonadism established later in foetal life presents with micropenis, cryptorchidism and/or micro-orchidism. After the period of neonatal activation of the gonadal axis has waned, the diagnosis of hypogonadism is challenging because androgen deficiency is not apparent until the age of puberty. Then, the differential diagnosis between constitutional delay of puberty and central hypogonadism may be difficult. During infancy and childhood, treatment is usually sought because of micropenis and/or cryptorchidism, whereas lack of pubertal development and relative short stature are the main complaints in teenagers. Testosterone therapy has been the standard, although off-label, in the vast majority of cases. However, more recently alternative therapies have been tested: aromatase inhibitors to induce the hypothalamic-pituitary-testicular axis in boys with constitutional delay of puberty and replacement with GnRH or gonadotrophins in those with central hypogonadism. Furthermore, follicle-stimulating hormone (FSH) priming prior to hCG or luteinizing hormone (LH) treatment seems effective to induce an enhanced testicular enlargement. Although the rationale for gonadotrophin or GnRH treatment is based on mimicking normal physiology, long-term results are still needed to assess their impact on adult fertility.
Collapse
Affiliation(s)
- Rodolfo A. Rey
- Rodolfo A. Rey Centro de Investigaciones
Endocrinológicas ‘Dr. César Bergadá’ (CEDIE), CONICET – FEI – División de
Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Gallo 1330, C1425EFD
Buenos Aires, Argentina
| |
Collapse
|
19
|
Acar S, Nalbantoğlu Ö, Gürsoy S, Özkaya B, Köprülü Ö, Arslan G, Hazan F, Özkan B. Identification of two AMH gene variants in two unrelated patients with persistent Müllerian duct syndrome: one novel variant. Gynecol Endocrinol 2021; 37:476-479. [PMID: 33787423 DOI: 10.1080/09513590.2021.1908253] [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] [Indexed: 10/21/2022] Open
Abstract
INTRODUCTION Persistent müllerian duct syndrome (PMDS) is a rare form of 46, XY disorder of sex development characterized by the persistence of the müllerian structures (uterus, fallopian tubes, the upper part of the vagina) in phenotypically and genotypically normal males. This disease occurs as a result of impairment in the synthesis, release or effect of anti-Müllerian hormone (AMH) during the embryonic period. Approximately 85-88% of PMDS cases have been reported to have AMH or AMHRII mutation. CASE Herein, we report two PMDS cases from unrelated two families who presented with bilateral undescended testes, persistence of müllerian remnants, and low/undetectable serum AMH levels. Molecular genetic analysis revealed two homozygous variants in AMH. The first one is a novel missense variant (c.1315C > T), the latter is a frameshift variant caused by a deletion (c.343_344delCT), which is less frequently reported type in AMH. CONCLUSION The diagnosis of PMDS should be kept in mind in patients with externally normal males, bilateral cryptorchidism, and signs of müllerian remnants on laparoscopy.
Collapse
Affiliation(s)
- Sezer Acar
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Özlem Nalbantoğlu
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Semra Gürsoy
- Division of Pediatric Genetics, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Beyhan Özkaya
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Özge Köprülü
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Gülçin Arslan
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Filiz Hazan
- Department of Medical Genetics, Dr. Behçe Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Behzat Özkan
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| |
Collapse
|
20
|
Hoyos LR, Visser JA, McLuskey A, Chazenbalk GD, Grogan TR, Dumesic DA. Loss of anti-Müllerian hormone (AMH) immunoactivity due to a homozygous AMH gene variant rs10417628 in a woman with classical polycystic ovary syndrome (PCOS). Hum Reprod 2021; 35:2294-2302. [PMID: 32918081 DOI: 10.1093/humrep/deaa199] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/06/2020] [Indexed: 12/24/2022] Open
Abstract
Anti-Müllerian hormone (AMH) is produced by granulosa cells of pre-antral and small antral ovarian follicles. In polycystic ovary syndrome (PCOS), higher levels of serum AMH are usually encountered due to the ample presence of small antral follicles and a high AMH production per follicular unit which have led to the proposal of AMH as a serum diagnostic marker for PCOS or as a surrogate for polycystic ovarian morphology (PCOM). However, heterozygous coding mutations of the AMH gene with decreased in vitro bioactivity have been described in some women with PCOS. Such mutation carriers have a trend toward reduced serum AMH levels compared to noncarriers, although both types of women with PCOS have similar circulating gonadotropin and testosterone (T) levels. This report describes a normal-weight woman with PCOS by NIH criteria with severely reduced AMH levels (index woman with PCOS). Our objective was to examine the molecular basis for her reduced serum AMH levels and to compare her endocrine characteristics to similar-weight women with PCOS and detectable AMH levels. Twenty normoandrogenic ovulatory (control) and 13 age- and BMI-matched women with PCOS (19-35 years; 19-25 kg/m2) underwent transvaginal sonography and serum hormone measures including gonadotropins, sex hormone-binding globulin, total and free T, androstenedione, dehydroepiandrosterone sulfate, estrone, estradiol and AMH. The latter was measured by ELISA (Pico-AMH: Ansh Labs, Webster, TX, USA). Women with PCOS and detectable AMH had higher serum AMH (10.82 (6.74-13.40) ng/ml, median (interquartile range)), total and free T (total T: 55.5 (49.5-62.5) ng/dl; free T: 5.65 (4.75-6.6) pg/ml) levels and greater total antral follicle count (AFC) (46 (39-59) follicles) than controls (AMH: 4.03 (2.47-6.11) ng/ml; total T: 30 (24.5-34.5) ng/dl; free T: 2.2 (1.8-2.45) pg/ml; AFC 16 (14.5-21.5) follicles, P < 0.05, all values), along with a trend toward LH hypersecretion (P = 0.06). The index woman with PCOS had severely reduced serum AMH levels (∼0.1 ng/ml), although she also had a typical NIH-defined PCOS phenotype resembling that of the other women with PCOS and elevated AMH levels. All women with PCOS, including the index woman with PCOS, exhibited LH hypersecretion, hyperandrogenism, reduced serum estrogen/androgen ratios and PCOM. A homozygous Ala515Val variant (rs10417628) in the mature region of AMH was identified in the index woman with PCOS. Recombinant hAMH-515Val displayed normal processing and bioactivity, yet had severely reduced immunoactivity when measured by the commercial pico-AMH ELISA assay by Ansh Labs. In conclusion, homozygous AMH variant rs10417628 may severely impair serum AMH immunoactivity without affecting its bioactivity or PCOS phenotypic expression. Variants in AMH can interfere with serum AMH immunoactivity without affecting the phenotype in PCOS. This observation can be accompanied by discordance between AMH immunoactivity and bioactivity.
Collapse
Affiliation(s)
- Luis R Hoyos
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jenny A Visser
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Anke McLuskey
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Gregorio D Chazenbalk
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Tristan R Grogan
- Department of Medicine Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
21
|
Evaluation of a new automated immunoassay for the quantification of anti-Müllerian hormone. Pract Lab Med 2021; 25:e00220. [PMID: 34095410 PMCID: PMC8145750 DOI: 10.1016/j.plabm.2021.e00220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/08/2021] [Indexed: 12/04/2022] Open
Abstract
Objectives A newly developed fully automated Lumipulse G AMH method (Fujirebio Diagnostics) was recently introduced in clinical laboratories for quantitative determination of anti-Müllerian hormone (AMH) level in human serum or plasma. AMH has emerged as value-added biomarker in the assessment of ovarian reserve, in diagnosis of granulosa cells cancer and in the investigation of gonadal disorders. We compared Lumipulse G AMH assay performances with other methods largely applied for AMH measurements. Design and Methods The Lumipulse G AMH method based on two-step sandwich chemiluminescence enzyme immunoassay was assessed on Lumipulse G600II analyzer. The evaluation study included imprecisions, sensitivity and linearity whereas a comparison study was performed on a heterogeneous population of 114 patients by using the Elecsys AMH Plus assay on COBAS 8000 e602 module (Roche Diagnostics). Results Lumipulse G AMH system showed good repeatability (within-run imprecision) with CV values below 1% (0.5% and 0.9% for high and low serum pools). Similarly within-laboratory imprecision was assessed with CV values of 2.5% and 1.6% for high and low level controls respectively. A linearity regression formula of 1.0119x-0.067 with a coefficient of determination (r2) equal to 0.999 was obtained in a range from 0.044 to 22.42 ng/ml. Passing-Bablok regression analysis was performed for assay comparability of AMH measurements. Results were closely correlated (correlation coefficient = 0.997) with a regression equation (y = 1.230x-0.025) showing a positive slope. Also, Bland-Altman analysis confirmed a good agreement between Lumipulse G AMH and Roche Elecsys AMH Plus assays with a bias of 17.76% in a large measurement range. Conclusions The performance of Lumipulse G AMH system was highly comparable with that of Roche Elecsys AMH Plus assay although approximately 10% higher values of AMH levels were observed for Lumipulse AMH system at all range of concentrations. Nevertheless the Lumipulse G system seems to be largely suitable for quantitative determination of AMH level in small-scale laboratory because of the reduced size and the use of single cartridge per test assuring flexibility and easy handling.
Collapse
|
22
|
Lambert S, Peycelon M, Samara-Boustani D, Hyon C, Dumeige L, Peuchmaur M, Fiot E, Léger J, Simon D, Paye-Jaouen A, Bouligand J, Siffroi JP, Carel JC, McElreavey K, El Ghoneimi A, Brachet C, Bouvattier C, Martinerie L. SRY-negative 46,XX testicular/ovotesticular DSD: Long-term outcomes and early blockade of gonadotropic axis. Clin Endocrinol (Oxf) 2021; 94:667-676. [PMID: 33296530 DOI: 10.1111/cen.14389] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/19/2020] [Accepted: 11/22/2020] [Indexed: 01/25/2023]
Abstract
OBJECTIVE SRY-negative 46,XX testicular and ovotesticular disorders/differences of sex development (T/OTDSD) represent a very rare and unique DSD condition where testicular tissue develops in the absence of a Y chromosome. To date, very few studies have described the phenotype, clinical and surgical management and long-term outcomes of these patients. Particularly, early blockade of the gonadotropic axis in patients raised in the female gender to minimize postnatal androgenization has never been reported. DESIGN Retrospective description of sixteen 46,XX T/OTDSD patients. RESULTS Sixteen 46,XX SRY-negative T/OTDSD were included. Most (12/16) were diagnosed in the neonatal period. Sex of rearing was male for six patients and female for ten, while the clinical presentation varied, with an external masculinization score from 1 to 10. Five patients raised as girl were successfully treated with GnRH analog to avoid virilization during minipuberty. Ovotestes/testes were found bilaterally for 54% of the patients and unilaterally for the others (with a contralateral ovary). Gonadal surgery preserved appropriate tissue in the majority of cases. Spontaneous puberty occurred in two girls and one boy, while two boys required hormonal induction of puberty. One of the girls conceived spontaneously and had an uneventful pregnancy. DNA analyses (SNP-array, next-generation sequencing and whole-exome sequencing) were performed. A heterozygous frameshit mutation in the NR2F2 gene was identified in one patient. CONCLUSIONS This study presents a population of patients with 46,XX SRY-negative T/OTDSD. Early blockade of gonadotropic axis appears efficient to reduce and avoid further androgenization in patients raised as girls.
Collapse
Affiliation(s)
- Sophie Lambert
- Pediatric Endocrinology Department, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Robert Debré Hospital, Assistance publique-Hôpitaux de Paris, Paris, France
- Pediatric Endocrinology Department, CHC, Liège, Belgium
- Pediatric Endocrinology Department, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | - Matthieu Peycelon
- Pediatric Endocrinology Department, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Robert Debré Hospital, Assistance publique-Hôpitaux de Paris, Paris, France
- Pediatric Surgery and Urology Department, Robert Debré Hospital, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Assistance publique-Hôpitaux de Paris, Paris, France
| | - Dinane Samara-Boustani
- Pediatric Endocrinology Department, CHU Necker-Enfants Malades, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Assistance-Publique Hôpitaux de Paris, Paris, France
| | - Capucine Hyon
- INSERM, Maladies génétiques d'expression pédiatrique, APHP, Hôpital d'Enfants Armand Trousseau, Sorbonne Université, Paris, France
| | - Laurence Dumeige
- Pediatric Endocrinology Department, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Robert Debré Hospital, Assistance publique-Hôpitaux de Paris, Paris, France
- INSERM UMR-S1185, Le Kremlin Bicêtre, France
| | - Michel Peuchmaur
- Université de Paris, Paris, France
- Pathology Department, Robert Debré Hospital, Assistance publique-Hôpitaux de Paris, Paris, France
| | - Elodie Fiot
- Pediatric Endocrinology Department, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Robert Debré Hospital, Assistance publique-Hôpitaux de Paris, Paris, France
| | - Juliane Léger
- Pediatric Endocrinology Department, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Robert Debré Hospital, Assistance publique-Hôpitaux de Paris, Paris, France
- Université de Paris, Paris, France
- INSERM UMR 1141, DHU Protect, Paris, France
| | - Dominique Simon
- Pediatric Endocrinology Department, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Robert Debré Hospital, Assistance publique-Hôpitaux de Paris, Paris, France
| | - Annabel Paye-Jaouen
- Pediatric Surgery and Urology Department, Robert Debré Hospital, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Assistance publique-Hôpitaux de Paris, Paris, France
| | - Jérome Bouligand
- Molecular Genetics and Hormonology Department, Assistance Publique-Hôpitaux de Paris, Bicêtre Hospital, Le Kremlin Bicêtre, France
| | - Jean-Pierre Siffroi
- INSERM, Maladies génétiques d'expression pédiatrique, APHP, Hôpital d'Enfants Armand Trousseau, Sorbonne Université, Paris, France
| | - Jean-Claude Carel
- Pediatric Endocrinology Department, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Robert Debré Hospital, Assistance publique-Hôpitaux de Paris, Paris, France
- Université de Paris, Paris, France
- INSERM UMR 1141, DHU Protect, Paris, France
| | - Ken McElreavey
- Department of Human Developmental Genetics, Institut Pasteur, Paris, France
| | - Alaa El Ghoneimi
- Pediatric Surgery and Urology Department, Robert Debré Hospital, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Assistance publique-Hôpitaux de Paris, Paris, France
- Université de Paris, Paris, France
| | - Cécile Brachet
- Pediatric Endocrinology Department, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | - Claire Bouvattier
- Pediatric Endocrinology Department, Bicêtre Hospital, Centre de Référence DEV-GEN, Paris-Saclay University, Kremlin Bicêtre, Paris, France
| | - Laetitia Martinerie
- Pediatric Endocrinology Department, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Robert Debré Hospital, Assistance publique-Hôpitaux de Paris, Paris, France
- Université de Paris, Paris, France
- INSERM UMR-S1185, Le Kremlin Bicêtre, France
| |
Collapse
|
23
|
Xue R, Lin W, Sun J, Watanabe M, Xu A, Araki M, Nasu Y, Tang Z, Huang P. The role of Wnt signaling in male reproductive physiology and pathology. Mol Hum Reprod 2021; 27:gaaa085. [PMID: 33543289 DOI: 10.1093/molehr/gaaa085] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/26/2020] [Indexed: 12/14/2022] Open
Abstract
Accumulating evidence has shown that Wnt signaling is deeply involved in male reproductive physiology, and malfunction of the signal path can cause pathological changes in genital organs and sperm cells. These abnormalities are diverse in manifestation and have been constantly found in the knockout models of Wnt studies. Nevertheless, most of the research solely focused on a certain factor in the Wnt pathway, and there are few reports on the overall relation between Wnt signals and male reproductive physiology. In our review, Wnt findings relating to the reproductive system were sought and summarized in terms of Wnt ligands, Wnt receptors, Wnt intracellular signals and Wnt regulators. By sorting out and integrating relevant functions, as well as underlining the controversies among different reports, our review aims to offer an overview of Wnt signaling in male reproductive physiology and pathology for further mechanistic studies.
Collapse
Affiliation(s)
- Ruizhi Xue
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Wenfeng Lin
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jingkai Sun
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Masami Watanabe
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Abai Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Motoo Araki
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yasutomo Nasu
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Zhengyan Tang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Huang
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Okayama Medical Innovation Center, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
24
|
Kalinchenko NY, Kolodkina AA, Raygorodskaya NY, Tiulpakov AN. [Clinical and molecular characteristics of patients with 46,XY DSD due to NR5A1 gene mutations]. ACTA ACUST UNITED AC 2020; 66:62-69. [PMID: 33351340 DOI: 10.14341/probl12445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/28/2020] [Accepted: 06/29/2020] [Indexed: 01/18/2023]
Abstract
Steroidogenic factor 1 (SF1, NR5A1) is a nuclear receptor that regulates multiple genes involved in adrenal and gonadal development, steroidogenesis, and the reproductive axis. Human mutations in SF1 were initially found in patients with severe gonadal dysgenesis and primary adrenal failure. However, more recent case reports have suggested that heterozygous mutations in SF1 may also be found in patients with 46,XY partial gonadal dysgenesis and underandrogenization but normal adrenal function. We have analyzed the gene encoding SF1 (NR5A1) in a cohort of 310 Russian patients with 46,XY disorders of sex development (DSD). Heterozygous SF1 variants were found in 36 out of 310 (11.6%) of cases, among them 15 were not previously described. We have not found any phenotype-genotype correlations and any clinical and laboratory markers that would allow to suspect this type of before conducting molecular genetic analysis.
Collapse
|
25
|
Valeri C, Lovaisa MM, Racine C, Edelsztein NY, Riggio M, Giulianelli S, Venara M, Bedecarrás P, Ballerini MG, di Clemente N, Lamb CA, Schteingart HF, Rey RA. Molecular mechanisms underlying AMH elevation in hyperoestrogenic states in males. Sci Rep 2020; 10:15062. [PMID: 32934281 PMCID: PMC7492256 DOI: 10.1038/s41598-020-71675-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/06/2020] [Indexed: 02/08/2023] Open
Abstract
Anti-Müllerian hormone (AMH) is secreted by Sertoli cells of the testes from early fetal life until puberty, when it is downregulated by androgens. In conditions like complete androgen insensitivity syndrome (CAIS), AMH downregulation does not occur and AMH increases at puberty, due in part to follicle-stimulating hormone (FSH) effect. However, other conditions like Peutz-Jeghers syndrome (PJS), characterised by low FSH, also have increased AMH. Because both CAIS and PJS may present as hyperoestrogenic states, we tested the hypothesis that oestradiol (E2) upregulates AMH expression in peripubertal Sertoli cells and explored the molecular mechanisms potentially involved. The results showed that E2 is capable of inducing an upregulation of endogenous AMH and of the AMH promoter activity in the prepubertal Sertoli cell line SMAT1, signalling through ERα binding to a specific ERE sequence present on the hAMH promoter. A modest action was also mediated through the membrane oestrogen receptor GPER. Additionally, the existence of ERα expression in Sertoli cells in patients with CAIS was confirmed by immunohistochemistry. The evidence presented here provides biological plausibility to the hypothesis that testicular AMH production increases in clinical conditions in response to elevated oestrogen levels.
Collapse
Affiliation(s)
- Clara Valeri
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - María M Lovaisa
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Chrystèle Racine
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine (CRSA), 75012, Paris, France.,Institut Hospitalo-Universitaire ICAN, 75013, Paris, France.,Sorbonne Paris Cité, Paris-Diderot Université, 75013, Paris, France
| | - Nadia Y Edelsztein
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Marina Riggio
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), C1428ADN, Buenos Aires, Argentina
| | - Sebastián Giulianelli
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), C1428ADN, Buenos Aires, Argentina.,Instituto de Biología de Organismos Marinos, IBIOMAR-CCT (CENPAT-CONICET), U9120ACD, Puerto Madryn, Argentina
| | - Marcela Venara
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Patricia Bedecarrás
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - María G Ballerini
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Nathalie di Clemente
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine (CRSA), 75012, Paris, France.,Institut Hospitalo-Universitaire ICAN, 75013, Paris, France
| | - Caroline A Lamb
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), C1428ADN, Buenos Aires, Argentina
| | - Helena F Schteingart
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina. .,Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, C1121ABG, Buenos Aires, Argentina.
| |
Collapse
|
26
|
Shrikhande L, Shrikhande B, Shrikhande A. AMH and Its Clinical Implications. J Obstet Gynaecol India 2020; 70:337-341. [PMID: 33041549 DOI: 10.1007/s13224-020-01362-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/19/2020] [Indexed: 12/21/2022] Open
Abstract
Anti-Müllerian Hormone (AMH) is critical for physiologic involution of the Mullerian ducts during sexual differentiation in the male fetus. In women, AMH is a product of the small antral follicles in the ovaries and serves to function as an autocrine and paracrine regulator of follicular maturation. As the size of the residual follicular pool depends on the quantity of small antral follicles and declines over time, the serum AMH level in women follows a characteristic trajectory: a gradual decline throughout the reproductive years and a precipitous drop at menopause, becoming undetectable soon after. Thus, AMH is clinically useful as a screening tool for diminished ovarian reserve (Cui et al. in Fertil Steril 105(02):481-485, 2016). Perturbations in serum AMH are linked with a variety of pathological conditions, for instance, polycystic ovaries syndrome (PCOS), the pathophysiology likely being the excess follicles in this syndrome which produce increased amounts of AMH (Dumont et al. in Reprod Biol Endocrinol 13:137, 2015). AMH is also elevated in some ovarian tumors such as adult granulosa cell tumors, and it can be used as a tumor marker to gauge response to therapy and monitor for recurrence. Within the domain of assisted reproductive technology, serum AMH assays are widely used to derive prognostic information such as the chance of successful ovarian stimulation, subsequent embryo quality and even pregnancy rates. Finally, in the rapidly evolving field of oncofertility, serum AMH holds great promise as a predictor of ovarian reserve after completion of cancer therapy. Our aim is to put forth an in-depth review of the clinical applications of AMH in contemporary practice.
Collapse
Affiliation(s)
- Laxmi Shrikhande
- Shrikhande Hospital and Research Centre Pvt Ltd, Nagpur, 34/2 Abhyankar Road, Dhantoli, Nagpur, Maharashtra 440012 India
| | - Bhushan Shrikhande
- Shrikhande Hospital and Research Centre Pvt Ltd, Nagpur, 34/2 Abhyankar Road, Dhantoli, Nagpur, Maharashtra 440012 India
| | - Aditya Shrikhande
- Shrikhande Hospital and Research Centre Pvt Ltd, Nagpur, 34/2 Abhyankar Road, Dhantoli, Nagpur, Maharashtra 440012 India
| |
Collapse
|
27
|
Rey RA. Biomarcadores de hipogonadismo masculino en la infancia y la adolescencia. ADVANCES IN LABORATORY MEDICINE 2020; 1:20190043. [PMCID: PMC10158747 DOI: 10.1515/almed-2019-0043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 01/19/2020] [Indexed: 06/28/2023]
Abstract
El eje hipotálamo-hipófiso-testicular es activo en la vida fetal y durante los primeros meses de la vida posnatal: la hipófisis secreta hormona luteinizante (LH) y folículo-estimulante (FSH), mientras que el testículo produce testosterona y factor insulino-símil 3 (INSL3) en las células de Leydig y hormona anti-Mülleriana (AMH) e inhibina B en las células de Sertoli. En la infancia, los niveles séricos de gonadotrofinas, testosterona y factor INSL3 disminuyen a valores prácticamente indetectables, pero los de AMH e inhibina B permanecen altos. En la pubertad, se reactivan las gonadotrofinas y la producción de testosterona e INSL3, aumenta la inhibina y disminuye la AMH, como signo de maduración de la célula de Sertoli. Sobre la base del conocimiento de la fisiología del desarrollo del eje, es posible utilizar clínicamente estos biomarcadores para interpretar la fisiopatología y diagnosticar las diferentes formas de hipogonadismo que pueden presentarse en la infancia y la adolescencia.
Collapse
Affiliation(s)
- Rodolfo A. Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET-FEI- División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo, 1330, C1425EFD, Buenos Aires, Argentina
| |
Collapse
|
28
|
Rey RA. Biomarkers of male hypogonadism in childhood and adolescence. ADVANCES IN LABORATORY MEDICINE 2020; 1:20200024. [PMID: 37363780 PMCID: PMC10159267 DOI: 10.1515/almed-2020-0024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 01/19/2020] [Indexed: 06/28/2023]
Abstract
Objectives The objective of this review was to characterize the use of biomarkers of male hypogonadism in childhood and adolescence. Contents The hypothalamic-pituitary-gonadal (HPG) axis is active during fetal life and over the first months of postnatal life. The pituitary gland secretes follicle stimulating hormone (FSH) and luteinizing hormone (LH), whereas the testes induce Leydig cells to produce testosterone and insulin-like factor 3 (INSL), and drive Sertoli cells to secrete anti-Müllerian hormone (AMH) and inhibin B. During childhood, serum levels of gonadotropins, testosterone and insulin-like 3 (INSL3) decline to undetectable levels, whereas levels of AMH and inhibin B remain high. During puberty, the production of gonadotropins, testosterone, and INSL3 is reactivated, inhibin B increases, and AMH decreases as a sign of Sertoli cell maturation. Summary and outlook Based on our knowledge of the developmental physiology of the HPG axis, these biomarkers can be used in clinical practice to interpret the physiopathology of hypogonadism. Additionally, these markers can have diagnostic value in different forms of hypogonadism that may appear during childhood and adolescence.
Collapse
Affiliation(s)
- Rodolfo A. Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET-FEI- División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD, Buenos Aires, Argentina
- Departamento de Histología, Biología Celular, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, C1121ABG, Buenos Aires, Argentina
| |
Collapse
|
29
|
Dapas M, Dunaif A. The contribution of rare genetic variants to the pathogenesis of polycystic ovary syndrome. ACTA ACUST UNITED AC 2020; 12:26-32. [PMID: 32440573 DOI: 10.1016/j.coemr.2020.02.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a highly heritable disorder, but only a small proportion of the heritability can be accounted for by common genetic risk variants identified to date. It is possible that variants with lower allele frequencies that cannot be detected using genome-wide association study arrays contribute to PCOS. Here, we discuss the challenges inherent to studying rare genetic variants in complex disease and review several recent studies that have used DNA sequencing techniques to investigate whether rare variants play a role in PCOS pathogenesis. We evaluate these findings in the context of the latest literature in PCOS and complex disease genetics.
Collapse
|
30
|
Sansone A, Isidori AM, Kliesch S, Schlatt S. Immunohistochemical characterization of the anti-Müllerian hormone receptor type 2 (AMHR-2) in human testes. Endocrine 2020; 68:215-221. [PMID: 32026338 PMCID: PMC7160062 DOI: 10.1007/s12020-020-02210-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/17/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE In males, AMH is secreted by immature Sertoli cells; following exposure to endogenous androgens, Sertoli cells undergo a process of maturation which ultimately inhibits AMH expression to undetectable levels in the serum. However, expression of AMH receptor (AMHR-2) has never been studied in human testes, and high intratubular concentrations of AMH have been reported in recent literature. We therefore assessed expression of AMHR-2 in several testicular tissue samples by immunohistochemistry (IHC). METHODS The IHC method was first validated on tissue samples from healthy human testis (n = 2) and from marmoset ovary (n = 1). The same method was then used for assessment on testicular histopathology specimens from patients with mixed atrophy (MA, n = 2), spermatogenetic arrest (SA, n = 2), Sertoli cell-only syndrome (SCO, n = 1), Klinefelter syndrome (KS, n = 1), and nonseminomatous germ cell tumors (NSGCT, n = 1). Tissue samples from two subjects at different pubertal stages (AndroProtect (AP), aged 5 and 14 years) with hematological malignancies were also retrieved. RESULTS In adult men, AMHR-2 was expressed on peritubular mesenchymal cells, with patterns closely mirroring α-smooth muscle actin expression. Similar patterns were preserved in almost all conditions; however, in nonseminomatous germ cell tumors the tissue architecture was lost, including AMHR-2 expression. More positive and diffuse staining was observed in tissue samples from prepubertal testes. CONCLUSIONS In specimens from both healthy and affected testes, AMHR-2 expression appears weaker in adult than in prepubertal tissue sections. The persistence of AMHR-2 expression seemingly hints at a possible effect of intratesticular AMH on the tubular walls.
Collapse
Affiliation(s)
- A Sansone
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149, Münster, Germany
- Department of Experimental Medicine, Food Science and Endocrinology, Sapienza University of Rome, 00161, Rome, Italy
| | - A M Isidori
- Department of Experimental Medicine, Food Science and Endocrinology, Sapienza University of Rome, 00161, Rome, Italy
| | - S Kliesch
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149, Münster, Germany
| | - S Schlatt
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149, Münster, Germany.
| |
Collapse
|
31
|
Huang X, Xu X, Dai Y, Cheng Z, Zheng X, Huo X. Association of prenatal exposure to PAHs with anti-Müllerian hormone (AMH) levels and birth outcomes of newborns. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 723:138009. [PMID: 32213412 DOI: 10.1016/j.scitotenv.2020.138009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/10/2020] [Accepted: 03/16/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Polycyclic aromatic hydrocarbons (PAHs) are chemicals that cause serious concerns because of their carcinogenicity and endocrine disrupting ability. OBJECTIVE In the current study, we studied how urinary PAH metabolites are related with the dose-effects of hormone levels and birth outcomes. METHOD 163 pregnant women without health problems and 163 newborns were enrolled in hospitals in Guiyu (e-waste-exposed area) and Haojiang (reference area) from May 2016 to May 2017. Urine samples were collected to measure hydroxylated PAH (OH-PAH) metabolite levels. Umbilical cord blood was used for measurement of hormone levels. Anthropometric parameters of newborns, such as anogenital distance (AGD), were also measured. RESULTS Eight of ten urinary PAH metabolites in the exposed group were significantly higher than in the reference group. Levels of umbilical cord serum estradiol (E2) and testosterone (T) in the exposed group were significantly lower than those in the reference group. Birth weight was positively correlated with 2-OHFlu (2-hydroxyfluorene). Head circumference was negatively correlated with 9-OHFlu, 3-OHPhe (3-hydroxyphenanthrene), 9-OHPhe, and ƩOHFlu (sum of 2-OHFlu and 9-OHFlu). Serum E2 and T levels were negatively correlated with most OH-PAHs. In addition, we found that serum anti-Müllerian hormone (AMH) level was positively correlated with AGD, and serum E2 level was negatively correlated with neonatal head circumference. CONCLUSIONS PAH exposure in pregnant women may adversely affect the birth outcomes of newborns, especially AGD; and AMH may be involved in the process. Establishing a baseline for the relationship between PAH exposure and health is important to protect the health of mothers and children living in electronic waste (e-waste) recycling areas.
Collapse
Affiliation(s)
- Xiaofan Huang
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, Guangdong, China; Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 511443, Guangdong, China
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, Guangdong, China; Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Yifeng Dai
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, Guangdong, China; Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands
| | - Zhiheng Cheng
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, Guangdong, China; Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands
| | - Xiangbin Zheng
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 511443, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 511443, Guangdong, China.
| |
Collapse
|
32
|
Karaoglan M. Correlation of anti-Mullerian hormone with humanchorionic gonadotropin test in the evaluation of testicular function of children with 46 XY male hypogonadism: Use of anti-Mullerian hormone as abiomarker. J Paediatr Child Health 2020; 56:411-419. [PMID: 31614067 DOI: 10.1111/jpc.14643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/06/2019] [Accepted: 09/09/2019] [Indexed: 01/11/2023]
Abstract
AIM It is challenging to evaluate reproductive potential during childhood. These challenges necessitate the use of invasive dynamic tests. Although the anti-Mullerian hormone (AMH) is a reliable biomarker in evaluating testicular function, especially in the pre-pubertal period, there are uncertainties concerning its use in a clinical setting. This study is focused on comparing the AMH and human chorionic gonadotropin (hCG) test in boys with hypogonadism. METHODS A total of 160 boys aged between 0 and 18 years who presented with complaints associated with hypogonadism were prospectively enrolled in the study. All children were assigned to the following five groups: gonadal disorders (n = 34), androgen synthesis and end organ effect disorder (n = 48), isolated genital malformation disorders (n = 57), hypogonadotropic hypogonadism (n = 15) and constitutional delayed puberty (n = 6). All children underwent a short 3-day hCG test (1500 U/m2 /day). The concordance and correlation were evaluated between the hCG test and AMH. RESULTS All groups exhibited a strong correlation (r160 = 0.689) and strong concordance (Kappa coefficient160 = 0.7) between the AMH and hCG test. Values of AMH higher than 32.7 pmol/L and hCG responses higher than 86 pmol/L were significant as indicative markers of functional testicular tissue presence. CONCLUSIONS This study has shown that there is a strong correlation between the AMH and short-term hCG test and that values of AMH higher than 32.7 pmol/L and stimulated testosterone higher than 86 pmol/L can be used as indicators of functionally sufficient testicular tissue. These results indicate that AMH value can be used as a reliable and useful biomarker in the evaluation of the testicular function in 46 XY hypogonadism.
Collapse
Affiliation(s)
- Murat Karaoglan
- Department of Pediatric Endocrinology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
33
|
Arato I, Grande G, Barrachina F, Bellucci C, Lilli C, Jodar M, Aglietti MC, Mancini F, Vincenzoni F, Pontecorvi A, Calafiore R, Oliva R, Luca G, Mancuso F, Milardi D. " In vitro" Effect of Different Follicle-Stimulating Hormone Preparations on Sertoli Cells: Toward a Personalized Treatment for Male Infertility. Front Endocrinol (Lausanne) 2020; 11:401. [PMID: 32625170 PMCID: PMC7314925 DOI: 10.3389/fendo.2020.00401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/19/2020] [Indexed: 01/02/2023] Open
Abstract
Follicle-stimulating hormone (FSH), a major regulator of spermatogenesis, has a crucial function in the development and function of the testis and it is extensively given as a fertility treatment to stimulate spermatogenesis. We analyzed the effects of different FSH preparations (α-follitropin, β-follitropin, and urofollitropin) in combination with testosterone on porcine pre-pubertal Sertoli cells. To study the effect of the different FSH treatments in the Sertoli cell function we performed Real Time PCR analysis of AMH, inhibin B, and FSH-r, an ELISA assay for AMH and inhibin B, and a high-throughput comparative proteomic analysis. We verified that all three preparations induced a reduction of AMH in terms of mRNA and secreted proteins, and an increase of inhibin B in terms of mRNA in all the FSH formulations, while solely α-follitropin produced an increase of secreted inhibin B in the culture medium. Comparative proteomic analysis of the three FSH preparations identified 46 proteins, 11 up-regulated and 2 down-regulated. Surprisingly, the combination of testosterone with β-follitropin specifically induced an up-regulation of eight specific secreted proteins. Our study, showing that the three different FSH preparations induce different effects, could offer the opportunity to shed light inside new applications to a personalized reproductive medicine.
Collapse
Affiliation(s)
- Iva Arato
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Giuseppe Grande
- Research Unit on Human Reproduction, International Scientific Institute Paul VI, Rome, Italy
- Division of Endocrinology, Fondazione Policlinico Universitario “Agostino Gemelli”, Rome, Italy
| | - Ferran Barrachina
- Molecular Biology of Reproduction and Development Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Catia Bellucci
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Cinzia Lilli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Meritxell Jodar
- Molecular Biology of Reproduction and Development Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic, Barcelona, Spain
| | | | - Francesca Mancini
- Research Unit on Human Reproduction, International Scientific Institute Paul VI, Rome, Italy
| | - Federica Vincenzoni
- Biochemistry and Clinical Biochemistry Institute, School of Medicine, Catholic University of Rome, Rome, Italy
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico “A. Gemelli” IRCCS, Rome, Italy
| | - Alfredo Pontecorvi
- Research Unit on Human Reproduction, International Scientific Institute Paul VI, Rome, Italy
- Division of Endocrinology, Fondazione Policlinico Universitario “Agostino Gemelli”, Rome, Italy
| | - Riccardo Calafiore
- Department of Medicine, University of Perugia, Perugia, Italy
- Division of Medical Andrology and Endocrinology of Reproduction, University of Perugia and Saint Mary Hospital, Terni, Italy
| | - Rafael Oliva
- Molecular Biology of Reproduction and Development Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic, Barcelona, Spain
| | - Giovanni Luca
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
- Division of Medical Andrology and Endocrinology of Reproduction, University of Perugia and Saint Mary Hospital, Terni, Italy
- *Correspondence: Giovanni Luca
| | - Francesca Mancuso
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Domenico Milardi
- Research Unit on Human Reproduction, International Scientific Institute Paul VI, Rome, Italy
- Division of Endocrinology, Fondazione Policlinico Universitario “Agostino Gemelli”, Rome, Italy
| |
Collapse
|
34
|
Grinspon RP, Bergadá I, Rey RA. Male Hypogonadism and Disorders of Sex Development. Front Endocrinol (Lausanne) 2020; 11:211. [PMID: 32351452 PMCID: PMC7174651 DOI: 10.3389/fendo.2020.00211] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/25/2020] [Indexed: 12/13/2022] Open
Abstract
Disorders of Sex Development (DSD) are congenital anomalies in which there is a discordance between chromosomal, genetic, gonadal, and/or internal/external genital sex. In XY individuals, the process of fetal sex differentiation can be disrupted at the stage of gonadal differentiation, resulting in gonadal dysgenesis, a form of early fetal-onset primary hypogonadism characterized by insufficient androgen and anti-Müllerian hormone (AMH) production, which leads to the development of ambiguous or female genitalia. The process of sex differentiation can also be disrupted at the stage of genital differentiation, due to isolated defects in androgen or AMH secretion, but not both. These are forms of fetal-onset hypogonadism with dissociated gonadal dysfunction. In this review, we present a perspective on impaired testicular endocrine function, i.e., fetal-onset male hypogonadism, resulting in incomplete virilization at birth.
Collapse
Affiliation(s)
- Romina P. Grinspon
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET—FEI—División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
- *Correspondence: Romina P. Grinspon
| | - Ignacio Bergadá
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET—FEI—División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Rodolfo A. Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET—FEI—División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
- Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
35
|
Grinspon RP, Rey RA. Molecular Characterization of XX Maleness. Int J Mol Sci 2019; 20:ijms20236089. [PMID: 31816857 PMCID: PMC6928850 DOI: 10.3390/ijms20236089] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 11/29/2019] [Accepted: 11/29/2019] [Indexed: 12/18/2022] Open
Abstract
Androgens and anti-Müllerian hormone (AMH), secreted by the foetal testis, are responsible for the development of male reproductive organs and the regression of female anlagen. Virilization of the reproductive tract in association with the absence of Müllerian derivatives in the XX foetus implies the existence of testicular tissue, which can occur in the presence or absence of SRY. Recent advancement in the knowledge of the opposing gene cascades driving to the differentiation of the gonadal ridge into testes or ovaries during early foetal development has provided insight into the molecular explanation of XX maleness.
Collapse
Affiliation(s)
- Romina P. Grinspon
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
- Correspondence: (R.P.G.); (R.A.R.); Tel.: +54-11-49635931 (R.P.G.)
| | - Rodolfo A. Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
- Departamento de Histología, Biología Celular, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
- Correspondence: (R.P.G.); (R.A.R.); Tel.: +54-11-49635931 (R.P.G.)
| |
Collapse
|
36
|
Grinspon RP, Arozarena M, Prada S, Bargman G, Sanzone M, Morales Bazurto M, Gutiérrez M, Bedecarrás P, Kannemann A, Elena GO, Gottlieb S, Berenstein AJ, Ropelato MG, Bergadá I, Aversa LA, Rey RA. Safety of standardised treatments for haematologic malignancies as regards to testicular endocrine function in children and teenagers. Hum Reprod 2019; 34:2480-2494. [PMID: 31768530 DOI: 10.1093/humrep/dez216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/02/2019] [Indexed: 12/23/2022] Open
Abstract
STUDY QUESTION Does standardised treatments used in children and adolescents with haematologic malignancies, including acute lymphoblastic (ALL) or myeloid leukaemia (AML) and non-Hodgkin lymphoma (NHL), affect endocrine function of the developing testes? SUMMARY ANSWER Therapy of haematologic malignancies do not provoke an overt damage of Sertoli and Leydig cell populations, as revealed by normal levels of anti-Müllerian hormone (AMH) and testosterone, but a mild primary testicular dysfunction may be observed, compensated by moderate gonadotropin elevation, during pubertal development. WHAT IS KNOWN ALREADY Evidence exists on the deleterious effect that chemotherapy and radiotherapy have on germ cells, and some attention has been given to the effects on Leydig and Sertoli cells of the adult gonads, but information is virtually non-existent on the effects of oncologic treatment on testicular somatic cell components during childhood and adolescence. STUDY DESIGN, SIZE, DURATION A retrospective, analytical, observational study included 97 boys with haematological malignancies followed at two tertiary paediatric public hospitals in Buenos Aires, Argentina, between 2002 and 2015. PARTICIPANTS/MATERIALS, SETTING, METHODS Clinical records of males aged 1-18 years, referred with the diagnoses of ALL, AML or NHL for the assessment of gonadal function, were eligible. We assessed serum levels of AMH and FSH as biomarkers of Sertoli cell endocrine function and testosterone and LH as biomarkers of Leydig cell function. MAIN RESULTS AND THE ROLE OF CHANCE All hormone levels were normal in the large majority of patients until early pubertal development. From Tanner stage G3 onwards, while serum AMH and testosterone kept within the normal ranges, gonadotropins reached mildly to moderately elevated values in up to 35.9% of the cases, indicating a compensated Sertoli and/or Leydig cell dysfunction, which generally did not require hormone replacement therapy. LIMITATIONS, REASONS FOR CAUTION Serum inhibin B determination and semen analysis were not available for most patients; therefore, we could not conclude on potential fertility impairment or identify whether primary Sertoli cell dysfunction resulted in secondary depleted spermatogenesis or whether primary germ cell damage impacted Sertoli cell function. WIDER IMPLICATIONS OF THE FINDINGS The regimens used in the treatment of boys and adolescents with ALL, AML or NHL in the past two decades seem relatively safe for endocrine testicular function; nonetheless, a mild primary testicular endocrine dysfunction may be observed, usually compensated by slightly elevated gonadotropin secretion by the pituitary in adolescents, and not requiring hormone replacement therapy. No clinically relevant risk factor, such as severity of the disease or treatment protocol, could be identified in association with the compensated endocrine dysfunction. STUDY FUNDING/COMPETING INTEREST(S) This work was partially funded by grants PIP 11220130100687 of Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and PICT 2016-0993 of Fondo para la Investigación Científica y Tecnológica (FONCYT), Argentina. R.A.R., R.P.G. and P.B. have received honoraria from CONICET (Argentina) for technology services using the AMH ELISA. L.A.A. is part-time employee of CSL Behring Argentina. The other authors have no conflicts of interest to disclose.
Collapse
Affiliation(s)
- Romina P Grinspon
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - María Arozarena
- Unidad de Hematología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Silvina Prada
- Unidad de Hematología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Graciela Bargman
- División de Endocrinología, Hospital de Niños Pedro de Elizalde, C1270AAN Buenos Aires, Argentina
| | - María Sanzone
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Marjorie Morales Bazurto
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Marcela Gutiérrez
- Unidad de Hematología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Patricia Bedecarrás
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Ana Kannemann
- Unidad de Hematología, Hospital Pedro de Elizalde, C1270AAN Buenos Aires, Argentina
| | - Graciela O Elena
- Unidad de Hematología, Hospital Pedro de Elizalde, C1270AAN Buenos Aires, Argentina
| | - Silvia Gottlieb
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Ariel J Berenstein
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP), CONICET-GCBA, Laboratorio de Biología Molecular, División Patología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - María Gabriela Ropelato
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Ignacio Bergadá
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Luis A Aversa
- Unidad de Hematología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| |
Collapse
|
37
|
Edelsztein NY, Kashimada K, Schteingart HF, Rey RA. CYP26B1 declines postnatally in Sertoli cells independently of androgen action in the mouse testis. Mol Reprod Dev 2019; 87:66-77. [DOI: 10.1002/mrd.23302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Nadia Y. Edelsztein
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de EndocrinologíaHospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - Kenichi Kashimada
- Department of Pediatrics and Developmental BiologyTokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Helena F. Schteingart
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de EndocrinologíaHospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - Rodolfo A. Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de EndocrinologíaHospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
- Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de MedicinaUniversidad de Buenos Aires Buenos Aires Argentina
| |
Collapse
|
38
|
Edelsztein NY, Racine C, di Clemente N, Schteingart HF, Rey RA. Androgens downregulate anti-Müllerian hormone promoter activity in the Sertoli cell through the androgen receptor and intact steroidogenic factor 1 sites. Biol Reprod 2019; 99:1303-1312. [PMID: 29985989 DOI: 10.1093/biolre/ioy152] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 07/06/2018] [Indexed: 12/14/2022] Open
Abstract
Testicular anti-Müllerian hormone (AMH) production is inhibited by androgens around pubertal onset, as observed under normal physiological conditions and in patients with precocious puberty. In agreement, AMH downregulation is absent in patients with androgen insensitivity. The molecular mechanisms underlying the negative regulation of AMH by androgens remain unknown. Our aim was to elucidate the mechanisms through which androgens downregulate AMH expression in the testis. A direct negative effect of androgens on the transcriptional activity of the AMH promoter was found using luciferase reporter assays in the mouse prepubertal Sertoli cell line SMAT1. A strong inhibition of AMH promoter activity was seen in the presence of both testosterone and DHT and of the androgen receptor. By site-directed mutagenesis and chromatin immunoprecipitation assays, we showed that androgen-mediated inhibition involved the binding sites for steroidogenic factor 1 (SF1) present in the proximal promoter of the AMH gene. In this study, we describe for the first time the mechanism behind AMH inhibition by androgens, as seen in physiological and pathological conditions in males. Inhibition of AMH promoter activity by androgens could be due to protein-protein interactions between the ligand-bound androgen receptor and SF1 or by blockage of SF1 binding to its sites on the AMH promoter.
Collapse
Affiliation(s)
- Nadia Y Edelsztein
- Centro de Investigaciones Endocrinológicas "Dr César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Chrystèle Racine
- Sorbonne Universitté, INSERM, Centre de Recherche Saint Antoine (CRSA), IHU ICAN, 75012 Paris, France
| | - Nathalie di Clemente
- Sorbonne Universitté, INSERM, Centre de Recherche Saint Antoine (CRSA), IHU ICAN, 75012 Paris, France
| | - Helena F Schteingart
- Centro de Investigaciones Endocrinológicas "Dr César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.,Departamento de Histología, Biología Celular, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
| |
Collapse
|
39
|
Rak AY, Trofimov AV, Ischenko AM. Anti-mullerian hormone receptor type II as a Potential Target for Antineoplastic Therapy. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2019. [DOI: 10.1134/s1990750819030053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
40
|
Mäkelä JA, Koskenniemi JJ, Virtanen HE, Toppari J. Testis Development. Endocr Rev 2019; 40:857-905. [PMID: 30590466 DOI: 10.1210/er.2018-00140] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/17/2018] [Indexed: 12/28/2022]
Abstract
Production of sperm and androgens is the main function of the testis. This depends on normal development of both testicular somatic cells and germ cells. A genetic program initiated from the Y chromosome gene sex-determining region Y (SRY) directs somatic cell specification to Sertoli cells that orchestrate further development. They first guide fetal germ cell differentiation toward spermatogenic destiny and then take care of the full service to spermatogenic cells during spermatogenesis. The number of Sertoli cells sets the limits of sperm production. Leydig cells secrete androgens that determine masculine development. Testis development does not depend on germ cells; that is, testicular somatic cells also develop in the absence of germ cells, and the testis can produce testosterone normally to induce full masculinization in these men. In contrast, spermatogenic cell development is totally dependent on somatic cells. We herein review germ cell differentiation from primordial germ cells to spermatogonia and development of the supporting somatic cells. Testicular descent to scrota is necessary for normal spermatogenesis, and cryptorchidism is the most common male birth defect. This is a mild form of a disorder of sex differentiation. Multiple genetic reasons for more severe forms of disorders of sex differentiation have been revealed during the last decades, and these are described along with the description of molecular regulation of testis development.
Collapse
Affiliation(s)
- Juho-Antti Mäkelä
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jaakko J Koskenniemi
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Helena E Virtanen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Pediatrics, Turku University Hospital, Turku, Finland
| |
Collapse
|
41
|
Rak AY, Trofimov AV, Ischenko AM. [Mullerian inhibiting substance type II receptor as a potential target for antineoplastic therapy]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 65:202-213. [PMID: 31258143 DOI: 10.18097/pbmc20196503202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The review considers properties of the type II anti-Mullerian hormone receptor (mullerian inhibiting substance receptor type II, MISRII), a transmembrane sensor with its own serine/threonine protein kinase activity, triggering apoptosis of the Mullerian ducts in mammalian embryogenesis and providing formation of the male type reproductive system. According to recent data, MISRII overexpression in the postnatal period is found in cells of a number of ovarian, mammary gland, and prostate tumors, and anti-Mullerian hormone (AMH) has a pro-apoptotic effect on MISRII-positive tumor cells. This fact makes MISRII a potential target for targeted anti-cancer therapy. Treatment based on targeting MISRII seems to be a much more effective alternative to the traditional one and will significantly reduce the drug dose. However, the mechanism of MISRII-AMH interaction is still poorly understood, so the development of new anticancer drugs is complicated. The review analyzes MISRII molecular structure and expression levels in various tissues and cell lines, as well as current understanding of the AMH binding mechanisms and data on the possibility of using MISRII as a target for the action of AMH-based antineoplastic drugs.
Collapse
Affiliation(s)
- A Ya Rak
- State Research Institute of Highly Pure Biopreparations, Saint-Petersburg, Russia; Saint-Petersburg State University, Saint-Petersburg, Russia
| | - A V Trofimov
- State Research Institute of Highly Pure Biopreparations, Saint-Petersburg, Russia
| | - A M Ischenko
- State Research Institute of Highly Pure Biopreparations, Saint-Petersburg, Russia
| |
Collapse
|
42
|
Acero MG, Moreno O, Gutiérrez A, Sánchez C, Cataño JG, Suárez-Obando F, Rojas A. Novel homozygous mutation in a colombian patient with persistent müllerian duct syndrome: expanded phenotype. Int Braz J Urol 2019; 45:1064-1070. [PMID: 31184456 PMCID: PMC6844334 DOI: 10.1590/s1677-5538.ibju.2018.0808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 03/17/2019] [Indexed: 05/30/2023] Open
Abstract
The anti-Müllerian hormone triggers the regression of uterus and fallopian tubes in male embryos; if there are problems in the synthesis or action of this protein, Müllerian structures persist in an otherwise phenotypic male. The most frequent clinical presentation of Persistent Mullerian Duct syndrome is cryptorchidism and inguinal hernia. The few cases reported in adults are incidental findings or inguinal hernias. However, we present an adult male with history of bilateral cryptorchidism with unsuccessful orchidopexy, who presents with a large abdominal mass with the finding of a seminomatous tumor and persistence of Müllerian structures, in whom the variant c.916delC (p.Leu306Cysfs*29) in the AMHR2 gene not previously reported was documented.
Collapse
Affiliation(s)
- Mary García Acero
- Human Genetic Institute, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Olga Moreno
- Human Genetic Institute, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Andrés Gutiérrez
- Department of Urology, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Catalina Sánchez
- Department of Urology, Hospital Universitario San Ignacio, Bogotá, Colombia
| | | | - Fernando Suárez-Obando
- Human Genetic Institute, Pontificia Universidad Javeriana, Bogotá, Colombia.,Genetic Service, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Adriana Rojas
- Human Genetic Institute, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
43
|
Abstract
The term primary gonadal failure encompasses not only testicular insufficiency in 46,XY males and ovarian insufficiency in 46,XX females, but also those disorders of sex development (DSD) which result in gender assignment that is at variance with the genotype and gonadal type. In boys, causes of gonadal failure include Klinefelter and other aneuploidy syndromes, bilateral cryptorchidism, testicular torsion, and forms of 46,XY DSD such as partial androgen insensitivity. Causes in girls include Turner syndrome and other aneuploidies, galactosemia, and autoimmune ovarian failure. Iatrogenic causes in both boys and girls include the late effects of childhood cancer treatment, total body irradiation prior to bone marrow transplantation, and iron overload in transfusion-dependent thalassaemia. In this paper, a brief description of the physiology of testicular and ovarian development is followed by a section on the causes and practical management of gonadal impairment in boys and girls. Protocols for pubertal induction and post-pubertal hormone replacement - intramuscular, oral and transdermal testosterone in boys; oral and transdermal oestrogen in girls - are then given. Finally, current and future strategies for assisted conception and fertility preservation are discussed.
Collapse
Affiliation(s)
- Asmahane Ladjouze
- Faculté de Médecine d'Alger, Service de Pédiatrie, Centre Hospitalo-Universitaire Bad El Oued, 1 Boulevard Said Touati, Algiers, Algeria.
| | - Malcolm Donaldson
- Section of Child Health, School of Medicine, Queen Elizabeth University Hospital, Govan Road, Glasgow, G51 4TF, United Kingdom.
| |
Collapse
|
44
|
Bouchard MF, Bergeron F, Grenier Delaney J, Harvey LM, Viger RS. In Vivo Ablation of the Conserved GATA-Binding Motif in the Amh Promoter Impairs Amh Expression in the Male Mouse. Endocrinology 2019; 160:817-826. [PMID: 30759208 PMCID: PMC6426834 DOI: 10.1210/en.2019-00047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 02/08/2019] [Indexed: 12/23/2022]
Abstract
GATA4 is an essential transcriptional regulator required for gonadal development, differentiation, and function. In the developing testis, proposed GATA4-regulated genes include steroidogenic factor 1 (Nr5a1), SRY-related HMG box 9 (Sox9), and anti-Müllerian hormone (Amh). Although some of these genes have been validated as genuine GATA4 targets, it remains unclear whether GATA4 is a direct regulator of endogenous Amh transcription. We used a CRISPR/Cas9-based approach to specifically inactivate or delete the sole GATA-binding motif of the proximal mouse Amh promoter. AMH mRNA and protein levels were assessed at developmental time points corresponding to elevated AMH levels: fetal and neonate testes in males and adult ovaries in females. In males, loss of GATA binding to the Amh promoter significantly reduced Amh expression. Although the loss of GATA binding did not block the initiation of Amh transcription, AMH mRNA and protein levels failed to upregulate in the developing fetal and neonate testis. Interestingly, adult male mice presented no anatomical anomalies and had no evidence of retained Müllerian duct structures, suggesting that AMH levels, although markedly reduced, were sufficient to masculinize the male embryo. In contrast to males, GATA binding to the Amh promoter was dispensable for Amh expression in the adult ovary. These results provide conclusive evidence that in males, GATA4 is a positive modulator of Amh expression that works in concert with other key transcription factors to ensure that the Amh gene is sufficiently expressed in a correct spatiotemporal manner during fetal and prepubertal testis development.
Collapse
Affiliation(s)
- Marie France Bouchard
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
| | - Francis Bergeron
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
| | - Jasmine Grenier Delaney
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
| | - Louis-Mathieu Harvey
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
| | - Robert S Viger
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
- Department of Obstetrics, Gynecology, and Reproduction, Université Laval, Quebec, Quebec, Canada
- Correspondence: Robert S. Viger, PhD, Reproduction, Mother and Child Health, Room T3-67, Centre de Recherche du CHU de Québec–Université Laval, 2705 Laurier Boulevard, Quebec, Quebec G1V 4G2, Canada. E-mail:
| |
Collapse
|
45
|
Yates AP, Jopling HM, Burgoyne NJ, Hayden K, Chaloner CM, Tetlow L. Paediatric reference intervals for plasma anti-Müllerian hormone: comparison of data from the Roche Elecsys assay and the Beckman Coulter Access assay using the same cohort of samples. Ann Clin Biochem 2019; 56:536-547. [PMID: 30889973 PMCID: PMC6688133 DOI: 10.1177/0004563219830733] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background Autoanalyser methods for the measurement of anti-Müllerian hormone have been
introduced into clinical laboratories but few reports of paediatric
reference intervals using these new assays have been published. Methods After prior evaluation of the Roche Elecsys anti-Müllerian hormone assay
against the Beckman Coulter modified second generation anti-Müllerian
Hormone enzyme-linked immunosorbent assay using samples from adult females,
a cohort of paediatric samples which had previously been assessed using the
Beckman Coulter Access anti-Müllerian hormone assay was analysed using the
Roche Elecsys anti-Müllerian hormone assay. Results The Roche Elecsys anti-Müllerian hormone assay measured significantly lower
than the Beckman Coulter modified second generation anti-Müllerian Hormone
enzyme-linked immunosorbent assay. In the paediatric cohort measured with
the Roche Elecsys assay, male levels are very high from birth to puberty
after which they fall towards postpubertal female levels. Male results were
similar to those previously obtained using the Beckman Coulter Access
anti-Müllerian hormone assay on the same cohort. Roche Elecsys
anti-Müllerian hormone in the females was very low in the neonatal and
prepubertal years and the postpubertal trend, with a steady rise from 15
years, was smoother than previously modelled using the Beckman Coulter
Access anti-Müllerian hormone assay. Conclusion Anti-Müllerian hormone levels measured with the Roche Elecsys assay were
significantly lower than the Beckman Coulter modified second generation
enzyme-linked immunosorbent assay suggesting the need for new reference
ranges. In the paediatric cohort, Roche Elecsys anti-Müllerian hormone
levels between boys and girls showed good prepubertal delineation and small
but statistically significant differences to previously measured levels
using the Beckman Coulter Access anti-Müllerian hormone assay on the same
sample cohort.
Collapse
Affiliation(s)
- Allen P Yates
- 1 Department of Clinical Biochemistry, Manchester University NHS Foundation Trust, Manchester, UK
| | - Helen M Jopling
- 1 Department of Clinical Biochemistry, Manchester University NHS Foundation Trust, Manchester, UK
| | | | - Katharine Hayden
- 1 Department of Clinical Biochemistry, Manchester University NHS Foundation Trust, Manchester, UK
| | - Christopher M Chaloner
- 1 Department of Clinical Biochemistry, Manchester University NHS Foundation Trust, Manchester, UK
| | - Lesley Tetlow
- 1 Department of Clinical Biochemistry, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
46
|
Kvernebo Sunnergren K, Ankarberg-Lindgren C, Dahlgren J. Adrenal and Gonadal Activity, Androgen Concentrations, and Adult Height Outcomes in Boys With Silver-Russell Syndrome. Front Endocrinol (Lausanne) 2019; 10:829. [PMID: 31920957 PMCID: PMC6914679 DOI: 10.3389/fendo.2019.00829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 11/13/2019] [Indexed: 11/24/2022] Open
Abstract
Background: We have previously shown that adult height (AH) in males with Silver-Russell syndrome (SRS) correlated negatively with prepubertal estradiol concentrations. We aimed to identify the source of estradiol by analyzing androgen secretion profiles and measuring anti-Müllerian hormone (AMH) and inhibin B concentrations during childhood and puberty in this group of patients. Methods: In a retrospective longitudinal single-center study, 13 males with SRS were classified as non-responders (NRs = 8) or responders (Rs = 5), depending on the AH outcome. From 6 years of age, androgens were determined by mass spectrometry, and AMH, inhibin B and sex hormone-binding globulin concentrations were analyzed by immunoassays. Results: AH outcome correlated negatively with dehydroepiandrosterone-sulfate (DHEAS) at 8 (r = -0.72), 10 (r = -0.79), and 12 years (r = -0.72); testosterone at 10 (r = -0.94), 12 (r = -0.70) and 14 years (r = -0.64); dihydrotestosterone (DHT) at 10 (r = -0.62) and 12 years; (r = -0.57) and AMH at 12 years (r = 0.62) of age. Compared with Rs, NRs had higher median concentrations of DHEAS (μmol/L) at 10 years (2.9 vs. 1.0); androstenedione (nmol/L) at 10 (1.1 vs. 0.6) and 12 years (1.7 vs. 0.8); testosterone (nmol/L) at 10 (0.3 vs. 0.1), 12 (7.8 vs. 0.2) and 14 years (15.6 vs. 10.4); and DHT (pmol/L) at 10 (122 vs. 28) and 12 years (652 vs. 59) of age. AMH (ng/mL) was lower in NRs than in Rs at 12 years of age (11 vs. 50). No significant differences were observed in the inhibin B concentrations at any age. Conclusions: The elevated androgen concentrations before and during puberty, originated from both adrenal and gonadal secretion and correlated negatively with AH outcomes in males with SRS.
Collapse
Affiliation(s)
- Kjersti Kvernebo Sunnergren
- Göteborg Pediatric Growth Research Center (GP-GRC), Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, Ryhov County Hospital, Jönköping, Sweden
| | - Carina Ankarberg-Lindgren
- Göteborg Pediatric Growth Research Center (GP-GRC), Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jovanna Dahlgren
- Göteborg Pediatric Growth Research Center (GP-GRC), Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Endocrinology, Sahlgrenska University Hospital, Queen Silva Hospital, Gothenburg, Sweden
| |
Collapse
|
47
|
Rodprasert W, Virtanen HE, Mäkelä JA, Toppari J. Hypogonadism and Cryptorchidism. Front Endocrinol (Lausanne) 2019; 10:906. [PMID: 32010061 PMCID: PMC6974459 DOI: 10.3389/fendo.2019.00906] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 12/11/2019] [Indexed: 01/24/2023] Open
Abstract
Congenital cryptorchidism (undescended testis) is one of the most common congenital urogenital malformations in boys. Prevalence of cryptorchidism at birth among boys born with normal birth weight ranges from 1.8 to 8.4%. Cryptorchidism is associated with a risk of low semen quality and an increased risk of testicular germ cell tumors. Testicular hormones, androgens and insulin-like peptide 3 (INSL3), have an essential role in the process of testicular descent from intra-abdominal position into the scrotum in fetal life. This explains the increased prevalence of cryptorchidism among boys with diseases or syndromes associated with congenitally decreased secretion or action of androgens, such as patients with congenital hypogonadism and partial androgen insensitivity syndrome. There is evidence to support that cryptorchidism is associated with decreased testicular hormone production later in life. It has been shown that cryptorchidism impairs long-term Sertoli cell function, but may also affect Leydig cells. Germ cell loss taking place in the cryptorchid testis is proportional to the duration of the condition, and therefore early orchiopexy to bring the testis into the scrotum is the standard treatment. However, the evidence for benefits of early orchiopexy for testicular endocrine function is controversial. The hormonal treatments using human chorionic gonadotropin (hCG) or gonadotropin-releasing hormone (GnRH) to induce testicular descent have low success rates, and therefore they are not recommended by the current guidelines for management of cryptorchidism. However, more research is needed to assess the effects of hormonal treatments during infancy on future male reproductive health.
Collapse
Affiliation(s)
- Wiwat Rodprasert
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- The Population Research Centre, University of Turku, Turku, Finland
- *Correspondence: Wiwat Rodprasert
| | - Helena E. Virtanen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- The Population Research Centre, University of Turku, Turku, Finland
| | - Juho-Antti Mäkelä
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- The Population Research Centre, University of Turku, Turku, Finland
| | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- The Population Research Centre, University of Turku, Turku, Finland
- Department of Pediatrics, Turku University Hospital, Turku, Finland
| |
Collapse
|
48
|
Çetinkaya M, Özen S, Uslu S, Gönç N, Acunas B, Akıncı A, Satar M, Berberoğlu M. Diagnostic and therapeutic approach in newborns with ambiguous genitale with disorder of sex development: consensus report of Turkish Neonatal and Pediatric Endocrinology and Diabetes Societies. TURK PEDIATRI ARSIVI 2018; 53:S198-S208. [PMID: 31236033 PMCID: PMC6568300 DOI: 10.5152/turkpediatriars.2018.01818] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Disorders of sex development are defined as conditions in which the chromosomal, gonadal, and anatomic sex is discordant. Patients usually present with atypical appearing genitalia. In the assessment of neonates with disorders of sex development, first, it is important to determine whether this situation requires prompt evaluation, and then the karyotype, hormone levels, and underlying etiology should be determined as soon as possible. All these procedures should be performed in the guidance of a multidisciplinary team in reference centers. As the physical examination of the infant is extremely important, the physcian should suspect and then perform a detailed history and physical examinationi and lastly plan the required laboratory and imaging procedures for the definite diagnosis. It is important not to be hurried in the choice of sex. The aim of this article, which includes the diagnostic and therapeutic approaches in infants with ambiguous genitalia, was to provide a common practice for all pediatricians.
Collapse
Affiliation(s)
- Merih Çetinkaya
- Division of Neonatology, Health Sicences University, İstanbul Kanuni Sultan Süleyman Training and Research Hospital, İstanbul, Turkey
| | - Samim Özen
- Division of Pediatric Endocrinology, Department of Pediatrics, Ege University, Faculty of Medicine, İzmir, Turkey
| | - Sinan Uslu
- Division of Neonatology Health Sicences University, İstanbul Şişli Hamidiye Etfal Training and Research Hospital, İstanbul, Turkey
| | - Nazlı Gönç
- Division of Pediatric Endocrinology, Department of Pediatrics, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Betül Acunas
- Division of Neonatology, Department of Pediatrics, Trakya University, Faculty of Medicine, Edirne, Turkey
| | - Ayşehan Akıncı
- Division of Pediatric Endocrinology, Department of Pediatrics, İnönü University, Faculty of Medicine, Malatya, Turkey
| | - Mehmet Satar
- Division of Neonatology, Department of Pediatrics, Çukurova University, Faculty of Medicine, Adana, Turkey
| | - Merih Berberoğlu
- Division of Pediatric Endocrinology, Department of Pediatrics, Ankara University, Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
49
|
Unal E, Yıldırım R, Tekin S, Demir V, Onay H, Haspolat YK. A Novel Mutation of AMHR2 in Two Siblings with Persistent Müllerian Duct Syndrome. J Clin Res Pediatr Endocrinol 2018; 10:387-390. [PMID: 29687786 PMCID: PMC6280330 DOI: 10.4274/jcrpe.0013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Persistent Müllerian Duct syndrome (PMDS) develops due to deficiency of anti-Müllerian hormone (AMH) or insensitivity of target organs to AMH in individuals with 46,XY karyotype. PMDS is characterized by normal male phenotype of external genitals, associated with persistence of Müllerian structures. This report includes the presentation of a 2.5 year old male patient due to bilateral undescended testis. His karyotype was 46,XY. The increase in testosterone following human chorionic gonadotropin stimulation test was normal. The patient was referred to our clinic after uterine, fallopian tube and vaginal remnants were recognized during the orchiopexy surgery. The family reported that the eight year old elder brother of the patient was operated on for right inguinal hernia and left undescended testis at the age of one year. A right transverse testicular ectopia was found in the elder brother. Both cases had normal AMH levels. AMHR2 gene was analyzed and a homozygous NM_020547.3:c.233-1G>A mutation was found that was not identified previously. In conclusion, we determined a novel mutation in the AMHR2 gene that was identified for the first time. This presented with different phenotypes in two siblings.
Collapse
Affiliation(s)
- Edip Unal
- Dicle University Faculty of Medicine, Department of Pediatric Endocrinology, Diyarbakır, Turkey,* Address for Correspondence: Dicle University Faculty of Medicine, Department of Pediatric Endocrinology, Diyarbakır, Turkey Phone: +90 412 248 80 01 E-mail:
| | - Ruken Yıldırım
- Diyarbakır Children’s Hospital, Clinic of Pediatric Endocrinology, Diyarbakır, Turkey
| | - Suat Tekin
- Dicle University Faculty of Medicine, Department of Pediatrics, Diyarbakır, Turkey
| | | | - Hüseyin Onay
- Ege University Faculty of Medicine, Department of Medical Genetics, İzmir, Turkey
| | - Yusuf Kenan Haspolat
- Dicle University Faculty of Medicine, Department of Pediatric Endocrinology, Diyarbakır, Turkey
| |
Collapse
|
50
|
Cheon KY, Chung YJ, Cho HH, Kim MR, Cha JH, Kang CS, Lee JY, Kim JH. Expression of Müllerian-Inhibiting Substance/Anti-Müllerian Hormone Type II Receptor in the Human Theca Cells. J Clin Endocrinol Metab 2018; 103:3376-3385. [PMID: 29947765 DOI: 10.1210/jc.2018-00549] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/21/2018] [Indexed: 02/13/2023]
Abstract
CONTEXT Müllerian-inhibiting substance/anti-Müllerian hormone (MIS/AMH) is produced in the ovarian granulosa cells, and it is believed to inhibit ovarian folliculogenesis and steroidogenesis in women of reproductive age. OBJECTIVE To investigate the expression of MIS/AMH type II receptor (MISRII/AMHRII) that binds MIS/AMH in the ovaries of reproductive-age women; to identify the exact targets of MIS/AMH. DESIGN Laboratory study using human ovarian tissue. SETTING University hospital. PATIENTS Tissue samples from 25 patients who had undergone ovarian surgery. INTERVENTIONS The segregation of ovarian granulosa and theca cells by laser microdissection was followed by RT-PCR, analyzing MISRII/AMHRII mRNA expression. Afterward, in situ hybridization and immunohistochemistry were performed to determine the localization of MISRII/AMHRII mRNA and protein expression. MAIN OUTCOME MEASURES MISRII/AMHRII mRNA expression by RT-PCR, in situ hybridization, and immunohistochemistry. RESULTS MISRII/AMHRII were expressed in granulosa and theca cells of preantral and antral follicles. The granulosa cells showed stronger MISRII/AMHRII expression than theca cells. MISRII/AMHRII mRNA staining of granulosa and theca cells in large antral follicles, early atretic follicles, and corpus luteum waned but were still detected weakly, showing higher expression in theca cells than in granulosa cells. However, MISRII/AMHRII protein in the granulosa layer of the atretic follicle and corpus luteum could not be assessed. CONCLUSIONS As MISRII/AMHRII is expressed in both granulosa and theca cells, this indicates that MIS/AMH, produced in the granulosa cells, is active in the theca cells as well. MIS/AMH is most likely actively involved not only in the autocrine and endocrine processes but also in the paracrine processes involving theca cells.
Collapse
Affiliation(s)
- Keun Young Cheon
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Youn Jee Chung
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Hee Cho
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mee Ran Kim
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Ho Cha
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chang Suk Kang
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Young Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jang Heub Kim
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|