1
|
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
|
2
|
Lăptoiu AR, Spoială EL, Stanciu GD, Hanganu E, Lupu VV, Ciongradi CI, Gavrilovici C. New Insights into the Role of INSL-3 in the Development of Cryptorchidism. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10040737. [PMID: 37189986 DOI: 10.3390/children10040737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/09/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023]
Abstract
Cryptorchidism, defined as the failure of at least one or both testicles to descend into the scrotal pouches, is the most frequent (1.6-9% at birth, 1/20 males at birth) congenital anomaly encountered in newborn males, resulting in one of the most frequent causes of non-obstructive azoospermia in men. Similar to other congenital malformations, cryptorchidism is thought to be caused by endocrine and genetic factors, combined with maternal and environmental influences. The etiology of cryptorchidism is unknown, as it involves complex mechanisms aiming to control the testicular development and descent from their initial intra-abdominal location in scrotal pouches. The implication of insulin-like 3 (INSL-3) associated with its receptor (LGR8) is critical. Genetic analysis discloses functionally deleterious mutations in INSL3 and GREAT/LGR8 genes. In this literature review, we discuss and analyze the implication of INSL3 and the INSL3/LGR8 mutation in the occurrence of cryptorchidism in both human and animal models.
Collapse
Affiliation(s)
- Alma-Raluca Lăptoiu
- Pediatrics Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Elena-Lia Spoială
- Pediatrics Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Gabriela Dumitrita Stanciu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Elena Hanganu
- Department of Biomedical Sciences, "Grigore T. Popa" University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
- Department of Pediatric and Orthopaedic Surgery, "Sfânta Maria" Emergency Children Hospital, 700309 Iași, Romania
| | - Vasile Valeriu Lupu
- Pediatrics Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Carmen-Iulia Ciongradi
- Department of Pediatric and Orthopaedic Surgery, "Sfânta Maria" Emergency Children Hospital, 700309 Iași, Romania
- 2nd Department of Surgery and Ortophedics, "Grigore T. Popa" University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Cristina Gavrilovici
- Pediatrics Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
3
|
Reny SE, Mukherjee A, Mol PM. The curious case of testicular descent: factors controlling testicular descent with a note on cryptorchidism. AFRICAN JOURNAL OF UROLOGY 2023. [DOI: 10.1186/s12301-023-00342-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
Abstract
Background
The testicular descent is a uniquely complex process depending upon multiple factors like growth and reorganisation of the specific gonadal ligaments, hormones, etc., which interplay with each other. Though an unambiguous event, it is still laced with incredulity since the data interpretation were intermingled between different species creating more ambiguity in certain aspects of this process. In order to understand the aetiopathology of cryptorchidism the extensive study of the factors controlling the descent is necessitous.
Main body
Though testes originate in the abdomen, they migrate to an extra abdominal site the scrotum, which makes it vulnerable to pathological conditions associated with the descent. The hormones that play vital role in the first phase of descent are insulin-like hormone 3 (INSL3), Anti-müllerian hormone as well as testosterone, whereas androgens, genitofemoral nerve and its neurotransmitter calcitonin gene-related peptide (CGRP) influence the second phase. Despite the vast research regarding the complex nexus of events involving the descent there are disparities among the cross species studies. However all these discrepancies make testicular descent yet again fascinating and perplexing. Our aim is to provide a comprehensive review including recent advances which provides thorough coverage of anatomical and hormonal factors in the descent as well as cryptorchidism.
Conclusion
Though our understanding on testicular descent has evolved over the decades there still has obscurity surrounding it and the studies on the factors responsible for descent are becoming more intense with the time. Our knowledge on many factors such as INSL3 and CGRP is more established now; however, on the other hand the role of androgens still remains speculative. As the knowledge and understanding of the biological process of testicular descent increases it will pave ways to new treatment plans to treat cryptorchidism more effectively.
Collapse
|
4
|
Ivell R, Mamsen LS, Andersen CY, Anand-Ivell R. Expression and Role of INSL3 in the Fetal Testis. Front Endocrinol (Lausanne) 2022; 13:868313. [PMID: 35464060 PMCID: PMC9019166 DOI: 10.3389/fendo.2022.868313] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022] Open
Abstract
Insulin-like peptide 3 (INSL3) is a small peptide hormone of the insulin-relaxin family which is produced and secreted by the fetal Leydig cells in the testes only. It appears to be undetectable in female fetuses. In the human fetus INSL3 synthesis begins immediately following gonadal sex determination at weeks 7 to 8 post coitum and the peptide can be detected in amniotic fluid 1 to 2 weeks later. INSL3 acts through a unique G-protein-coupled receptor, called RelaXin-like Family Peptide receptor 2 (RXFP2), which is expressed by the mesenchymal cells of the gubernacular ligament linking the testes to the inguinal wall. The role of INSL3 in the male fetus is to cause a thickening of the gubernaculum which then retains the testes in the inguinal region, while the remainder of the abdominal organs grow away in an antero-dorsal direction. This represents the first phase of testis descent and is followed later in pregnancy by the second inguino-scrotal phase whereby the testes pass into the scrotum through the inguinal canal. INSL3 acts as a significant biomarker for Leydig cell differentiation in the fetus and may be reduced by maternal exposure to endocrine disrupting chemicals, such as xenoestrogens or phthalates, leading to cryptorchidism. INSL3 may have other roles within the fetus, but as a Leydig cell biomarker its reduction acts also as a surrogate for anti-androgen action.
Collapse
Affiliation(s)
- Richard Ivell
- School of Bioscience, University of Nottingham, Sutton Bonington, United Kingdom
| | - Linn Salto Mamsen
- Laboratory of Reproductive Biology, Section 5712, Juliane Marie Centre for Women, Children and Reproduction, Rigshospitalet, University Hospital of Copenhagen, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, Section 5712, Juliane Marie Centre for Women, Children and Reproduction, Rigshospitalet, University Hospital of Copenhagen, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ravinder Anand-Ivell
- School of Bioscience, University of Nottingham, Sutton Bonington, United Kingdom
| |
Collapse
|
5
|
Shokri S, Tavalaee M, Ebrahimi SM, Ziaeipour S, Nasr-Esfahani MH, Nejatbakhsh R. Expression of RXFP2 receptor on human spermatozoa and the anti-apoptotic and antioxidant effects of insulin-like factor 3. Andrologia 2020; 52:e13715. [PMID: 32557760 DOI: 10.1111/and.13715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 01/16/2023] Open
Abstract
Insulin-like factor 3 (INSL3) has an important role in the human reproductive system; however, its detailed function is still mysterious. We aimed to investigate the possibility of expression of RXFP2 receptor on human spermatozoa and to determine the anti-apoptotic and antioxidant mechanism derived the binding of INSL3 and RXFP2. In this experimental study, the expression/location of the RXFP2 receptor was determined on the spermatozoa of fertile and infertile men. Twenty samples from 20 fertile men were collected and divided into 6 parts (control group, and five groups treated with INSL3 10, 100, 250, 500, 1,000 ng/ml). DNA damage, active caspase, reactive oxygen species (ROS) and sperm parameters were evaluated by TUNEL, flow cytometry, optical microscope and computer-assisted sperm analysis. The expression of RXFP2 was confirmed by Western blot. Immunocytochemistry illustrated that this receptor is expressed in the posterior half of the spermatozoa's head. The INSL3 at concentrations of 500 and 1,000 ng/ml reduced the active caspase and mitochondrial ROS, and also reduced DNA fragmentation at 1,000 ng/ml. Besides, INSL3 500 and 1,000 ng/ml significantly increased the sperm motility. This study confirmed the presence of RXFP2 receptor in fertile and infertile men's spermatozoa, indicating the highly dose-dependent efficacy of the INSL3, which may have promising impacts on the in-vitro fertilisation outcomes.
Collapse
Affiliation(s)
- Saeed Shokri
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Marziyeh Tavalaee
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Seyyed Meisam Ebrahimi
- Department of Medical Surgical Nursing, Abhar School of Nursing, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Sanaz Ziaeipour
- Department of Anatomical Sciences, School of Medicine, Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Reza Nejatbakhsh
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
6
|
Kanamori M, Oikawa K, Tanemura K, Hara K. Mammalian germ cell migration during development, growth, and homeostasis. Reprod Med Biol 2019; 18:247-255. [PMID: 31312103 PMCID: PMC6613016 DOI: 10.1002/rmb2.12283] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Germ cells represent one of the typical cell types that moves over a long period of time and large distance within the animal body. To continue its life cycle, germ cells must migrate to spatially distinct locations for proper development. Defects in such migration processes can result in infertility. Thus, for more than a century, the principles of germ cell migration have been a focus of interest in the field of reproductive biology. METHODS Based on published reports (mainly from rodents), investigations of germ cell migration before releasing from the body, including primordial germ cells (PGCs), gonocytes, spermatogonia, and immature spermatozoon, were summarized. MAIN FINDINGS Germ cells migrate with various patterns, with each migration step regulated by distinct mechanisms. During development, PGCs actively and passively migrate from the extraembryonic region toward genital ridges through the hindgut epithelium. After sex determination, male germline cells migrate heterogeneously in a developmental stage-dependent manner within the testis. CONCLUSION During migration, there are multiple gates that disallow germ cells from re-entering the proper developmental pathway after wandering off the original migration path. The presence of gates may ensure the robustness of germ cell development during development, growth, and homeostasis.
Collapse
Affiliation(s)
- Mizuho Kanamori
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural ScienceTohoku UniversitySendaiJapan
| | - Kenta Oikawa
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural ScienceTohoku UniversitySendaiJapan
| | - Kentaro Tanemura
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural ScienceTohoku UniversitySendaiJapan
| | - Kenshiro Hara
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural ScienceTohoku UniversitySendaiJapan
| |
Collapse
|
7
|
Li X, Mo J, Zhu Q, Ni C, Wang Y, Li H, Lin ZK, Ge RS. The structure-activity relationship (SAR) for phthalate-mediated developmental and reproductive toxicity in males. CHEMOSPHERE 2019; 223:504-513. [PMID: 30784757 DOI: 10.1016/j.chemosphere.2019.02.090] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/07/2019] [Accepted: 02/14/2019] [Indexed: 06/09/2023]
Abstract
Testicular dysgenesis syndrome includes the hypospadias, cryptorchidism and abnormal fetal testis in male neonate. This is possibly caused by the environmental phthalates, which down-regulate the expression of androgen synthetic genes and Insl3 or directly inhibits steroidogenic enzymes. There are distinct structure-activity relationships (SARs) for phthalate-mediated developmental and reproductive toxicity. Here, we review the SAR for phthalate-mediated testicular dysgenesis syndrome. Of phthalates of straight side chains, C5-C6 ones are the most potent, C4 or C7 are moderate, C3 is weakest, and C1-2 or C8-13 are ineffective. The branching and unsaturation of side chains increases the toxicity. The cycling of side chains does not increase the toxicity.
Collapse
Affiliation(s)
- Xiaoheng Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiaying Mo
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiqi Zhu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chaobo Ni
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiyan Wang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huitao Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhen-Kun Lin
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ren-Shan Ge
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
8
|
Sharma V, Lehmann T, Stuckas H, Funke L, Hiller M. Loss of RXFP2 and INSL3 genes in Afrotheria shows that testicular descent is the ancestral condition in placental mammals. PLoS Biol 2018; 16:e2005293. [PMID: 29953435 PMCID: PMC6023123 DOI: 10.1371/journal.pbio.2005293] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 05/24/2018] [Indexed: 01/13/2023] Open
Abstract
Descent of testes from a position near the kidneys into the lower abdomen or into the scrotum is an important developmental process that occurs in all placental mammals, with the exception of five afrotherian lineages. Since soft-tissue structures like testes are not preserved in the fossil record and since key parts of the placental mammal phylogeny remain controversial, it has been debated whether testicular descent is the ancestral or derived condition in placental mammals. To resolve this debate, we used genomic data of 71 mammalian species and analyzed the evolution of two key genes (relaxin/insulin-like family peptide receptor 2 [RXFP2] and insulin-like 3 [INSL3]) that induce the development of the gubernaculum, the ligament that is crucial for testicular descent. We show that both RXFP2 and INSL3 are lost or nonfunctional exclusively in four afrotherians (tenrec, cape elephant shrew, cape golden mole, and manatee) that completely lack testicular descent. The presence of remnants of once functional orthologs of both genes in these afrotherian species shows that these gene losses happened after the split from the placental mammal ancestor. These “molecular vestiges” provide strong evidence that testicular descent is the ancestral condition, irrespective of persisting phylogenetic discrepancies. Furthermore, the absence of shared gene-inactivating mutations and our estimates that the loss of RXFP2 happened at different time points strongly suggest that testicular descent was lost independently in Afrotheria. Our results provide a molecular mechanism that explains the loss of testicular descent in afrotherians and, more generally, highlight how molecular vestiges can provide insights into the evolution of soft-tissue characters. While fossils of whales with legs demonstrate that these species evolved from legged ancestors, the ancestral state of nonfossilizing soft-tissue structures can only be indirectly inferred. This difficulty is also confounded by uncertainties in the phylogenetic relationships between the animals concerned. A prime example is the case of testicular descent, a developmental process that determines the final position of testes, which occurs in most placental mammals but is absent from several afrotherian lineages. Here, we discovered that afrotherians possess remnants of genes known to be required for testicular descent. These “molecular vestiges” show that testicular descent was already present in the placental ancestor and was subsequently lost in Afrotheria. Our study highlights the potential of molecular vestiges in resolving contradictory ancestral states of soft-tissue characters.
Collapse
Affiliation(s)
- Virag Sharma
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
- Center for Systems Biology Dresden, Germany
| | - Thomas Lehmann
- Senckenberg Research Institute and Natural History Museum Frankfurt, Frankfurt am Main, Germany
| | | | - Liane Funke
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Michael Hiller
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
- Center for Systems Biology Dresden, Germany
- * E-mail:
| |
Collapse
|
9
|
The Role of the Appendix Testis in Normal Testicular Descent: Is There a Connection? BIOMED RESEARCH INTERNATIONAL 2018; 2018:3078031. [PMID: 29850503 PMCID: PMC5937597 DOI: 10.1155/2018/3078031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/24/2018] [Accepted: 03/18/2018] [Indexed: 11/17/2022]
Abstract
Objective The presence of testicular appendices was prospectively evaluated in 89 boys with 96 undescended testes who underwent orchidopexy over the period of 4 years. Results The patients were divided into two groups. Group A included 42 boys with 49 undescended testes positioned close to the internal inguinal ring, and Group B included 47 boys with 47 undescended testes close to the external inguinal ring. The incidence of appendix testis (AT) in Group A was 57.1% (28 in 49) and 78.7% (37 in 47) in Group B. The results of our study showed significantly decreased incidence of testicular appendices in undescended testes positioned close to the internal inguinal ring compared with undescended testes positioned close to the external inguinal ring (p < 0.05). Conclusion AT may play a role in normal testicular descent and the undescended testis positioned close to the external inguinal ring can be considered as a separate entity of the true congenital undescended testis.
Collapse
|
10
|
Morgan JT, Robbins AK, Mateson AB, Sawamoto K, Tomatsu S, Gray DR, Gleghorn JP, Barthold JS. Regional Variation in Androgen Receptor Expression and Biomechanical Properties May Contribute to Cryptorchidism Susceptibility in the LE/orl Rat. Front Endocrinol (Lausanne) 2018; 9:738. [PMID: 30568634 PMCID: PMC6290328 DOI: 10.3389/fendo.2018.00738] [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: 05/01/2018] [Accepted: 11/20/2018] [Indexed: 11/13/2022] Open
Abstract
Background: The process of testicular descent requires androgen and insulin-like 3, hormones secreted by fetal Leydig cells. Knowledge concerning distinct and common functions of these hormones in regulating development of the fetal gubernaculum remains limited and/or conflicting. The current studies were designed to better define characteristics of androgen receptor (AR) expression, function and regulation, as well as the biomechanical properties of normal and cryptorchid gubernaculum during fetal development. Methods: We studied fetal gubernacula from Long Evans outbred (LE/wt) rats and an inbred (LE/orl) strain with an inherited form of cryptorchidism associated with an AR signaling defect. Gubernacular cells or whole organs obtained from LE/wt and LE/orl fetal gubernacula underwent AR immunostaining and quantitative image analysis. The effects of dihydrotestosterone (DHT) on AR expression, muscle fiber morphology, hyaluronan (HA) levels and glycosaminoglycan (GAG) content were measured in LE/wt gubernacula. Finally, the spatial mechanics of freshly harvested LE/wt and LE/orl fetal gubernacula were compared using micropipette aspiration. Results: AR is expressed in the nucleus of mesenchymal core, tip and cord cells of the embryonic (E) day 17 and 21 fetal gubernaculum, and is enhanced by DHT in primary cultures of gubernacular mesenchymal cells. Enhanced AR expression at the tip was observed in LE/wt but not LE/orl gubernacula. In in vitro studies of whole mount fetal gubernaculum, DHT did not alter muscle fiber morphology, HA content or GAG production. Progressive swelling with reduced cellular density of the LE/wt gubernaculum at E19-21 was associated with increased central stiffness in LE/wt but not in LE/orl fetuses. Conclusions: These data confirm nuclear AR expression in gubernacular mesenchyme with distal enhancement at the tip/cord region in LE/wt but not LE/orl rat fetuses. DHT enhanced cellular AR expression but had no major effects on muscle morphology or matrix composition in the rat fetal gubernaculum in vitro. Regional increased stiffness and decreased cell density between E19 and E21 were observed in LE/wt but not LE/orl fetal gubernacula. Developmental differences in cell-specific AR expression in LE/orl fetal gubernacula may contribute to the dysmorphism and aberrant function that underlies cryptorchidism susceptibility in this strain.
Collapse
Affiliation(s)
- Joshua T. Morgan
- Department of Bioengineering, University of California, Riverside, Riverside, CA, United States
- *Correspondence: Joshua T. Morgan
| | - Alan K. Robbins
- Nemours Biomedical Research, Division of Urology, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Abigail B. Mateson
- Nemours Biomedical Research, Division of Urology, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Kazuki Sawamoto
- Department of Orthopedics, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Shunji Tomatsu
- Department of Orthopedics, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Dione R. Gray
- Nemours Biomedical Research, Division of Urology, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Jason P. Gleghorn
- Department of Biomedical Engineering, University of Delaware, Newark, DE, United States
| | - Julia Spencer Barthold
- Nemours Biomedical Research, Division of Urology, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| |
Collapse
|
11
|
Barthold JS, Ivell R. Perspective: A Neuro-Hormonal Systems Approach to Understanding the Complexity of Cryptorchidism Susceptibility. Front Endocrinol (Lausanne) 2018; 9:401. [PMID: 30083133 PMCID: PMC6065160 DOI: 10.3389/fendo.2018.00401] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/28/2018] [Indexed: 12/26/2022] Open
Abstract
Nonsyndromic cryptorchidism is a common multifactorial, condition with long-term risks of subfertility and testicular cancer. Revealing the causes of cryptorchidism will likely improve prediction and prevention of adverse outcomes. Herein we provide our current perspective of cryptorchidism complexity in a synthesis of cumulative clinical and translational data generated by ourselves and others. From our recent comparison of genome-wide association study (GWAS) data of cryptorchidism with or without testicular germ cell tumor, we identified RBFOX family genes as candidate susceptibility loci. Notably, RBFOX proteins regulate production of calcitonin gene-related peptide (CGRP), a sensory neuropeptide linked to testicular descent in animal models. We also re-analyzed existing fetal testis transcriptome data from a rat model of inherited cryptorchidism (the LE/orl strain) for enrichment of Leydig cell progenitor genes. The majority are coordinately downregulated, consistent with known reduced testicular testosterone levels in the LE/orl fetus, and similarly suppressed in the gubernaculum. Using qRT-PCR, we found dysregulation of dorsal root ganglia (DRG) sensory transcripts ipsilateral to undescended testes. These data suggest that LE/orl cryptorchidism is associated with altered signaling in possibly related cell types in the testis and gubernaculum as well as DRG. Complementary rat and human studies thus lead us to propose a multi-level, integrated neuro-hormonal model of testicular descent. Variants in genes encoding RBFOX family proteins and/or their transcriptional targets combined with environmental exposures may disrupt this complex pathway to enhance cryptorchidism susceptibility. We believe that a systems approach is necessary to provide further insight into the causes and consequences of cryptorchidism.
Collapse
Affiliation(s)
- Julia S. Barthold
- Nemours Biomedical Research, Division of Urology, Alfred I. duPont Hospital for Children, Wilmington, DE, United States
- *Correspondence: Julia S. Barthold
| | - Richard Ivell
- School of Biosciences and School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, United Kingdom
| |
Collapse
|
12
|
Hutson JM, Lopez-Marambio FA. The possible role of AMH in shortening the gubernacular cord in testicular descent: A reappraisal of the evidence. J Pediatr Surg 2017; 52:1656-1660. [PMID: 28599968 DOI: 10.1016/j.jpedsurg.2017.05.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 04/12/2017] [Accepted: 05/19/2017] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIM Anti-Müllerian hormone (AMH), also called Müllerian inhibiting substance (MIS), is glycoprotein hormone secreted by the fetal Sertoli cells to regulate regression of the Müllerian ducts, the anlagen of the uterus, fallopian tubes, and upper vagina. After its existence was predicted in 1946 and its isolation and purification in the 1970's, a huge amount of information has been gathered on its molecular biology and function in the last 30-40years. Once thought to be a locally acting factor in the male fetus during sexual differentiation, it is now recognized as an endocrine hormone present in both sexes and with functions throughout life. One of the remaining controversies is the possible role of AMH during fetal testicular descent. In the human with aberrant AMH function, the boy has cryptorchidism with persistent Müllerian duct syndrome (PMDS), where the testes are often intraabdominal and on an abnormally long gubernacular cord. By contrast, in rodent models knockout of the AMH gene does not cause cryptorchidism. METHODS/RESULTS In this review we examined the evidence in the literature for and against a role for AMH in testicular descent and considered the implications of the different anatomy of the gubernacular cord in rodents versus children. CONCLUSION We conclude that AMH may have a role in shortening the gubernacular cord in humans which is concealed in rodent models by differences in anatomy of the gubernacular cord in rodents. The controversy could be resolved by re-examination of the gubernacular cord in boys with PMDS and mice with AMHKO. TYPE OF STUDY Review. LEVEL OF EVIDENCE V.
Collapse
Affiliation(s)
- John M Hutson
- Department of Paediatrics, University of Melbourne, Australia; Urology Department, The Royal Children's Hospital, Melbourne, Australia; F Douglas Stephens Surgical Research Group, Murdoch Childrens Research Institute, Melbourne, Australia.
| | | |
Collapse
|
13
|
Adverse effects of endocrine disruptors on the foetal testis development: focus on the phthalates. Folia Histochem Cytobiol 2016; 47:S67-74. [PMID: 20067897 DOI: 10.2478/v10042-009-0056-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
There are great concerns about the increasing incidence of abnormalities in male reproductive function. Human sperm counts have markedly dropped and the rate of testicular cancer has clearly augmented over the past four decades. Moreover, the prevalence rates of cryptorchidism and hypospadias are also probably increasing. It has been hypothesized that all these adverse trends in male reproduction result from abnormalities in the development of the testis during foetal and neonatal life. Furthermore, many recent epidemiological, clinical and experimental data suggest that these male reproductive disorders could be due to the effects of xenobiotics termed endocrine disruptors, which are becoming more and more concentrated and prevalent in our environment. Among these endocrine disruptors, we chose to focus this review on the phthalates for different reasons: 1) they are widespread in the environment; 2) their concentrations in many human biological fluids have been measured; 3) the experimental data using rodent models suggesting a reprotoxicity are numerous and are the most convincing; 4) their deleterious effects on the in vivo and in vitro development and function of the rat foetal testis have been largely studied; 5) some epidemiological data in humans suggest a reprotoxic effect at environmental concentrations at least during neonatal life. However, the direct effects of phthalates on human foetal testis have never been explored. Thus, as we did for the rat in the 1990s, we recently developed and validated an organ culture system which allows maintenance of the development of the different cell types of human foetal testis. In this system, addition of 10-4 M MEHP (mono-2-ethylhexyl phthalate), the most produced phthalate, had no effect on basal or LH-stimulated production of testosterone, but it reduced the number of germ cells by increasing their apoptosis, without modification of their proliferation. This is the first experimental demonstration that phthalates alter the development of the foetal testis in humans. Using our organotypic culture system, we and others are currently investigating the effect of MEHP in the mouse and the rat, and it will be interesting to compare the results between these species to analyse the relevance of toxicological tests based on rodent models.
Collapse
|
14
|
Barthold JS, Wang Y, Kolon TF, Kollin C, Nordenskjöld A, Olivant Fisher A, Figueroa TE, BaniHani AH, Hagerty JA, Gonzaléz R, Noh PH, Chiavacci RM, Harden KR, Abrams DJ, Kim CE, Li J, Hakonarson H, Devoto M. Pathway analysis supports association of nonsyndromic cryptorchidism with genetic loci linked to cytoskeleton-dependent functions. Hum Reprod 2015; 30:2439-51. [PMID: 26209787 PMCID: PMC4573451 DOI: 10.1093/humrep/dev180] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 06/16/2015] [Accepted: 06/30/2015] [Indexed: 12/30/2022] Open
Abstract
STUDY QUESTION What are the genetic loci that increase susceptibility to nonsyndromic cryptorchidism, or undescended testis? SUMMARY ANSWER A genome-wide association study (GWAS) suggests that susceptibility to cryptorchidism is heterogeneous, with a subset of suggestive signals linked to cytoskeleton-dependent functions and syndromic forms of the disease. WHAT IS KNOWN ALREADY Population studies suggest moderate genetic risk of cryptorchidism and possible maternal and environmental contributions to risk. Previous candidate gene analyses have failed to identify a major associated locus, although variants in insulin-like 3 (INSL3), relaxin/insulin-like family peptide receptor 2 (RXFP2) and other hormonal pathway genes may increase risk in a small percentage of patients. STUDY DESIGN, SIZE, DURATION This is a case-control GWAS of 844 boys with nonsyndromic cryptorchidism and 2718 control subjects without syndromes or genital anomalies, all of European ancestry. PARTICIPANTS/MATERIALS, SETTING, METHODS All boys with cryptorchidism were diagnosed and treated by a pediatric specialist. In the discovery phase, DNA was extracted from tissue or blood samples and genotyping performed using the Illumina HumanHap550 and Human610-Quad (Group 1) or OmniExpress (Group 2) platform. We imputed genotypes genome-wide, and combined single marker association results in meta-analyses for all cases and for secondary subphenotype analyses based on testis position, laterality and age, and defined genome-wide significance as P = 7 × 10(-9) to correct for multiple testing. Selected markers were genotyped in an independent replication group of European cases (n = 298) and controls (n = 324). We used several bioinformatics tools to analyze top (P < 10(-5)) and suggestive (P < 10(-3)) signals for significant enrichment of signaling pathways, cellular functions and custom gene lists after multiple testing correction. MAIN RESULTS AND THE ROLE OF CHANCE In the full analysis, we identified 20 top loci, none reaching genome-wide significance, but one passing this threshold in a subphenotype analysis of proximal testis position (rs55867206, near SH3PXD2B, odds ratio = 2.2 (95% confidence interval 1.7, 2.9), P = 2 × 10(-9)). An additional 127 top loci emerged in at least one secondary analysis, particularly of more severe phenotypes. Cytoskeleton-dependent molecular and cellular functions were prevalent in pathway analysis of suggestive signals, and may implicate loci encoding cytoskeletal proteins that participate in androgen receptor signaling. Genes linked to human syndromic cryptorchidism, including hypogonadotropic hypogonadism, and to hormone-responsive and/or differentially expressed genes in normal and cryptorchid rat gubernaculum, were also significantly overrepresented. No tested marker showed significant replication in an independent population. The results suggest heterogeneous, multilocus and potentially multifactorial susceptibility to nonsyndromic cryptorchidism. LIMITATIONS, REASONS FOR CAUTION The present study failed to identify genome-wide significant markers associated with cryptorchidism that could be replicated in an independent population, so further studies are required to define true positive signals among suggestive loci. WIDER IMPLICATIONS OF THE FINDINGS As the only GWAS to date of nonsyndromic cryptorchidism, these data will provide a basis for future efforts to understand genetic susceptibility to this common reproductive anomaly and the potential for additive risk from environmental exposures. STUDY FUNDING/COMPETING INTERESTS This work was supported by R01HD060769 (the Eunice Kennedy Shriver National Institute for Child Health and Human Development (NICHD)), P20RR20173 (the National Center for Research Resources (NCRR), currently P20GM103464 from the National Institute of General Medical Sciences (NIGMS)), an Institute Development Fund to the Center for Applied Genomics at The Children's Hospital of Philadelphia, and Nemours Biomedical Research. The authors have no competing interests to declare.
Collapse
Affiliation(s)
- Julia Spencer Barthold
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA Division of Urology, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA
| | - Yanping Wang
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA
| | - Thomas F Kolon
- Division of Urology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Claude Kollin
- Department of Women's and Children's Health, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Agneta Nordenskjöld
- Department of Women's and Children's Health, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Alicia Olivant Fisher
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA
| | - T Ernesto Figueroa
- Division of Urology, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA
| | - Ahmad H BaniHani
- Division of Urology, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA
| | - Jennifer A Hagerty
- Division of Urology, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA
| | - Ricardo Gonzaléz
- Division of Urology, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA Present address: Auf der Bult Kinder- und Jugendkrankenhaus, Hannover, Germany
| | - Paul H Noh
- Division of Urology, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA Present address: Division of Pediatric Urology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Rosetta M Chiavacci
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kisha R Harden
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Debra J Abrams
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Cecilia E Kim
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jin Li
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA Division of Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marcella Devoto
- Division of Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA Department of Molecular Medicine, Sapienza University, Rome, Italy
| |
Collapse
|
15
|
Expression of insulin-like factor 3 hormone-receptor system in the reproductive organs of male goats. Cell Tissue Res 2015; 362:407-20. [PMID: 26017634 DOI: 10.1007/s00441-015-2206-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 04/24/2015] [Indexed: 10/23/2022]
Abstract
Relaxin-like factor (RLF), generally known as insulin-like factor 3 (INSL3), is essential for testis descent during fetal development. However, its role in adult males is not fully understood. We investigate the function of INSL3 in male Saanen goats by identifying cell types expressing its receptor, relaxin/insulin-like family peptide receptor (RXFP)2 and by characterizing the developmental expression pattern of INSL3 and RXFP2 and the binding of INSL3 to target cells in the male reproductive system. A highly specific RXFP2 antibody that co-localizes with an anti-FLAG antibody in HEK-293 cells recognizes RXFP2-transcript-expressing cells in the testis. INSL3 and RXFP2 mRNA expression is upregulated in the testis, starting from puberty. INSL3 mRNA and protein expression has been detected in Leydig cells, whereas RXFP2 mRNA and protein localize to Leydig cells, to meiotic and post-meiotic germ cells and to the epithelium and smooth muscle of the cauda epididymis and vas deferens. INSL3 binds to all of these tissues and cell types, with the exception of Leydig cells, in a hormone-specific and saturable manner. These results provide evidence for a functional intra- and extra-testicular INSL3 ligand-receptor system in adult male goats.
Collapse
|
16
|
Genome-wide association study using deregressed breeding values for cryptorchidism and scrotal/inguinal hernia in two pig lines. Genet Sel Evol 2015; 47:18. [PMID: 25886970 PMCID: PMC4367917 DOI: 10.1186/s12711-015-0096-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 01/16/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cryptorchidism and scrotal/inguinal hernia are the most frequent congenital defects in pigs. Identification of genomic regions that control these congenital defects is of great interest to breeding programs, both from an animal welfare point of view as well as for economic reasons. The aim of this genome-wide association study (GWAS) was to identify single nucleotide polymorphisms (SNPs) that are strongly associated with these congenital defects. Genotypes were available for 2570 Large White (LW) and 2272 Landrace (LR) pigs. Breeding values were estimated based on 1 359 765 purebred and crossbred male offspring, using a binary trait animal model. Estimated breeding values were deregressed (DEBV) and taken as the response variable in the GWAS. RESULTS Heritability estimates were equal to 0.26 ± 0.02 for cryptorchidism and to 0.31 ± 0.01 for scrotal/inguinal hernia. Seven and 31 distinct QTL regions were associated with cryptorchidism in the LW and LR datasets, respectively. The top SNP per region explained between 0.96% and 1.10% and between 0.48% and 2.77% of the total variance of cryptorchidism incidence in the LW and LR populations, respectively. Five distinct QTL regions associated with scrotal/inguinal hernia were detected in both LW and LR datasets. The top SNP per region explained between 1.22% and 1.60% and between 1.15% and 1.46% of the total variance of scrotal/inguinal hernia incidence in the LW and LR populations, respectively. For each trait, we identified one overlapping region between the LW and LR datasets, i.e. a region on SSC8 (Sus scrofa chromosome) between 65 and 73 Mb for cryptorchidism and a region on SSC13 between 34 and 37 Mb for scrotal/inguinal hernia. CONCLUSIONS The use of DEBV in combination with a binary trait model was a powerful approach to detect regions associated with difficult traits such as cryptorchidism and scrotal/inguinal hernia that have a low incidence and for which affected animals are generally not available for genotyping. Several novel QTL regions were detected for cryptorchidism and scrotal/inguinal hernia, and for several previously known QTL regions, the confidence interval was narrowed down.
Collapse
|
17
|
Jones BC, Hutson JM. The syndrome of Spigelian hernia and cryptorchidism: a review of paediatric literature. J Pediatr Surg 2015; 50:325-30. [PMID: 25638630 DOI: 10.1016/j.jpedsurg.2014.10.059] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 10/28/2014] [Accepted: 10/29/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND/PURPOSE The purpose of this study is to present a summary of paediatric Spigelian hernia (SH) reported to date, and discuss possible aetiology of SH associated with ipsilateral ectopic testis (SH-ET). METHODS Search of PubMed, Medline, Embase, and CINAHL was performed using keywords "Spigelian hernia". The following were extracted from articles describing paediatric SH: demographics, site and contents of SH, comorbidities, proposed aetiology, presence of ipsilateral inguinal canal (IC) and gubernaculum (G). RESULTS There were 78 patients with 88 hernias (69 male, 19 female), including 55 male (19 left, 22 right, 7 bilateral) and 16 female (5 left, 5 right, 3 bilateral) nontraumatic SHs. In nontraumatic male SH, 29 hernias contained testis (10 left, 11 right, 4 bilateral), 15 did not, 10 had no data. Of 29 SH-EH, 15 were lacking IC and G, 3 were missing IC (no G data) and 2 had absent G (no IC data). The combination of SH and cryptorchidism is increasingly recognised as a distinct syndrome. However, there is controversy as to the pathogenic mechanism of this association. We hypothesise SH-ET develops because the G, and therefore IC and line of descent, become cranially 'mislocated' along the mammary line, which overlies the Spigelian fascia. CONCLUSION SH is rare in children. SH-ET may result by testicular descent to an ectopic site along the embryological mammary line.
Collapse
Affiliation(s)
- Brendan C Jones
- F. Douglas Stephens Surgical Research Laboratory, Murdoch Children's Research Institute, Flemington Road, Parkville, Victoria, Australia, 3052; Department of Urology, Royal Children's Hospital, Flemington Road, Parkville, Victoria, Australia, 3052.
| | - John M Hutson
- F. Douglas Stephens Surgical Research Laboratory, Murdoch Children's Research Institute, Flemington Road, Parkville, Victoria, Australia, 3052; Department of Urology, Royal Children's Hospital, Flemington Road, Parkville, Victoria, Australia, 3052.
| |
Collapse
|
18
|
Duan S, Zhang X, Jiang X, Xie L, Sun Z, Ma S, Li J. An insight into insulin-like factor 3 regulate its receptor RXFP2 in mouse gubernaculum testis cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:14806-11. [PMID: 26823808 PMCID: PMC4713594 DOI: pmid/26823808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 10/21/2015] [Indexed: 02/05/2023]
Abstract
The etiology of testicular dysgenesis syndrome is multifactorial and involves abnormalities in the anatomical structures and endocrine factors. Several studies have shown that the abnormal development of the gubernaculum may affect testicular descent, and the insulin-like factor 3 (INSL3) appears to play an important role in development of the gubernaculum have been proved. INSL3 binds its specific receptor (Relaxin family peptide 2, RXFP2), which was highly expressed in gubernaculum, to produce a crucial effect in the first transabdominal descent stage, but its mechanism still remain unclear. In this study, in order to explore how does INSL3 regulate its receptor RXFP2, we cultured mouse gubernaculum testis cells in vitro, which was treated by INSL3, and examined the expression of RXFP2 in mouse gubernaculum testis cells. The results displayed that INSL3 changed RXFP2 expression, and we found that low dose INSL3 can increase RXFP2 expression, the mechanism of above-mentioned might be related with the hormesis of INSL3.
Collapse
Affiliation(s)
- Shouxing Duan
- Department of Pediatric Surgery, The Second Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong, China
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong, China
- Guangdong Key Laboratory of Medical Molecular ImagingShantou, Guangdong, China
| | - Xuan Zhang
- Department of Pediatric Surgery, The Affiliated Maternal and Child Health Hospital of Shenzhen University Medical CollegeShenzhen, Guangdong, China
| | - Xuewu Jiang
- Department of Pediatric Surgery, The Affiliated Maternal and Child Health Hospital of Shenzhen University Medical CollegeShenzhen, Guangdong, China
| | - Lei Xie
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong, China
- Guangdong Key Laboratory of Medical Molecular ImagingShantou, Guangdong, China
| | - Zongbo Sun
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong, China
- Guangdong Key Laboratory of Medical Molecular ImagingShantou, Guangdong, China
| | - Shuhua Ma
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong, China
- Guangdong Key Laboratory of Medical Molecular ImagingShantou, Guangdong, China
| | - Jianhong Li
- Department of Pediatric Surgery, The Second Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong, China
| |
Collapse
|
19
|
Johansen ML, Anand-Ivell R, Mouritsen A, Hagen CP, Mieritz MG, Søeborg T, Johannsen TH, Main KM, Andersson AM, Ivell R, Juul A. Serum levels of insulin-like factor 3, anti-Müllerian hormone, inhibin B, and testosterone during pubertal transition in healthy boys: a longitudinal pilot study. Reproduction 2014; 147:529-35. [DOI: 10.1530/rep-13-0435] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Insulin-like factor 3 (INSL3) is a promising marker of Leydig cell function with potentially high clinical relevance. Limited data of INSL3 levels in relation to other reproductive hormones in healthy pubertal boys exist. In this study, we aimed to evaluate longitudinal serum changes in INSL3 compared with LH, FSH, testosterone, inhibin B, and anti-Müllerian hormone (AMH) during puberty in healthy boys. Ten boys were included from the longitudinal part of the COPENHAGEN Puberty Study. Pubertal evaluation, including testicular volume, was performed and blood samples were drawn every 6 months for 5 years. Serum concentrations of testosterone were determined by a newly developed LC–MS/MS method, and serum concentrations of INSL3, AMH, inhibin B, FSH, and LH respectively were determined by validated immunoassays. The results showed that serum INSL3 levels increased progressively with increasing age, pubertal onset, and testicular volume. In six of the ten boys, LH increased before the first observed increase in INSL3. In the remaining four boys, the increase in LH and INSL3 was observed at the same examination. The increases in serum concentrations of LH, testosterone, and INSL3 were not parallel or in ordered succession and varied interindividually. We demonstrated that INSL3 concentrations were tightly associated with pubertal onset and increasing testicular volume. However, the pubertal increases in LH, INSL3, and testosterone concentrations were not entirely parallel, suggesting that INSL3 and testosterone may be regulated differently. Thus, we speculate that INSL3 provides additional information on Leydig cell differentiation and function during puberty compared with traditional markers of testicular function.
Collapse
|
20
|
Ivell R, Heng K, Anand-Ivell R. Insulin-Like Factor 3 and the HPG Axis in the Male. Front Endocrinol (Lausanne) 2014; 5:6. [PMID: 24478759 PMCID: PMC3902607 DOI: 10.3389/fendo.2014.00006] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/13/2014] [Indexed: 11/30/2022] Open
Abstract
The hypothalamic-pituitary-gonadal (HPG) axis comprises pulsatile GnRH from the hypothalamus impacting on the anterior pituitary to induce expression and release of both LH and FSH into the circulation. These in turn stimulate receptors on testicular Leydig and Sertoli cells, respectively, to promote steroidogenesis and spermatogenesis. Both Leydig and Sertoli cells exhibit negative feedback to the pituitary and/or hypothalamus via their products testosterone and inhibin B, respectively, thereby allowing tight regulation of the HPG axis. In particular, LH exerts both acute control on Leydig cells by influencing steroidogenic enzyme activity, as well as chronic control by impacting on Leydig cell differentiation and gene expression. Insulin-like peptide 3 (INSL3) represents an additional and different endpoint of the HPG axis. This Leydig cell hormone interacts with specific receptors, called RXFP2, on Leydig cells themselves to modulate steroidogenesis, and on male germ cells, probably to synergize with androgen-dependent Sertoli cell products to support spermatogenesis. Unlike testosterone, INSL3 is not acutely regulated by the HPG axis, but is a constitutive product of Leydig cells, which reflects their number and/or differentiation status and their ability therefore to produce various factors including steroids, together this is referred to as Leydig cell functional capacity. Because INSL3 is not subject to the acute episodic fluctuations inherent in the HPG axis itself, it serves as an excellent marker for Leydig cell differentiation and functional capacity, as in puberty, or in monitoring the treatment of hypogonadal patients, and at the same time buffering the HPG output.
Collapse
Affiliation(s)
- Richard Ivell
- School of Molecular and Biomedical Science, University of Adelaide , Adelaide, SA , Australia ; Leibniz Institute for Farm Animal Biology , Dummerstorf , Germany
| | - Kee Heng
- School of Molecular and Biomedical Science, University of Adelaide , Adelaide, SA , Australia
| | | |
Collapse
|
21
|
Barthold JS, Wang Y, Robbins A, Pike J, McDowell E, Johnson KJ, McCahan SM. Transcriptome analysis of the dihydrotestosterone-exposed fetal rat gubernaculum identifies common androgen and insulin-like 3 targets. Biol Reprod 2013; 89:143. [PMID: 24174575 DOI: 10.1095/biolreprod.113.112953] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Androgens and insulin-like 3 (INSL3) are required for development of the fetal gubernaculum and testicular descent. Previous studies suggested that the INSL3-exposed fetal gubernacular transcriptome is enriched for genes involved in neural pathways. In the present study, we profiled the transcriptome of fetal gubernaculum explants exposed to dihydrotestosterone (DHT) and compared this response to that with INSL3. We exposed fetal (Embryonic Day 17) rat gubernacula to DHT for 24 h (10 and 30 nM) or 6 h (1 and 10 nM) in organ culture and analyzed gene expression relative to that of vehicle-treated controls using Affymetrix arrays. Results were annotated using functional, pathway, and promoter analyses and independently validated for selected transcripts using quantitative RT-PCR (qRT-PCR). Transcripts were differentially expressed after 24 h but not 6 h. Most highly overrepresented functional categories included those related to gene expression, skeletal and muscular development and function, and Wnt signaling. Promoter response elements enriched in the DHT-specific transcriptome included consensus sequences for c-ETS1, ELK1, CREB, CRE-BP1/c-June, NRF2, and USF. We observed that 55% of DHT probe sets were also differentially expressed after INSL3 exposure and that the direction of change was the same in 96%. The qRT-PCR results confirmed that DHT increased expression of the INSL3-responsive genes Crlf1 and Chrdl2 but reduced expression of Wnt4. We also validated reduced Tgfb2 and Cxcl12 and increased Slit3 expression following DHT exposure. These data suggest a robust overlap in the DHT- and INSL3-regulated transcriptome that may be mediated in part by CREB signaling and a common Wnt pathway response for both hormones in the fetal gubernaculum.
Collapse
Affiliation(s)
- Julia S Barthold
- Nemours Biomedical Research/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | | | | | | | | | | | | |
Collapse
|
22
|
A genome-wide association study for canine cryptorchidism in Siberian Huskies. J Anim Breed Genet 2013; 131:202-9. [DOI: 10.1111/jbg.12064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 10/16/2013] [Indexed: 12/13/2022]
|
23
|
Hutson JM, Southwell BR, Li R, Lie G, Ismail K, Harisis G, Chen N. The regulation of testicular descent and the effects of cryptorchidism. Endocr Rev 2013; 34:725-52. [PMID: 23666148 DOI: 10.1210/er.2012-1089] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The first half of this review examines the boundary between endocrinology and embryonic development, with the aim of highlighting the way hormones and signaling systems regulate the complex morphological changes to enable the intra-abdominal fetal testes to reach the scrotum. The genitoinguinal ligament, or gubernaculum, first enlarges to hold the testis near the groin, and then it develops limb-bud-like properties and migrates across the pubic region to reach the scrotum. Recent advances show key roles for insulin-like hormone 3 in the first step, with androgen and the genitofemoral nerve involved in the second step. The mammary line may also be involved in initiating the migration. The key events in early postnatal germ cell development are then reviewed because there is mounting evidence for this to be crucial in preventing infertility and malignancy later in life. We review the recent advances in what is known about the etiology of cryptorchidism and summarize the syndromes where a specific molecular cause has been found. Finally, we cover the recent literature on timing of surgery, the issues around acquired cryptorchidism, and the limited role of hormone therapy. We conclude with some observations about the differences between animal models and baby boys with cryptorchidism.
Collapse
Affiliation(s)
- John M Hutson
- Urology Department, Royal Children's Hospital, Parkville 3052, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
24
|
Jelinic M, Leo CH, Post Uiterweer ED, Sandow SL, Gooi JH, Wlodek ME, Conrad KP, Parkington H, Tare M, Parry LJ. Localization of relaxin receptors in arteries and veins, and region-specific increases in compliance and bradykinin-mediated relaxation after in vivo serelaxin treatment. FASEB J 2013; 28:275-87. [PMID: 24036884 DOI: 10.1096/fj.13-233429] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Relaxin is a potent vasodilator of small resistance arteries and modifies arterial compliance in some systemic vascular beds, yet receptors for relaxin, such as RXFP1, have only been localized to vascular smooth muscle. This study first aimed to localize RXFP1 in rat arteries and veins from different organ beds and determine whether receptors are present in endothelial cells. We then tested the hypothesis that region-specific vascular effects of relaxin may be influenced by the cellular localization of RXFP1 within different blood vessels. The aorta, vena cava, mesenteric artery, and vein had significantly higher (P<0.05) RXFP1 immunostaining in endothelial cells compared with vascular smooth muscle, whereas the femoral artery and vein and small pulmonary arteries had higher (P<0.01) RXFP1 immunostaining in the vascular smooth muscle. Male rats were treated subcutaneously with recombinant human relaxin-2 (serelaxin; 4 μg/h) for 5 d; vasodilation and compliance in mesenteric and femoral arteries and veins were compared with placebo controls. Serelaxin significantly (P=0.04) reduced wall stiffness and increased volume compliance in mesenteric arteries but not in the other vessels examined. This was associated with changes in geometrical properties, and not compositional changes in the extracellular matrix. Serelaxin treatment had no effect on acetylcholine-mediated relaxation but significantly (P<0.001) enhanced bradykinin (BK)-mediated relaxation in mesenteric arteries, involving enhanced nitric oxide but not endothelium-derived hyperpolarization or vasodilatory prostanoids. In conclusion, there is differential distribution of RXFP1 on endothelial and smooth muscle across the vasculature. In rats, mesenteric arteries exhibit the greatest functional response to chronic serelaxin treatment.
Collapse
Affiliation(s)
- Maria Jelinic
- 2Department of Zoology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Sasser JM. The emerging role of relaxin as a novel therapeutic pathway in the treatment of chronic kidney disease. Am J Physiol Regul Integr Comp Physiol 2013; 305:R559-65. [PMID: 23883673 DOI: 10.1152/ajpregu.00528.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Emerging evidence supports a potential therapeutic role of relaxin in fibrotic diseases, including chronic kidney disease. Relaxin is a pleiotropic hormone, best characterized for its role in the reproductive system; however, recent studies have demonstrated a role of relaxin in the cardiorenal system. Both relaxin and its receptor, RXFP1, are expressed in the kidney, and relaxin has been shown to play a role in renal vasodilation, in sodium excretion, and as an antifibrotic agent. Together, these findings suggest that the kidney is a target organ of relaxin. Therefore, the purpose of this review is to describe the functional and structural impacts of relaxin treatment on the kidney and to discuss evidence that relaxin prevents disease progression in several experimental models of kidney disease. In addition, this review will present potential mechanisms that are involved in the therapeutic actions of relaxin.
Collapse
Affiliation(s)
- Jennifer M Sasser
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
26
|
Bathgate RAD, Halls ML, van der Westhuizen ET, Callander GE, Kocan M, Summers RJ. Relaxin family peptides and their receptors. Physiol Rev 2013; 93:405-80. [PMID: 23303914 DOI: 10.1152/physrev.00001.2012] [Citation(s) in RCA: 379] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
There are seven relaxin family peptides that are all structurally related to insulin. Relaxin has many roles in female and male reproduction, as a neuropeptide in the central nervous system, as a vasodilator and cardiac stimulant in the cardiovascular system, and as an antifibrotic agent. Insulin-like peptide-3 (INSL3) has clearly defined specialist roles in male and female reproduction, relaxin-3 is primarily a neuropeptide involved in stress and metabolic control, and INSL5 is widely distributed particularly in the gastrointestinal tract. Although they are structurally related to insulin, the relaxin family peptides produce their physiological effects by activating a group of four G protein-coupled receptors (GPCRs), relaxin family peptide receptors 1-4 (RXFP1-4). Relaxin and INSL3 are the cognate ligands for RXFP1 and RXFP2, respectively, that are leucine-rich repeat containing GPCRs. RXFP1 activates a wide spectrum of signaling pathways to generate second messengers that include cAMP and nitric oxide, whereas RXFP2 activates a subset of these pathways. Relaxin-3 and INSL5 are the cognate ligands for RXFP3 and RXFP4 that are closely related to small peptide receptors that when activated inhibit cAMP production and activate MAP kinases. Although there are still many unanswered questions regarding the mode of action of relaxin family peptides, it is clear that they have important physiological roles that could be exploited for therapeutic benefit.
Collapse
Affiliation(s)
- R A D Bathgate
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology, Monash University, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
27
|
Tanwar PS, Commandeur AE, Zhang L, Taketo MM, Teixeira JM. The Müllerian inhibiting substance type 2 receptor suppresses tumorigenesis in testes with sustained β-catenin signaling. Carcinogenesis 2012; 33:2351-61. [PMID: 22962306 DOI: 10.1093/carcin/bgs281] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Dysregulated WNT/β-catenin signaling in murine testes results in a phenotype with complete germ cell loss that resembles human Sertoli cell-only syndrome. In other systems, including the ovary, dysregulated WNT/β-catenin induces tumorigenesis but no tumors are observed in the mutant testes without deletion of a tumor suppressor, such as phosphatase and tensin homolog (PTEN). Müllerian inhibiting substance (MIS, also known as AMH), a member of the transforming growth factor-β family of growth factors responsible for Müllerian duct regression in fetal males, has been shown to inhibit tumor growth in vitro and in vivo but its role as an endogenous tumor suppressor has never been reported. We have deleted the MIS type 2 receptor (MISR2), and thus MIS signaling, in mice with dysregulated WNT/β-catenin and show that these mice develop testicular stromal tumors with 100% penetrance within a few months postnatal. The tumors are highly proliferative and have characteristics of either Sertoli cell tumors or progenitor Leydig cell tumors based on their marker profiles and histology. Phosphorylated Sma and mothers against decapentaplegic-related homolog 1/5/8 is absent in the tumors and β-catenin target genes are induced. The tumor suppressor TP53 is also highly expressed in the tumors, as is phosphorylated γH2AX, which is indicative of DNA damage. The phenotype of these tumors closely resembles those observed when PTEN is also deleted in mice with dysregulated WNT/β-catenin. Tumorigenesis in these mice provides conclusive evidence that physiological MIS signaling is a tumor suppressor mechanism and suggests that targeted treatment of MISR2-expressing cancers with therapeutic MIS should have a beneficial effect on tumor progression.
Collapse
Affiliation(s)
- Pradeep S Tanwar
- Vincent Center for Reproductive Biology, Department of Obstetrics, Gynecology, and Reproductive Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | | | | |
Collapse
|
28
|
Barthold JS, Wang Y, Reilly A, Robbins A, Figueroa TE, Banihani A, Hagerty J, Akins RE. Reduced expression of androgen receptor and myosin heavy chain mRNA in cremaster muscle of boys with nonsyndromic cryptorchidism. J Urol 2012; 188:1411-6. [PMID: 22906643 DOI: 10.1016/j.juro.2012.06.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Indexed: 11/28/2022]
Abstract
PURPOSE To better define the developmental mechanisms of nonsyndromic cryptorchidism, we measured the expression of hormone receptor and muscle type specific mRNAs in target tissues of boys with and those without nonsyndromic cryptorchidism. MATERIALS AND METHODS Prospectively collected cremaster muscle and/or hernia sac tissues from boys with congenital (79) or acquired (66) nonsyndromic cryptorchidism and hernia/hydrocele (controls, 84) were analyzed for hormone receptor (RXFP2, AR, ESR1, ESR2) and myosin heavy chain specific (MYH1, MYH2, MYH7) mRNA expression using real-time reverse transcriptase polymerase chain reaction. Log transformed mRNA, phenotype and feeding history data were statistically analyzed using Pearson's correlation, ANOVA and 2-sample t tests. RESULTS AR mRNA expression was higher in cremaster muscle than in sac tissue, and significantly lower in congenital and acquired nonsyndromic cryptorchidism cases vs controls (p <0.01). Type 1 (slow/cardiac) MYH7 mRNA expression was also significantly reduced in both nonsyndromic cryptorchidism groups (p ≤ 0.002), while a reduction in type 2 (fast) MYH2 expression was more modest and significant only for the congenital cryptorchidism group (p <0.05). Cremasteric MYH7 and AR levels were strongly correlated (r(2) = 0.751, p <0.001). MYH7 and ESR1 mRNA levels were higher and lower, respectively, in boys with nonsyndromic cryptorchidism who were fed soy formula. Expression of other genes was not measurable. CONCLUSIONS Our data suggest that boys with congenital and acquired nonsyndromic cryptorchidism differentially express AR and slow twitch specific MYH7 mRNA in the cremaster muscle, and that MYH7 expression is correlated with AR levels and soy formula use. These differences in gene expression may reflect aberrant hormonal signaling and/or innervation during development with the potential for secondary functional effects and failed testicular descent.
Collapse
Affiliation(s)
- Julia Spencer Barthold
- Surgery/Urology and Nemours Biomedical Research, AI duPont Hospital for Children, Wilmington, Delaware 19803, USA.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Bay K, Andersson AM. Human testicular insulin-like factor 3: in relation to development, reproductive hormones and andrological disorders. ACTA ACUST UNITED AC 2011; 34:97-109. [PMID: 20550598 DOI: 10.1111/j.1365-2605.2010.01074.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Knockout of the gene encoding insulin-like factor 3 (INSL3) results in cryptorchidism in mice due to disruption of the transabdominal phase of testicular descent. This finding was essential for understanding the complete course of testis descensus, and wound up years of speculations regarding the endocrine regulation of this process. INSL3 is, along with testosterone, a major secretory product of testicular Leydig cells. In addition to its crucial function in testicular descent, INSL3 is suggested to play a paracrine role in germ cell survival and an endocrine role in bone metabolism. INSL3 is produced in human prenatal and neonatal, and in adult Leydig cells to various extents, and is in a developmental context regulated like testosterone, with production during second trimester, an early postnatal peak and increasing secretion during puberty, resulting in high adult serum levels. INSL3 production is entirely dependent on the state of Leydig cell differentiation, and is stimulated by the long-term trophic effects mediated by luteinizing hormone (LH). Once differentiated, Leydig cells apparently express INSL3 in a constitutive manner, and the hormone is thereby insensitive to the acute, steroidogenic effects of LH, which for example is an important factor in the regulation of testosterone. Clinically, serum INSL3 levels can turn out to be a usable tool to monitor basal Leydig cell function in patients with various disorders affecting Leydig cell function. According to animal studies, foetal INSL3 production is, directly or indirectly, sensitive to oestrogenic or anti-androgenic compounds. This provides important insight into the mechanism by which maternal exposure to endocrine disrupters can result in cryptorchidism in the next generation. Conclusively, INSL3 is an interesting testicular hormone with potential clinical value as a marker for Leydig cell function. It should be considered on a par with testosterone in the evaluation of testicular function and the consequences of Leydig cell dysfunction.
Collapse
Affiliation(s)
- K Bay
- University Department of Growth and Reproduction, Rigshospitalet, Copenhagen, Denmark.
| | | |
Collapse
|
30
|
Angenard G, Muczynski V, Coffigny H, Duquenne C, Frydman R, Habert R, Livera G, Rouiller-Fabre V. In vitro effects of Uranium on human fetal germ cells. Reprod Toxicol 2011; 31:470-6. [DOI: 10.1016/j.reprotox.2010.12.058] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 11/25/2010] [Accepted: 12/22/2010] [Indexed: 12/21/2022]
|
31
|
Abstract
Complete testicular descent is a sign of, and a prerequisite for, normal testicular function in adult life. The process of testis descent is dependent on gubernacular growth and reorganization, which is regulated by the Leydig cell hormones insulin-like peptide 3 (INSL3) and testosterone. Investigation of the role of INSL3 and its receptor, relaxin-family peptide receptor 2 (RXFP2), has contributed substantially to our understanding of the hormonal control of testicular descent. Cryptorchidism is a common congenital malformation, which is seen in 2-9% of newborn boys, and confers an increased risk of infertility and testicular cancer in adulthood. Although some cases of isolated cryptorchidism in humans can be ascribed to known genetic defects, such as mutations in INSL3 or RXFP2, the cause of cryptorchidism remains unknown in most patients. Several animal and human studies are currently underway to test the hypothesis that in utero factors, including environmental and maternal lifestyle factors, may be involved in the etiology of cryptorchidism. Overall, the etiology of isolated cryptorchidism seems to be complex and multifactorial, involving both genetic and nongenetic components.
Collapse
|
32
|
Altérations environnementales du développement du testicule foetal: zoom sur les phtalates. Basic Clin Androl 2011. [DOI: 10.1007/s12610-011-0121-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Résumé
L’augmentation de plusieurs anomalies de la fonction de reproduction masculine suscite de grandes inquiétudes. Au cours des quatre dernières décennies, le nombre de spermatozoïdes chez l’homme a nettement diminué, et l’incidence du cancer testiculaire a doublé. De plus, les cas de cryptorchidie et d’hypospadias sont également en augmentation. L’hypothèse la plus couramment admise est que tous ces effets néfastes sur la fonction reproductive masculine résulteraient d’anomalies survenant lors du développement du testicule pendant la vie foetale et néonatale. En outre, de nombreuses données épidémiologiques, cliniques et expérimentales suggèrent que ces troubles pourraient être dus aux effets de xénobiotiques appelés perturbateurs endocriniens qui sont de plus en plus concentrés et présents dans notre environnement. Parmi les perturbateurs endocriniens, nous avons choisi de focaliser cette revue sur les phtalates pour diverses raisons: 1) ils sont très répandus dans l’environnement; 2) leurs concentrations dans de nombreux fluides biologiques humains ont été mesurées y compris pendant la grossesse; 3) les données expérimentales utilisant le modèle rat et suggérant une reprotoxicité sont nombreuses et pertinentes; 4) les effets délétères des phtalates sur le développement et sur les fonctions du testicule foetal de rat ont largement été étudiés; 5) quelques données épidémiologiques humaines suggèrent un effet reprotoxique des phtalates aux concentrations retrouvées dans l’environnement, au moins durant la vie néonatale. Cependant, les effets directs des phtalates sur le testicule foetal humain n’avaient jamais été étudiés. Comme nous l’avions fait chez le rat dans les années 1990, nous avons récemment développé et validé un système de culture organotypique de testicule foetal humain qui permet de maintenir in vitro le développement des différents types cellulaires. Dans ce système, l’ajout de 10−4 M de MEHP (mono-2-éthylhexyl phtalate), le phtalate le plus répandu, n’a aucun effet sur la production de testostérone basale ou stimulée par l’hormone lutéinisante (LH), mais il réduit le nombre de cellules germinales en augmentant leur apoptose et sans modifier leur prolifération. Nos données constituent la première donnée expérimentale montrant que les phtalates altèrent le développement du testicule foetal humain. En outre, en utilisant le même système de culture organotypique, il est intéressant de comparer la réponse au MEHP chez l’Homme et chez les rongeurs pour analyser la pertinence des tests toxicologiques basés sur le modèle rongeur.
Collapse
|
33
|
The development and anatomy of the gubernaculum in Hoxa11 knockout mice. J Pediatr Surg 2011; 46:387-92. [PMID: 21292093 DOI: 10.1016/j.jpedsurg.2010.11.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 11/04/2010] [Indexed: 11/23/2022]
Abstract
BACKGROUND The gubernaculum is central to testicular descent, with recent evidence suggesting that it elongates to the scrotum like a limb bud. Homeobox (Hox) genes involved in limb bud outgrowth are expressed within the gubernaculum. Mice with homozygous Hoxa11 gene deletions have bilateral cryptorchidism. This study investigated the precise anatomical effects of Hoxa11 mutation on the mouse gubernaculum. METHODS The pelvises of postnatal mice (n = 46; days 1-10) with Hoxa11 knockout (n = 19), heterozygotes (n = 11), and wild-type (n = 16) mice were serially sectioned and stained with hematoxylin and eosin. Immunohistochemistry was performed for the presence of desmin. RESULTS Hoxa11 mutant mice had intraabdominal cryptorchid testes and highly convoluted vas deferentia. The gubernacular bulbs were abnormal, with no "outgrowth" and persistence of the prenatal "swelling reaction." Desmin immunostaining revealed the lack of undifferentiated mesenchymal cells usually seen as a "swirl" within the bulb and decreased formation of cremaster muscle. CONCLUSIONS Hoxa11 may be involved in forming the growth center seen as the "swirl" of mesenchyme within the gubernacular bulb, consistent with these cells being required for gubernacular elongation during testicular descent. Hoxa11 mutations may well contribute to failure of gubernacular migration in boys with cryptorchidism.
Collapse
|
34
|
Abstract
Cryptorchidism is a very common anomaly of the male genitalia, affecting 2%-4% of male infants and is more common in premature infants. There are two separate stages of testicular descent. The first stage occurs at 8-15 weeks' gestation in the human fetus and is characterized by enlargement of the genito-inguinal ligament, or gubernaculum, and regression of the cranial suspensory ligament. The testis remains close to the future inguinal region as the fetal abdomen grows. Leydig cells in the testis produce insulin-like hormone 3, which stimulates the caudal gubernaculum to grow and become thicker. Mullerian inhibiting substance may have a role in the first phase of descent by stimulating the swelling reaction in the gubernaculum. The second phase of testicular descent requires migration of the gubernaculum and testis from the inguinal region to the scrotum, between 25 and 35 weeks' gestation. The genitofemoral nerve releases calcitonin gene-related peptide, a neurotransmitter that provides a chemotactic gradient to guide migration. The exact cause of cyrptorchidism remains elusive. Information is mainly derived from animal studies (especially in rodents), which may not extrapolate to the human setting. These findings, however, do have some similarities among mammalian species. The current recommended timing for orchidopexy is between 6 and 12 months of life in an effort to preserve the spermatogonia--the stem cells for subsequent spermatogenesis. Despite surgical treatment by orchidopexy, the long-term outcome still remains problematic and controversial. Impaired fertility (33% in unilateral cases and 66% in bilateral undescended testes) and a cancer risk 5-10 times greater than normal is observed over time. Further research into the cause and management of undescended testes is necessary.
Collapse
Affiliation(s)
- John M Hutson
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.
| | | | | | | |
Collapse
|
35
|
Affiliation(s)
- Jacques J Tremblay
- Reproduction, Perinatal and Child Health, CHUQ Research Centre, Laval University, Quebec City, Quebec, Canada.
| |
Collapse
|
36
|
Johnson KJ, Robbins AK, Wang Y, McCahan SM, Chacko JK, Barthold JS. Insulin-like 3 exposure of the fetal rat gubernaculum modulates expression of genes involved in neural pathways. Biol Reprod 2010; 83:774-82. [PMID: 20631401 DOI: 10.1095/biolreprod.110.085175] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Insulin-like 3 (INSL3) signaling directs fetal gubernacular development and testis descent, but the actions of INSL3 in the gubernaculum are poorly understood. Using microarray gene expression profiling of fetal male rat gubernaculum explants exposed to 10 or 100 nM INSL3, significant changes in expression were identified for approximately 900 genes. Several of the genes showing the largest inductions regulate neuronal development or activity, including Pnoc (34-fold), Nptx2 (9-fold), Nfasc (4-fold), Gfra3 (3-fold), Unc5d (3-fold), and Crlf1 (3-fold). Bioinformatics analysis revealed BMP and WNT signaling pathways and several gene ontologies related to neurogenesis were altered by INSL3. Promoter response elements significantly enriched in the INSL3-regulated gene list included consensus sequences for MYB, REL, ATF2, and TEF transcription factors. Comparing in vivo gene expression profiles of male and female rat fetal gubernaculum showed expression of the Bmp, Wnt, and neurodevelopmental genes induced by INSL3 was higher in males. Using quantitative RT-PCR, the microarray data were confirmed, and the induction of Bmp3, Chrdl2, Crlf1, Nptx2, Pnoc, Wnt4, and Wnt5a mRNA levels were examined over a range of INSL3 concentrations (0.1-100 nM) in male and female gubernaculum. In both sexes, an increasing gene expression response was observed between 0.1 and 10 nM INSL3. These data suggest that INSL3 signaling in the fetal gubernaculum induces morphogenetic programs, including BMP and WNT signaling, and support other rodent data suggesting a role for these pathways in development of the gubernaculum.
Collapse
Affiliation(s)
- Kamin J Johnson
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware 19803, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
Cryptorchidism is a common genital anomaly diagnosed at birth or during childhood. Genetic and/or environmental factors that alter expression or function of hormones crucial for testicular descent, insulin-like 3, and testosterone, may contribute to cryptorchidism. When identified at birth, surgical treatment is indicated by 6 months of age if testes fail to descend, or at the time of diagnosis in older children. A laparoscopic approach is preferred for abdominal testes. Early surgical therapy may reduce the risk of subfertility and/or malignancy.
Collapse
|
38
|
Kong RCK, Shilling PJ, Lobb DK, Gooley PR, Bathgate RAD. Membrane receptors: structure and function of the relaxin family peptide receptors. Mol Cell Endocrinol 2010; 320:1-15. [PMID: 20138959 DOI: 10.1016/j.mce.2010.02.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Revised: 12/14/2009] [Accepted: 02/02/2010] [Indexed: 01/17/2023]
Abstract
The receptors for members of the relaxin peptide family have only recently been discovered and are G-protein-coupled receptors (GPCRs). Relaxin and insulin-like peptide 3 (INSL3) interact with the leucine-rich-repeat-containing GPCRs (LGRs) LGR7 and LGR8, respectively. These receptors show closest similarity to the glycoprotein hormone receptors and contain large ectodomains with 10 leucine-rich repeats (LRRs) but are unique members of the LGR family (class C) as they have an LDL class A (LDLa) module at their N-terminus. In contrast, relaxin-3 and INSL5 interact with another class of type I GPCRs which lack a large ectodomain, the peptide receptors GPCR135 and GPCR142, respectively. These receptors are now classified as relaxin family peptide (RXFP) receptors, RXFP1 (LGR7), RXFP2 (LGR8), RXFP3 (GPCR135) and RXFP4 (GPCR142). This review outlines the identification of the peptides and receptors, their expression profiles and physiological roles and the functional interactions of the peptides with their unique receptors.
Collapse
Affiliation(s)
- Roy C K Kong
- Florey Neuroscience Institutes, University of Melbourne, Victoria 3010, Australia
| | | | | | | | | |
Collapse
|
39
|
Angenard G, Muczynski V, Coffigny H, Pairault C, Duquenne C, Frydman R, Habert R, Rouiller-Fabre V, Livera G. Cadmium increases human fetal germ cell apoptosis. ENVIRONMENTAL HEALTH PERSPECTIVES 2010; 118:331-7. [PMID: 20064782 PMCID: PMC2854759 DOI: 10.1289/ehp.0900975] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Accepted: 10/14/2009] [Indexed: 05/08/2023]
Abstract
BACKGROUND Cadmium (Cd) is a common environmental pollutant and a major constituent of tobacco smoke. Adverse effects of this heavy metal on reproductive function have been identified in adults; however, no studies have examined its effects on human reproductive organs during development. OBJECTIVES Using our previously developed organ culture system, we investigated the effects of cadmium chloride on human gonads at the beginning of fetal life, a critical stage in the development of reproductive function. METHODS Human fetal gonads were recovered during the first trimester (711 weeks postconception) and cultured with or without Cd. We used different concentrations of Cd and compared results with those obtained with mouse fetal gonads at similar stages. RESULTS Cd, at concentrations as low as 1 microM, significantly decreased the germ cell density in human fetal ovaries. This correlated with an increase in germ cell apoptosis, but there was no effect on proliferation. Similarly, in the human fetal testis, Cd (1 microM) reduced germ cell number without affecting testosterone secretion. In mouse fetal gonads, Cd increased only female germ cell apoptosis. CONCLUSIONS This is the first experimental demonstration that Cd, at low concentrations, alters the survival of male and female germ cells in humans. Considering data demonstrating extensive human exposure, we believe that current environmental levels of Cd could be deleterious to early gametogenesis.
Collapse
Affiliation(s)
- Gaëlle Angenard
- Laboratory of Differentiation and Radiobiology of the Gonads, CEADSV/iRCM/SCSR, Fontenay aux Roses, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Yuan FP, Li X, Lin J, Schwabe C, Büllesbach EE, Rao CV, Lei ZM. The role of RXFP2 in mediating androgen-induced inguinoscrotal testis descent in LH receptor knockout mice. Reproduction 2010; 139:759-69. [PMID: 20154177 DOI: 10.1530/rep-09-0518] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
LH receptor knockout (LhrKO) male mice exhibit a bilateral cryptorchidism resulting from a developmental defect in the gubernaculum during the inguinoscrotal phase of testis descent, which is corrected by testosterone replacement therapy (TRT). In vivo and in vitro experiments were conducted to investigate the roles of the androgen receptor (AR) and RXFP2 signals in regulation of gubernacular development in LhrKO animals. This study demonstrated that AR and RXFP2 proteins were expressed in the gubernaculum during the entire postnatal period. TRT normalized gubernacular RXFP2 protein levels inLhrKO mice. Organ and primary cell cultures of gubernacula showed that 5alpha-dihydrotestosterone (DHT) upregulated the expression of Rxfp2 which was abolished by the addition of an AR antagonist, flutamide. A single s.c. testosterone injection also led to a significant increase in Rxfp2 mRNA levels in a time-dependent fashion in LhrKO animals. DHT, natural and synthetic insulin-like peptide 3 (INSL3), or relaxin alone did not affect proliferation of gubernacular mesenchymal cells, while co-treatments of DHT with either INSL3 or relaxin resulted in an increase in cell proliferation, and they also enhanced the mesenchymal cell differentiation toward the myogenic pathway, which included a decrease in a mesenchymal cell marker, CD44 and the expression of troponin. These effects were attenuated by the addition of flutamide, siRNA-mediated Rxfp2 knockdown, or by an INSL3 antagonist. Co-administration of an INSL3 antagonist curtailed TRT-induced inguinoscrotal testis descent in LhrKO mice. Our findings indicate that the RXFP2 signaling pathway plays an important role in mediating androgen action to stimulate gubernaculum development during inguinoscrotal testis descent.
Collapse
Affiliation(s)
- F P Yuan
- Department of Obstetrics, Gynecology and Women's Health, School of Medicine, Health Sciences Center, University of Louisville, Louisville, Kentucky 40292, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Arrighi S, Bosi G, Groppetti D, Aralla M, Cremonesi F. An insight into testis and gubernaculum dynamics of INSL3 - RXFP2 signalling during testicular descent in the dog. Reprod Fertil Dev 2010; 22:751-60. [DOI: 10.1071/rd09260] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Accepted: 11/24/2009] [Indexed: 11/23/2022] Open
Abstract
Insulin-like 3 (INSL3) plays a prominent role in male development and is supposed to induce the growth of the gubernaculum testis (g.t.), thus being directly involved in testicular descent in humans and rodents. This happens through activation of the RXFP2 receptor (GREAT or LGR8). The INSL3–RXFP2 complex is reputed to play an additional paracrine role in the testis, possibly acting as part of an autocrine feedback loop. The present work provides evidence of the immunolocalisation of INSL3 in the Leydig cells of canine fetuses and of the expression of RXFP2 receptor in different tissues of the g.t. of the same specimens. RXFP2 was localised at the cell membrane of g.t. muscle and connective cells, as well as in the epithelial cells of the developing excurrent ducts. Notably, RXFP2 immunoreactivity of the g.t. was limited to fetuses at ~35–45 days of gestation, which is also the fetal period when the endocrine compartment of the dog testis is active endocrinologically, as confirmed by the anti-P450c17 and anti-INSL3 immunoreactivities of the fetal Leydig cells, and by anti-Müllerian hormone immunoreactivity of the Sertoli cells. The same immunoreactivities were also evaluated in the testes of cryptorchid dogs of different ages. RXFP2 immunoreactivity was absent from genital tracts of cryptorchid testes and g.t. remnants.
Collapse
|
42
|
Zhao X, Du ZQ, Vukasinovic N, Rodriguez F, Clutter AC, Rothschild MF. Association of HOXA10, ZFPM2, and MMP2 genes with scrotal hernias evaluated via biological candidate gene analyses in pigs. Am J Vet Res 2009; 70:1006-12. [PMID: 19645582 DOI: 10.2460/ajvr.70.8.1006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate the associations between 14 biological candidate genes and scrotal hernias in pigs. ANIMALS 1,534 Pietrain-based pigs, including 692 individuals from 298 pig families and 842 male pigs without family information. PROCEDURES Pigs were classified as affected or unaffected for scrotal hernias. Single nucleotide polymorphisms of candidate genes were analyzed via PCR assays and genotyped. Statistical analyses were performed on the family-trio and the case-control data. RESULTS 2 genes involved in collagen metabolism (homeobox A10 [HOXA10] and matrix metalloproteinases 2 [MMP2]) and 1 gene encoding zinc finger protein multitype 2 (ZFPM2, important in the development of diaphragmatic hernia) were significantly associated with hernias. Pigs with these genotypes had high odds of developing scrotal hernias in the case and control groups (2 ZFPM2 variants: odds ratio, 4.3 [95% confidence interval, 2.78 to 6.64] and 4.45[95%confidenceinterval,2.88to6.88]). Anothergene, collagentypeII A 1(COL2A1),was potentially involved in hernia development. CONCLUSIONS AND CLINICAL RELEVANCE HOXA10, ZFPM2, MMP2, and COL2A1 could have important roles in pig hernia development and potentially be useful for marker-assisted selection in the pig industry. IMPACT FOR HUMAN MEDICINE Pigs are used for the study of many human diseases because of their physiologic similarities. Genes associated with scrotal hernias in this study may be directly used in understanding the molecular mechanisms underlying this defect in humans.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Animal Science and Center for Integrated Animal Genomics, College of Agriculture and Life Sciences, Iowa State University, Ames, IA 50011, USA
| | | | | | | | | | | |
Collapse
|
43
|
Arrighi S, Bosi G, Groppetti D, Aralla M, Cremonesi F. Immunolocalization of INSL3 in dog foetal Leydig cells and the LGR8 receptor in the gubernaculum testis. Vet Res Commun 2009; 33 Suppl 1:67-71. [DOI: 10.1007/s11259-009-9251-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
44
|
Ivell R, Anand-Ivell R. Biology of insulin-like factor 3 in human reproduction. Hum Reprod Update 2009; 15:463-76. [DOI: 10.1093/humupd/dmp011] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
45
|
Lambrot R, Muczynski V, Lécureuil C, Angenard G, Coffigny H, Pairault C, Moison D, Frydman R, Habert R, Rouiller-Fabre V. Phthalates impair germ cell development in the human fetal testis in vitro without change in testosterone production. ENVIRONMENTAL HEALTH PERSPECTIVES 2009; 117:32-7. [PMID: 19165384 PMCID: PMC2627862 DOI: 10.1289/ehp.11146] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Accepted: 09/08/2008] [Indexed: 05/04/2023]
Abstract
BACKGROUND Several studies have described an increasing frequency of male reproductive disorders, which may have a common origin in fetal life and which are hypothesized to be caused by endocrine disruptors. Phthalate esters represent a class of environmental endocrine-active chemicals known to disrupt development of the male reproductive tract by decreasing testosterone production in the fetal rat. OBJECTIVES Using the organ culture system we developed previously, we investigated the effects on the development of human fetal testis of one phthalate--mono-2-ethylhexyl phthalate (MEHP)--an industrial chemical found in many products, which has been incriminated as a disruptor of male reproductive function. METHODS Human fetal testes were recovered during the first trimester (7-12 weeks) of gestation, a critical period for testicular differentiation, and cultured for 3 days with or without MEHP in basal conditions or stimulated with luteinizing hormone (LH). RESULTS Whatever the dose, MEHP treatment had no effect on basal or LH-stimulated testosterone produced by the human fetal testis in vitro, although testosterone production can be modulated in our culture system. MEHP (10(-4) M) did not affect proliferation or apoptosis of Sertoli cells, but it reduced the mRNA expression of anti-Müllerian hormone. MEHP (10(-4) M) reduced the number of germ cells by increasing their apoptosis, measured by the detection of caspase-3-positive germ cells, without modification of their proliferation. CONCLUSIONS This is the first experimental demonstration that phthalates alter the development of the germ cell lineage in humans. However, in contrast to results observed in the rat, phthalates did not affect steroidogenesis.
Collapse
Affiliation(s)
- Romain Lambrot
- Laboratory of Differentiation and Radiobiology of the Gonads, Unit of Gametogenesis and Genotoxicity, Commissariat à l’Energie Atomique, Direction des Sciences du Vivant, Institute of Cellular and Molecular Radiation Biology, Stem Cells and Radiation Department, Fontenay aux Roses, France
- Université Paris Diderot-Paris, Fontenay aux Roses, France
- Unité 566, INSERM, Fontenay aux Roses, France
| | - Vincent Muczynski
- Laboratory of Differentiation and Radiobiology of the Gonads, Unit of Gametogenesis and Genotoxicity, Commissariat à l’Energie Atomique, Direction des Sciences du Vivant, Institute of Cellular and Molecular Radiation Biology, Stem Cells and Radiation Department, Fontenay aux Roses, France
- Université Paris Diderot-Paris, Fontenay aux Roses, France
- Unité 566, INSERM, Fontenay aux Roses, France
| | - Charlotte Lécureuil
- Laboratory of Differentiation and Radiobiology of the Gonads, Unit of Gametogenesis and Genotoxicity, Commissariat à l’Energie Atomique, Direction des Sciences du Vivant, Institute of Cellular and Molecular Radiation Biology, Stem Cells and Radiation Department, Fontenay aux Roses, France
- Université Paris Diderot-Paris, Fontenay aux Roses, France
- Unité 566, INSERM, Fontenay aux Roses, France
| | - Gaëlle Angenard
- Laboratory of Differentiation and Radiobiology of the Gonads, Unit of Gametogenesis and Genotoxicity, Commissariat à l’Energie Atomique, Direction des Sciences du Vivant, Institute of Cellular and Molecular Radiation Biology, Stem Cells and Radiation Department, Fontenay aux Roses, France
- Université Paris Diderot-Paris, Fontenay aux Roses, France
- Unité 566, INSERM, Fontenay aux Roses, France
| | - Hervé Coffigny
- Laboratory of Differentiation and Radiobiology of the Gonads, Unit of Gametogenesis and Genotoxicity, Commissariat à l’Energie Atomique, Direction des Sciences du Vivant, Institute of Cellular and Molecular Radiation Biology, Stem Cells and Radiation Department, Fontenay aux Roses, France
- Université Paris Diderot-Paris, Fontenay aux Roses, France
- Unité 566, INSERM, Fontenay aux Roses, France
| | - Catherine Pairault
- Laboratory of Differentiation and Radiobiology of the Gonads, Unit of Gametogenesis and Genotoxicity, Commissariat à l’Energie Atomique, Direction des Sciences du Vivant, Institute of Cellular and Molecular Radiation Biology, Stem Cells and Radiation Department, Fontenay aux Roses, France
- Université Paris Diderot-Paris, Fontenay aux Roses, France
- Unité 566, INSERM, Fontenay aux Roses, France
| | - Delphine Moison
- Laboratory of Differentiation and Radiobiology of the Gonads, Unit of Gametogenesis and Genotoxicity, Commissariat à l’Energie Atomique, Direction des Sciences du Vivant, Institute of Cellular and Molecular Radiation Biology, Stem Cells and Radiation Department, Fontenay aux Roses, France
- Université Paris Diderot-Paris, Fontenay aux Roses, France
- Unité 566, INSERM, Fontenay aux Roses, France
| | - René Frydman
- Service de Gynécologie-Obstétrique, Université Paris Sud, Hôpital Antoine Béclère, Clamart, France
- Unité 782, INSERM, Clamart, France
| | - René Habert
- Laboratory of Differentiation and Radiobiology of the Gonads, Unit of Gametogenesis and Genotoxicity, Commissariat à l’Energie Atomique, Direction des Sciences du Vivant, Institute of Cellular and Molecular Radiation Biology, Stem Cells and Radiation Department, Fontenay aux Roses, France
- Université Paris Diderot-Paris, Fontenay aux Roses, France
- Unité 566, INSERM, Fontenay aux Roses, France
| | - Virginie Rouiller-Fabre
- Laboratory of Differentiation and Radiobiology of the Gonads, Unit of Gametogenesis and Genotoxicity, Commissariat à l’Energie Atomique, Direction des Sciences du Vivant, Institute of Cellular and Molecular Radiation Biology, Stem Cells and Radiation Department, Fontenay aux Roses, France
- Université Paris Diderot-Paris, Fontenay aux Roses, France
- Unité 566, INSERM, Fontenay aux Roses, France
- Address correspondence to V. Rouiller-Fabre, Unit of Gametogenesis and Genotoxicity, LDRG/SCSR/ iRCM/DSV, Centre CEA, BP6, F-92265, Fontenay aux Roses, France. Telephone: 33-1-46-54-99-23. Fax: 33-1-46-54-99-06. E-mail:
| |
Collapse
|
46
|
Agoulnik AI. Relaxin and related peptides in male reproduction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 612:49-64. [PMID: 18161481 DOI: 10.1007/978-0-387-74672-2_5] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The relaxin hormone is renowned for its function in pregnancy, parturition and other aspects of female reproduction. At the same time, the role of relaxin in male reproduction is still debated. Relaxin is prominently expressed in prostate and its receptors are found in several male reproductive organs; however, the data indicative of its contribution to differentiation and functioning of prostate or testis are contradictory. Prostate relaxin is a main source of this peptide in the seminal plasma. The relaxin effects on sperm motility and fertilization have been reported. The expression of other relaxin related peptides, such as INSL5 and INSL6 was described in testis; yet, currently there are no experimental data to pinpoint their biological functions. The other member of relaxin peptide family, insulin-like 3 peptide (INSL3), is a major player in male development. The INSL3 peptide is expressed in testicular fetal and adult Leydig cells and is directly responsible for the process of abdominal testicular descent (migration of the testes towards the scrotum during male development). Genetic targeting of the Insl3 gene or INSL3 GPCR receptor Lgr8/Rxfp2 causes high intra-abdominal cryptorchidism due to a differentiation failure of testicular ligaments, the gubernacula. Several mutations of these two genes rendering nonfunctional proteins have been described in human patients with testicular maldescent. Thus, in this chapter we review the data related to the expression and function of relaxin and related peptides in male reproduction.
Collapse
Affiliation(s)
- Alexander I Agoulnik
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas 77030, USA.
| |
Collapse
|
47
|
Ribarski I, Lehavi O, Yogev L, Hauser R, Bar-Shira Maymon B, Botchan A, Paz G, Yavetz H, Kleiman SE. USP26 gene variations in fertile and infertile men. Hum Reprod 2008; 24:477-84. [PMID: 18927127 DOI: 10.1093/humrep/den374] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The human X chromosome is enriched with testis-specific genes that may be crucial for male fertility. One is the ubiquitin-specific protease 26 (USP26). Five frequent mutations have been identified: 1737G>A, 1090C > T, 370-371insACA, 494T > C and 1423C>T (with the latter three usually detected in a cluster). Their role in infertility is still controversial. This study assesses the association of the most frequent USP26 mutations with male infertility and male infertility etiology factors. METHODS The study included 300 infertile and 287 fertile men. Data were collected on ethnicity (according to maternal origin) and family history of reproduction. Clinical records from 235 infertile and 62 fertile (sperm bank donors) men were available and summarized. The five mutations were investigated by bioinformatic tools and their frequencies were assessed by restriction analysis. The results were correlated with clinical findings. Segregation of the mutations in four families was analyzed. RESULTS The five analyzed mutations were detected in 44 men from both fertile and infertile groups. The cluster and the 1090C>T mutations showed the highest frequency among Arabs and Sephardic Jews of the infertile group, respectively. Inheritance studies showed that mutations were not always associated with the infertility trait. Mutations 1090C>T and 1737G>A were significantly associated with a history of inguinal hernia (P = 0.007 and P = 0.043, respectively). The prevalence of inguinal hernia among men with the 1090C > T mutation was 33.3% (5/15 men), higher than that reported in infertile men (6.7%). CONCLUSIONS Mutation 1090C > T may be a new genetic risk factor for developing inguinal hernia which may be associated with impaired male fertility.
Collapse
Affiliation(s)
- I Ribarski
- Institute for the Study of Fertility, Lis Maternity Hospital, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, 6 Weizman Street, Tel Aviv, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Foresta C, Zuccarello D, Garolla A, Ferlin A. Role of hormones, genes, and environment in human cryptorchidism. Endocr Rev 2008; 29:560-80. [PMID: 18436703 DOI: 10.1210/er.2007-0042] [Citation(s) in RCA: 152] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cryptorchidism is the most frequent congenital birth defect in male children (2-4% in full-term male births), and it has the potential to impact the health of the human male. In fact, although it is often considered a mild malformation, it represents the best-characterized risk factor for reduced fertility and testicular cancer. Furthermore, some reports have highlighted a significant increase in the prevalence of cryptorchidism over the last few decades. Etiology of cryptorchidism remains for the most part unknown, and cryptorchidism itself might be considered a complex disease. Major regulators of testicular descent from intraabdominal location into the bottom of the scrotum are the Leydig-cell-derived hormones testosterone and insulin-like factor 3. Research on possible genetic causes of cryptorchidism has increased recently. Abundant animal evidence supports a genetic cause, whereas the genetic contribution to human cryptorchidism is being elucidated only recently. Mutations in the gene for insulin-like factor 3 and its receptor and in the androgen receptor gene have been recognized as causes of cryptorchidism in some cases, but some chromosomal alterations, above all the Klinefelter syndrome, are also frequently involved. Environmental factors acting as endocrine disruptors of testicular descent might also contribute to the etiology of cryptorchidism and its increased incidence in recent years. Furthermore, polymorphisms in different genes have recently been investigated as contributing risk factors for cryptorchidism, alone or by influencing susceptibility to endocrine disruptors. Obviously, the interaction of environmental and genetic factors is fundamental, and many aspects have been clarified only recently.
Collapse
Affiliation(s)
- Carlo Foresta
- University of Padova, Department of Histology, Microbiology and Medical Biotechnologies, Section of Clinical Pathology and Centre for Male Gamete Cryopreservation, Via Gabelli 63, 35121 Padova, Italy.
| | | | | | | |
Collapse
|
49
|
One-generation reproductive toxicity study of dietary 17β-estradiol (E2; CAS No. 50-28-2) in CD-1® (Swiss) mice. Reprod Toxicol 2008; 25:144-60. [DOI: 10.1016/j.reprotox.2007.11.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2007] [Revised: 11/16/2007] [Accepted: 11/29/2007] [Indexed: 11/18/2022]
|
50
|
Ferlin A, Zuccarello D, Garolla A, Selice R, Foresta C. Hormonal and genetic control of testicular descent. Reprod Biomed Online 2008; 15:659-65. [PMID: 18062862 DOI: 10.1016/s1472-6483(10)60532-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cryptorchidism has the potential to affect the health of the human male. Although it is often considered a mild malformation, it represents the best-characterized risk factor for reduced fertility and testicular cancer. The aetiology of cryptorchidism remains, for the most part, unknown and cryptorchidism itself might be considered a complex disease. This reflects the intricate mechanisms regulating testicular development and descent from intra-abdominal location into the bottom of the scrotum, involving different anatomical and hormonal factors. Major actors of testicular descent are the Leydig cell-derived hormones testosterone and insulin-like factor 3, even if other factors may play a role. Although considerable evidence exists in animals to support a genetic cause, the genetic contribution to human cryptorchidism is only recently being elucidated. Environmental factors might also contribute to the aetiology of cryptorchidism and its increased incidence in recent years. Mutations in the gene for insulin-like factor 3 and its receptor and in the androgen receptor gene explain a minority of cases of cryptorchidism, but research on genetic polymorphisms that may also influence susceptibility to endocrine disruptors is shedding light on this field.
Collapse
Affiliation(s)
- Alberto Ferlin
- University of Padova, Department of Histology, Microbiology and Medical Biotechnologies, Centre for Male Gamete Cryopreservation, Via Gabelli 63, 35121 Padova, Italy.
| | | | | | | | | |
Collapse
|