1
|
Liu Z, Luo X, Zhang Z, Zhang Q, Wang C, Chen H, Long C, Liu X, Wei G. MAFB-mediated CEBPA regulated human urothelium growth through Wnt/β-catenin signaling pathway. Genes Dis 2025; 12:101432. [PMID: 39569391 PMCID: PMC11577151 DOI: 10.1016/j.gendis.2024.101432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 08/29/2024] [Indexed: 11/22/2024] Open
Abstract
MAFB is essential for regulating male-type urethral differentiation, and especially, its variation can contribute to hypospadias in mice. However, the potential mechanism is still unclear. Here we observed that the basic leucine zipper (bZIP) transcription factor MAFB and CCAAT/enhancer-binding protein alpha (CEBPA) could promote human urothelium SV-HUC-1 growth. Moreover, MAFB and CEBPA expression were reduced in the prepuce tissues of hypospadias patients. Based on transcriptome sequencing analysis and Western blot, MAFB knockdown was found to suppress CEBPA protein expression and repress Wnt/β-catenin signaling in urothelium cells. Meanwhile, we observed blocked cell-cycle progression from the G1 to the S phase, inhibited cell proliferation, and activated apoptosis. Furthermore, MAFB could facilitate CEBPA transcription and regulate the proliferation of urothelium. The above results indicated that MAFB-mediated inhibition of urothelial SV-HUC-1 growth resulted from inhibiting the Wnt/β-catenin signaling pathway by down-regulating CEBPA. Our findings provide new insight into the understanding of genes associated with hypospadias and the pathogenic mechanism of this disorder.
Collapse
Affiliation(s)
- Zhenmin Liu
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Xingguo Luo
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Zhicheng Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Qiang Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Chong Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Hongsong Chen
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Chunlan Long
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Xing Liu
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| |
Collapse
|
2
|
Zhang Y, Wang J, Yang H, Guan Y. The potential mechanisms underlying phthalate-induced hypospadias: a systematic review of rodent model studies. Front Endocrinol (Lausanne) 2024; 15:1490011. [PMID: 39698037 PMCID: PMC11652206 DOI: 10.3389/fendo.2024.1490011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Objectives Maternal exposure to environmental endocrine disruptors, such as phthalates, during pregnancy is a significant risk factor for the development of hypospadias. By consolidating existing research on the mechanisms by which phthalates induce hypospadias in rodent models, this systematic review aims to organize and analyze the discovered mechanisms and their potential connections. Methods The study involved all articles that explored the mechanisms of phthalate-induced hypospadias using rodent models. A comprehensive search of the PubMed and Web of Science databases was conducted using the terms "hypospadias" and "phthalates" before January 20, 2024. Then, two investigators screened for studies worthy of inclusion by setting inclusion and exclusion criteria. Results Of the initial 326 search results, 22 were included in the subsequent analysis. Based on the commonalities among different results, the mechanisms of phthalate-induced hypospadias could be categorized into the following five groups: sex steroids-related signaling pathways (n=10), epithelial-mesenchymal transition (n=6), autophagy (n=5), apoptosis (n=4) and angiogenesis (n=2). Among these, sex steroids-related signaling pathways might serve as a central regulator among all mechanisms, and reactive oxygen species (ROS) also played an important mediating role. Conclusion The systematic review indicates that phthalates may initially disrupt the balance of sex steroids-related pathways, leading to abnormally elevated levels of ROS and subsequently to other functional abnormalities, ultimately resulting in the development of hypospadias. All these findings will help to improve prevention strategies during pregnancy to reduce the adverse effects of phthalates on the offspring.
Collapse
Affiliation(s)
- Youtian Zhang
- Department of Urology, Tianjin Children’s Hospital/Tianjin University Children’s Hospital, Tianjin, China
| | - Jian Wang
- Department of Urology, Tianjin Children’s Hospital/Tianjin University Children’s Hospital, Tianjin, China
| | - Hongchao Yang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People’s Hospital), Dezhou, Shandong, China
| | - Yong Guan
- Department of Urology, Tianjin Children’s Hospital/Tianjin University Children’s Hospital, Tianjin, China
| |
Collapse
|
3
|
Kałużna M, Budny B, Rabijewski M, Dubiel A, Trofimiuk-Müldner M, Szutkowski K, Piotrowski A, Wrotkowska E, Hubalewska-Dydejczyk A, Ruchała M, Ziemnicka K. Variety of genetic defects in GnRH and hypothalamic-pituitary signaling and development in normosmic patients with IHH. Front Endocrinol (Lausanne) 2024; 15:1396805. [PMID: 39010903 PMCID: PMC11246878 DOI: 10.3389/fendo.2024.1396805] [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: 03/06/2024] [Accepted: 05/27/2024] [Indexed: 07/17/2024] Open
Abstract
Introduction Normosmic isolated hypogonadotropic hypogonadism (nIHH) is a clinically and genetically heterogeneous disorder. Deleterious variants in over 50 genes have been implicated in the etiology of IHH, which also indicates a possible role of digenicity and oligogenicity. Both classes of genes controlling GnRH neuron migration/development and hypothalamic/pituitary signaling and development are strongly implicated in nIHH pathogenesis. The study aimed to investigate the genetic background of nIHH and further expand the genotype-phenotype correlation. Methods A total of 67 patients with nIHH were enrolled in the study. NGS technology and a 38-gene panel were applied. Results Causative defects regarded as at least one pathogenic/likely pathogenic (P/LP) variant were found in 23 patients (34%). For another 30 individuals, variants of unknown significance (VUS) or benign (B) were evidenced (45%). The most frequently mutated genes presenting P/LP alterations were GNRHR (n = 5), TACR3 (n = 3), and CHD7, FGFR1, NSMF, BMP4, and NROB1 (n = 2 each). Monogenic variants with solid clinical significance (P/LP) were observed in 15% of subjects, whereas oligogenic defects were detected in 19% of patients. Regarding recurrence, 17 novel pathogenic variants affecting 10 genes were identified for 17 patients. The most recurrent pathogenic change was GNRHR:p.Arg139His, detected in four unrelated subjects. Another interesting observation is that P/LP defects were found more often in genes related to hypothalamic-pituitary pathways than those related to GnRH. Conclusions The growing importance of the neuroendocrine pathway and related genes is drawing increasing attention to nIHH. However, the underestimated potential of VUS variants in IHH etiology, particularly those presenting recurrence, should be further elucidated.
Collapse
Affiliation(s)
- Małgorzata Kałużna
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Bartłomiej Budny
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Michał Rabijewski
- Department of Reproductive Health, Centre for Postgraduate Medical Education, Warsaw, Poland
| | - Agnieszka Dubiel
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Kraków, Poland
| | | | - Kosma Szutkowski
- NanoBioMedical Centre at Adam Mickiewicz University in Poznan, Poznan, Poland
| | - Adam Piotrowski
- Department of Biomedical Physics at Adam Mickiewicz University in Poznan, Poznan, Poland
| | - Elżbieta Wrotkowska
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Marek Ruchała
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Ziemnicka
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
4
|
Hashimoto D, Fujimoto K, Nakata M, Suzuki T, Kumegawa S, Ueda Y, Suzuki K, Asamura S, Yamada G. Developmental and functional roles of androgen and interactive signals for external genitalia and erectile tissues. Reprod Med Biol 2024; 23:e12611. [PMID: 39372370 PMCID: PMC11456227 DOI: 10.1002/rmb2.12611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024] Open
Abstract
Background Recent progress in molecular and signal analyses revealed essential functions of cellular signals including androgen and related growth factors such as Wnt regulators for external genitalia (ExG) development and its pathogenesis. Accumulated data showed their fundamental functions also for erectile tissue (corporal body) development and its abnormalities. The current review focuses on such signals from developmental and functional viewpoints. Methods Experimental strategies including histological and molecular signal analyses with conditional mutant mice for androgen and Wnt signals have been extensively utilized. Main findings Essential roles of androgen for the development of male-type ExG and urethral formation are shown. Wnt signals are associated with androgen for male-type ExG organogenesis. Androgen plays essential roles in the development of erectile tissue, the corporal body and it also regulates the duration time of erection. Wnt and other signals are essential for the regulation of mesenchymal cells of erectile tissue as shown by its conditional mutant mouse analyses. Stress signals, continuous erection, and the potential of lymphatic characteristics of the erectile vessels with sinusoids are also shown. Conclusion Reiterated involvement of androgen, Wnt, and other regulatory factors is stated for the development and pathogenesis of ExG and erectile tissues.
Collapse
Affiliation(s)
- Daiki Hashimoto
- Department of Physiology, Faculty of MedicineWakayama Medical UniversityWakayamaJapan
| | - Kota Fujimoto
- Department of UrologyUrological Science Institute, Yonsei University College of MedicineSeoulSouth Korea
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Masanori Nakata
- Department of Physiology, Faculty of MedicineWakayama Medical UniversityWakayamaJapan
| | - Takuya Suzuki
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Shinji Kumegawa
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Yuko Ueda
- Department of UrologyOsaka Women's and Children's HospitalOsakaJapan
| | - Kentaro Suzuki
- Faculty of Life and Environmental SciencesUniversity of YamanashiYamanashiJapan
| | - Shinichi Asamura
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Gen Yamada
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| |
Collapse
|
5
|
Faria JAD, Moraes DR, Kulikowski LD, Batista RL, Gomes NL, Nishi MY, Zanardo E, Nonaka CKV, de Freitas Souza BS, Mendonca BB, Domenice S. Cytogenomic Investigation of Syndromic Brazilian Patients with Differences of Sexual Development. Diagnostics (Basel) 2023; 13:2235. [PMID: 37443631 DOI: 10.3390/diagnostics13132235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Cytogenomic methods have gained space in the clinical investigation of patients with disorders/differences in sexual development (DSD). Here we evaluated the role of the SNP array in achieving a molecular diagnosis in Brazilian patients with syndromic DSD of unknown etiology. METHODS Twenty-two patients with DSD and syndromic features were included in the study and underwent SNP-array analysis. RESULTS In two patients, the diagnosis of 46,XX SRY + DSD was established. Additionally, two deletions were revealed (3q29 and Xp22.33), justifying the syndromic phenotype in these patients. Two pathogenic CNVs, a 10q25.3-q26.2 and a 13q33.1 deletion encompassing the FGFR2 and the EFNB2 gene, were associated with genital atypia and syndromic characteristics in two patients with 46,XY DSD. In a third 46,XY DSD patient, we identified a duplication in the 14q11.2-q12 region of 6.5 Mb associated with a deletion in the 21p11.2-q21.3 region of 12.7 Mb. In a 46,XY DSD patient with delayed neuropsychomotor development and congenital cataracts, a 12 Kb deletion on chromosome 10 was found, partially clarifying the syndromic phenotype, but not the genital atypia. CONCLUSIONS The SNP array is a useful tool for DSD patients, identifying the molecular etiology in 40% (2/5) of patients with 46,XX DSD and 17.6% (3/17) of patients with 46,XY DSD.
Collapse
Affiliation(s)
- José Antonio Diniz Faria
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador 40110-909, Brazil
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-010, Brazil
| | - Daniela R Moraes
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-010, Brazil
| | - Leslie Domenici Kulikowski
- Laboratório de Citogenômica e Patologia Molecular LIM/03, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-010, Brazil
| | - Rafael Loch Batista
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-010, Brazil
| | - Nathalia Lisboa Gomes
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-010, Brazil
| | - Mirian Yumie Nishi
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-010, Brazil
| | - Evelin Zanardo
- Laboratório de Citogenômica e Patologia Molecular LIM/03, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-010, Brazil
| | - Carolina Kymie Vasques Nonaka
- Centro de Biotecnologia e Terapia Celular, Hospital São Rafael, Salvador 41253-190, Brazil
- Instituto D'Or de Pesquisa e Ensino (IDOR), Salvador 41253-190, Brazil
| | - Bruno Solano de Freitas Souza
- Centro de Biotecnologia e Terapia Celular, Hospital São Rafael, Salvador 41253-190, Brazil
- Instituto D'Or de Pesquisa e Ensino (IDOR), Salvador 41253-190, Brazil
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador 40296-710, Brazil
| | - Berenice Bilharinho Mendonca
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-010, Brazil
| | - Sorahia Domenice
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-010, Brazil
| |
Collapse
|
6
|
Chen L, Wang J, Lu W, Xiao Y, Ni J, Wang W, Ma X, Dong Z. Characterization With Gene Mutations in Han Chinese Patients With Hypospadias and Function Analysis of a Novel AR Genevariant. Front Genet 2021; 12:673732. [PMID: 34276780 PMCID: PMC8278054 DOI: 10.3389/fgene.2021.673732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/09/2021] [Indexed: 11/13/2022] Open
Abstract
It is estimated that around 10-20% of hypospadias are caused by genetic abnormalities worldwide although the spectrum of associated genes does vary across different ethnicities. The prevalence of hypospadias among the Chinese population has been increasing the last couple of decades. However, the pathogenesis underlying the disease and its associated genetic abnormality remains unclear. Here we performed a genetic analysis of 81 children with karyotype 46, XY and the hypospadias phenotype in order to characterize the genetic components that contribute to the development of hypospadias in Chinese patients. 15 candidate genes, including sex determination genes-SOX9, SRY, NR0B1 (DAX1), NR5A1 (SF1), DHH, sex differentiation genes-AR, SRD5A2, MAMLD1, INSL3, and hypospadias-associated genes-FGF8, FGF10, BMP4, BMP7, ATF3, and MID1 were screened by using next generation sequencing. A total of 18 patients were found to have mutations identified by PCR and sequencing, including 11 cases of SRD5A2 genes, 6 cases of AR genes, and 1 case of MID1 gene, respectively. One novel missense mutation p.I817N was discovered in AR gene. Further molecular analysis found that subcellular localization of the ARI 81 7N was the same as that of wild type ARWT in the absence or presence of hormone. But it led to 50% reduction in AR-induced transcriptional activity in the presence of either the synthetic androgen R1881 or the natural ligand dihydrotestosterone. Our results indicate that SRD5A2 and AR genes are two top candidate genes associated with 46, XY hypospadias in Chinese patients. Further epidemiological and genetic analysis are still needed to further clarify the pathogenesis of hypospadias in Han Chinese patients.
Collapse
Affiliation(s)
- Lifen Chen
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junqi Wang
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenli Lu
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Xiao
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jihong Ni
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Wang
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyu Ma
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiya Dong
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
7
|
Endocrine disrupting chemicals in the pathogenesis of hypospadias; developmental and toxicological perspectives. Curr Res Toxicol 2021; 2:179-191. [PMID: 34345859 PMCID: PMC8320613 DOI: 10.1016/j.crtox.2021.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/11/2022] Open
Abstract
Penis development is regulated by a tight balance of androgens and estrogens. EDCs that impact androgen/estrogen balance during development cause hypospadias. Cross-disciplinary collaborations are needed to define a mechanistic link.
Hypospadias is a defect in penile urethral closure that occurs in approximately 1/150 live male births in developed nations, making it one of the most common congenital abnormalities worldwide. Alarmingly, the frequency of hypospadias has increased rapidly over recent decades and is continuing to rise. Recent research reviewed herein suggests that the rise in hypospadias rates can be directly linked to our increasing exposure to endocrine disrupting chemicals (EDCs), especially those that affect estrogen and androgen signalling. Understanding the mechanistic links between endocrine disruptors and hypospadias requires toxicologists and developmental biologists to define exposures and biological impacts on penis development. In this review we examine recent insights from toxicological, developmental and epidemiological studies on the hormonal control of normal penis development and describe the rationale and evidence for EDC exposures that impact these pathways to cause hypospadias. Continued collaboration across these fields is imperative to understand the full impact of endocrine disrupting chemicals on the increasing rates of hypospadias.
Collapse
Key Words
- Androgen
- BBP, benzyl butyl phthalate
- BPA, bisphenol A
- DBP, Σdibutyl phthalate
- DDT, dichlorodiphenyltrichloroethane
- DEHP, Σdi-2(ethylhexyl)-phthalate
- DHT, dihydrotestosterone
- EDC, endocrine disrupting chemicals
- EMT, epithelial to mesenchymal transition
- ER, estrogen receptor
- Endocrine disruptors
- Estrogen
- GT, genital tubercle
- Hypospadias
- NOAEL, no observed adverse effect level
- PBB, polybrominated biphenyl
- PBDE, polybrominated diphenyl ether
- PCB, polychlorinated biphenyl
- PCE, tetrachloroethylene
- Penis
Collapse
|
8
|
Kong X, Luo J, Xiang H, Wang S, Shen L, Long C, Liu F, Lin T, He D, Liu X, Wei GH. Expression of Mafb is down-regulated in the foreskin of children with hypospadias. J Pediatr Urol 2021; 17:70.e1-70.e6. [PMID: 33268316 DOI: 10.1016/j.jpurol.2020.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 09/17/2020] [Accepted: 10/07/2020] [Indexed: 01/23/2023]
Abstract
BACKGROUND Hypospadias is the second most common congenital malformation in males. Although the aetiology of hypospadias is not clear, it is generally thought to be affected by both genetic and environmental endocrine-disrupting factors that affect the development of the urethra, leading to deformity. OBJECTIVE To investigate the difference in expression of the transcription factor Mafb in hypospadias and normal penile tissues and to assess whether it is related to the occurrence of hypospadias. STUDY DESIGN Penile tissue was obtained from children with hypospadias who underwent surgical repair at the Children's Hospital of Chongqing Medical University. Patients diagnosed with undescended testicles, intersex status or endocrine abnormalities were excluded from the study. Twenty-five cases with hypospadias (average 3.5 years old) and 15 cases with circumcisions (as control) (average 5 years old) were included in this study. Real-time quantitative polymerase chain reaction, Immunochemistry and Western blot were used to detect the expression of Mafb. RESULTS Mafb mRNA expressions in the prepuce of cases with hypospadias was significantly reduced compared with that in the controls [(1.179 ± 0.1275), (0.6652 ± 0.07506), p < 0.05)]. Hypospadias cases also showed decreased Mafb protein expression in the preputial subcutaneous mesenchymal cell layer. Mafb protein levels were significantly decreased in those with hypospadias compared with controls [(1.932 ± 0.1139), (1.006 ± 0.03312), p < 0.05]. However, no such differences were found in Mafb expression between subjects with mild and severe hypospadias. DISCUSSION Compared to the normal foreskin, expression of the Mafb gene was down-regulated at both mRNA and protein levels, which was consistent with our RNA-seq sequencing results in Diethylhexyl phthalate (DEHP)-induced hypospadias rats. This study is the first to report abnormal expression of Mafb in the preputial tissue of hypospadias cases. An in-depth study of the relationship between Mafb and cell proliferation, apoptosis, and urethra development may reveal the pathogenesis of hypospadias. CONCLUSION Expression of the Mafb gene and protein in the foreskin of children with hypospadias is lower than that in normal foreskin. We postulate that such abnormal expression of the Mafb gene may be related to the occurrence of hypospadias and that this abnormal expression may affect the development of the urethra during the embryonic period.
Collapse
Affiliation(s)
- Xiaoyan Kong
- Department of Urology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, China
| | - Jin Luo
- Department of Urology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Han Xiang
- Department of Urology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Shao Wang
- Department of Urology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Lianju Shen
- Department of Urology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, China
| | - Chunlan Long
- Department of Urology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, China
| | - Feng Liu
- Department of Urology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, China
| | - Tao Lin
- Department of Urology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, China
| | - Dawei He
- Department of Urology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, China
| | - Xing Liu
- Department of Urology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, China.
| | - Guang-Hui Wei
- Department of Urology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, China
| |
Collapse
|
9
|
Chang J, Wang S, Zheng Z. Etiology of Hypospadias: A Comparative Review of Genetic Factors and Developmental Processes Between Human and Animal Models. Res Rep Urol 2021; 12:673-686. [PMID: 33381468 PMCID: PMC7769141 DOI: 10.2147/rru.s276141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 09/28/2020] [Indexed: 11/23/2022] Open
Abstract
Hypospadias is a congenital anomaly of the penis with an occurrence of approximately 1 in 200 boys, but the etiology of the majority of hypospadias has remained unknown. Numerous genes have been reported as having variants in hypospadias patients, and many studies on genetic deletion of key genes in mouse genital development have also been published. Until now, no comparative analysis in the genes related literature has been reported. The basic knowledge of penile development and hypospadias is mainly obtained from animal model studies. Understanding of the differences and similarities between human and animal models is crucial for studies of hypospadias. In this review, mutations and polymorphisms of hypospadias-related genes have been compared between humans and mice, and differential genotype–phenotype relationships of certain genes between humans and mice have been discussed using the data available in PubMed and MGI online databases, and our analysis only revealed mutations in seven out of 43 human hypospadias related genes which have been reported to show similar phenotypes in mutant mice. The differences and similarities in the processes of penile development and hypospadias malformation among human and commonly used animal models suggest that the guinea pig may be a good model to study the mechanism of human penile development and etiology of hypospadias.
Collapse
Affiliation(s)
- Jun Chang
- Department of Physiology, School of Medicine, Southern Illinois University Carbondale, Carbondale, IL 62901, USA.,School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, People's Republic of China
| | - Shanshan Wang
- Department of Physiology, School of Medicine, Southern Illinois University Carbondale, Carbondale, IL 62901, USA
| | - Zhengui Zheng
- Department of Physiology, School of Medicine, Southern Illinois University Carbondale, Carbondale, IL 62901, USA
| |
Collapse
|
10
|
Punjani N, Lamb DJ. Male infertility and genitourinary birth defects: there is more than meets the eye. Fertil Steril 2020; 114:209-218. [PMID: 32741459 PMCID: PMC10590568 DOI: 10.1016/j.fertnstert.2020.06.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/24/2020] [Accepted: 06/24/2020] [Indexed: 12/17/2022]
Abstract
Male factor infertility is a significant problem present in up to 50% of infertile couples. The relationship between male infertility and systemic disease is of significant interest, and emerging evidence suggests a relationship between male infertility and male genitourinary (GU) birth defects (cryptorchidism, hypospadias, ambiguous genitalia, and congenital anomalies of the kidney and urinary tract). Many of these birth defects are treated in isolation by busy urologists without acknowledgment that these may be related to more global syndromic conditions. Conversely, geneticists and nonurologists who treat variable systemic phenotypes may overlook GU defects, which are indeed related conditions. Many of these defects are attributed to copy number variants dosage-sensitive genes due to chromosome microdeletions or microduplications. These variants are responsible for disease phenotypes seen in the general population. The copy number variants described in this review are syndromic in some cases and responsible for both GU birth defects as well as other systemic phenotypes. This review highlights the emerging evidence between these birth defects, male infertility, and other systemic conditions.
Collapse
Affiliation(s)
- Nahid Punjani
- James Buchanan Brady Foundation Institute of Urology, Weill Cornell Medical College, New York, New York
| | - Dolores J Lamb
- James Buchanan Brady Foundation Institute of Urology, Weill Cornell Medical College, New York, New York; Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, New York; Center for Reproductive Genomics, Weill Cornell Medical College, New York, New York.
| |
Collapse
|
11
|
Piñeyro-Ruiz C, Serrano H, Pérez-Brayfield MR, Jorge JC. New frontiers on the molecular underpinnings of hypospadias according to severity. Arab J Urol 2020; 18:257-266. [PMID: 33312738 PMCID: PMC7717703 DOI: 10.1080/2090598x.2020.1760589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/13/2020] [Indexed: 10/31/2022] Open
Abstract
Hypospadias, which is characterised by the displacement of the urethral meatus from its typical anatomical location in males, shows various degrees of severity. In this systematic review, we surveyed our current understanding of the genetics of isolated hypospadias in humans according to the severity of the condition. We found that sequencing and genotyping approaches were the preferred methods of study and that single nucleotide polymorphisms were the most common finding associated with hypospadias. Most genes fell into four gene-pathway categories related to androgens, oestrogens, growth factors, or transcription factors. Few hypospadias studies classify their findings by severity. Taken together, we argue that it is advantageous to take into consideration the severity of the condition in search of novel candidates in the aetiology of hypospadias. Abbreviations: AR: androgen receptor; ATF3: activating transcription factor 3; BMP4: bone morphogenetic protein 4; BMP7: bone morphogenetic protein 7; CYP17: steroid 17-alpha-hydroxylase/17,20 lyase; CYP1A1: cytochrome P450 1A1; CYP3A4: cytochrome P450 3A4; CNVs: copy number variants; DGKK: diacylglycerol kinase kappa; ESR1: oestrogen receptor 1; ESR2: oestrogen receptor 2; FGF8: fibroblast growth factor 8; FGF10: fibroblast growth factor 10; FGFR2: fibroblast growth factor receptor 2; HOXA4: homeobox protein Hox-A4; HOXB6: homeobox protein Hox-B6; HSD17B3: hydroxysteroid 17-beta dehydrogenase 3; MAMLD1: mastermind-like domain-containing protein 1; SF-1: splicing factor 1; SHH: sonic hedgehog; SNPs: single nucleotide polymorphisms; SOX9: SRY-box 9; SRD5A2: steroid 5 alpha-reductase 2; SRY: sex-determining region Y protein; STAR: steroidogenic acute regulatory protein; STARD3: StAR-related lipid transfer protein 3; STS: steryl-sulfatase; WT1: Wilms tumour protein; ZEB1: zinc finger oestrogen-box binding homeobox 1.
Collapse
Affiliation(s)
- Coriness Piñeyro-Ruiz
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico, USA
| | - Horacio Serrano
- Department of Internal Medicine and Department of Biochemistry, University of Puerto Rico, San Juan, Puerto Rico, USA
| | - Marcos R. Pérez-Brayfield
- Department of Surgery, Section of Urology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico, USA
| | - Juan Carlos Jorge
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico, USA
| |
Collapse
|
12
|
Flück CE, Audí L, Fernández-Cancio M, Sauter KS, Martinez de LaPiscina I, Castaño L, Esteva I, Camats N. Broad Phenotypes of Disorders/Differences of Sex Development in MAMLD1 Patients Through Oligogenic Disease. Front Genet 2019; 10:746. [PMID: 31555317 PMCID: PMC6726737 DOI: 10.3389/fgene.2019.00746] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Disorders/differences of sex development (DSD) are the result of a discordance between chromosomal, gonadal, and genital sex. DSD may be due to mutations in any of the genes involved in sex determination and development in general, as well as gonadal and/or genital development specifically. MAMLD1 is one of the recognized DSD genes. However, its role is controversial as some MAMLD1 variants are present in normal individuals, several MAMLD1 mutations have wild-type activity in functional studies, and the Mamld1-knockout male mouse presents with normal genitalia and reproduction. We previously tested nine MAMLD1 variants detected in nine 46,XY DSD patients with broad phenotypes for their functional activity, but none of the mutants, except truncated L210X, had diminished transcriptional activity on known target promoters CYP17A1 and HES3. In addition, protein expression of MAMLD1 variants was similar to wild-type, except for the truncated L210X. We hypothesized that MAMLD1 variants may not be sufficient to explain the phenotype in 46,XY DSD individuals, and that further genetic studies should be performed to search for additional hits explaining the broad phenotypes. We therefore performed whole exome sequencing (WES) in seven of these 46,XY patients with DSD and in one 46,XX patient with ovarian insufficiency, who all carried MAMLD1 variants. WES data were filtered by an algorithm including disease-tailored lists of MAMLD1-related and DSD-related genes. Fifty-five potentially deleterious variants in 41 genes were identified; 16/55 variants were reported in genes in association with hypospadias, 8/55 with cryptorchidism, 5/55 with micropenis, and 13/55 were described in relation with female sex development. Patients carried 1-16 variants in 1-16 genes together with their MAMLD1 variation. Network analysis of the identified genes revealed that 23 genes presented gene/protein interactions with MAMLD1. Thus, our study shows that the broad phenotypes of individual DSD might involve multiple genetic variations contributing towards the complex network of sexual development.
Collapse
Affiliation(s)
- Christa E Flück
- Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics and Department of BioMedical Research, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Laura Audí
- Growth and Development Research Unit, Vall d'Hebron Research Institute (VHIR), Center for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Mónica Fernández-Cancio
- Growth and Development Research Unit, Vall d'Hebron Research Institute (VHIR), Center for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Kay-Sara Sauter
- Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics and Department of BioMedical Research, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Idoia Martinez de LaPiscina
- Endocrinology and Diabetes Research Group, BioCruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERDEM, CIBERER, University of the Basque Country (UPV-EHU), Barakaldo, Spain
| | - Luis Castaño
- Pediatric Endocrinology Section, Cruces University Hospital, Endocrinology and Diabetes Research Group, BioCruces Bizkaia Health Research Institute, CIBERDEM, CIBERER, University of the Basque Country (UPV-EHU), Barakaldo, Spain
| | - Isabel Esteva
- Endocrinology Section, Gender Identity Unit, Regional University Hospital of Malaga, Málaga, Spain
| | - Núria Camats
- Growth and Development Research Unit, Vall d'Hebron Research Institute (VHIR), Center for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| |
Collapse
|
13
|
Abstract
Aims: The aim of this study was to try to explain the pathogenesis of proximal hypospadias based on anatomical and histological findings. Methods: During 9 years, we performed systematic biopsies (in the lateral areas of the urethral plate, as well as under this plate) in 81 patients treated for proximal hypospadias. The histological study was performed by routine coloring, hematoxylin and eosin, and Masson's trichrome, which colors the collagen fibers in blue, and monoclonal antibody against alpha-smooth muscle actin. Results: There is a fibrosis tissue abnormally present on the ventral side of the penis. This tissue consists of a mixture of fibrous connective tissue, nerve nets, short vessels, and smooth muscle fibers. The penis' dartos does not contain smooth muscle fibers. These fibers can come from a blood vessel or spongy tissue which existed during the neonatal period in the distal part of the penis before disappearing. Conclusions: The proximal hypospadias is due presumably to avascular necrosis of the distal part poorly vascularized of the corpus spongiosum.
Collapse
Affiliation(s)
- Smail Acimi
- Department of Visceral Surgery, Faculty of Medicine, Children's Hospital Canastel, University of Oran, Oran, Algeria
| |
Collapse
|
14
|
Chen Y, Kuroki Y, Shaw G, Pask AJ, Yu H, Toyoda A, Fujiyama A, Renfree MB. Androgen and Oestrogen Affect the Expression of Long Non-Coding RNAs During Phallus Development in a Marsupial. Noncoding RNA 2018; 5:E3. [PMID: 30598023 PMCID: PMC6468475 DOI: 10.3390/ncrna5010003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/19/2018] [Accepted: 12/27/2018] [Indexed: 12/24/2022] Open
Abstract
There is increasing evidence that long non-coding RNAs (lncRNAs) are important for normal reproductive development, yet very few lncRNAs have been identified in phalluses so far. Unlike eutherians, phallus development in the marsupial tammar wallaby occurs post-natally, enabling manipulation not possible in eutherians in which differentiation occurs in utero. We treated with sex steroids to determine the effects of androgen and oestrogen on lncRNA expression during phallus development. Hormonal manipulations altered the coding and non-coding gene expression profile of phalluses. We identified several predicted co-regulatory lncRNAs that appear to be co-expressed with the hormone-responsive candidate genes regulating urethral closure and phallus growth, namely IGF1, AR and ESR1. Interestingly, more than 50% of AR-associated coding genes and lncRNAs were also associated with ESR1. In addition, we identified and validated three novel co-regulatory and hormone-responsive lncRNAs: lnc-BMP5, lnc-ZBTB16 and lncRSPO4. Lnc-BMP5 was detected in the urethral epithelium of male phalluses and was downregulated by oestrogen in males. Lnc-ZBTB16 was downregulated by oestrogen treatment in male phalluses at day 50 post-partum (pp). LncRSPO4 was downregulated by adiol treatment in female phalluses but increased in male phalluses after castration. Thus, the expression pattern and hormone responsiveness of these lncRNAs suggests a physiological role in the development of the phallus.
Collapse
Affiliation(s)
- Yu Chen
- School of BioSciences, The University of Melbourne 3010, VIC, Australia.
| | - Yoko Kuroki
- RIKEN, Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan.
| | - Geoff Shaw
- School of BioSciences, The University of Melbourne 3010, VIC, Australia.
| | - Andrew J Pask
- School of BioSciences, The University of Melbourne 3010, VIC, Australia.
| | - Hongshi Yu
- School of BioSciences, The University of Melbourne 3010, VIC, Australia.
| | - Atsushi Toyoda
- Advanced Genomics Center, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan.
| | - Asao Fujiyama
- Advanced Genomics Center, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan.
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne 3010, VIC, Australia.
| |
Collapse
|
15
|
Joodi M, Amerizadeh F, Hassanian SM, Erfani M, Ghayour-Mobarhan M, Ferns GA, Khazaei M, Avan A. The genetic factors contributing to hypospadias and their clinical utility in its diagnosis. J Cell Physiol 2018; 234:5519-5523. [PMID: 30238986 DOI: 10.1002/jcp.27350] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 08/17/2018] [Indexed: 12/24/2022]
Abstract
Hypospadias is among the most common congenital malformations in male neonates. It results from abnormal penile and urethral development, but is a multifactorial disorder that is highly heterogeneous, with several genetic and environmental determinants. Monogenic and chromosomal abnormalities are present in approximately 30% of cases, although the genetic factors contributing to hypospadias remain unknown in 70% of cases. While defects in androgen synthesis can lead to this malformation, mutational analyses have shown several genes, such as sonic hedgehog, fibroblast growth factors, bone morphogenetic proteins, homeobox genes, and the Wnt family, are involved in the normal development of male external genitalia. Mutations in the genes of penile development (e.g., HOX, FGF, Shh) and testicular determination (e.g., WT1, SRY), luteinizing hormone receptor, and androgen receptor have also been proposed to be implicated in hypospadias. Here we review the recent advances in this field and discuss the potential genes that could determine the risk of hypospadias.
Collapse
Affiliation(s)
- Marjan Joodi
- Department of Pediatric Surgery, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Sarvar Children's Hospital, Endoscopic and Minimally Invasive Surgery Research Center, Mashhad, Iran
| | - Forouzan Amerizadeh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marjan Erfani
- Department of Neurology, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School Falmer, Brighton, UK
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
16
|
Acimi S. What is the pathogenesis of proximal hypospadias? Turk J Urol 2018; 44:357-361. [PMID: 29799398 DOI: 10.5152/tud.2018.85530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/23/2017] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To report the information concerning the pathogenesis of proximal hypospadias and causes of curvature associated with it. MATERIAL AND METHODS From January 2009 to December 2015, 74 patients underwent repair of proximal hypospadias. In 70 patients we performed a systematic biopsies in the lateral areas of the urethral plate, as well as under this plate. The study of the histological structure of these areas was performed using routine staining with hematoxylin and eosin, and the Masson's trichrome which color the collagen fibers in blue and monoclonal antibody against alpha-smooth muscle actin. RESULTS This prospective study shows that the fibrotic tissue abnormally present on the ventral side of the penis consists of a mixture of fibrous connective tissue, nerve nets, short vessels, and smooth muscle fibers. In contrast to the scrotal dartos, penile dartos fascia does not contain smooth muscle fibers. Therefore, these fibers may come from a blood vessel or spongy tissue which existed during neonatal period in the distal part of the penis before disappearing. In addition, in 13 cases, the presence downstream of the urethral meatus, of a bifurcation of corpus spongiosum into two branches supposes that the corpus spongiosum is form by fusion around the urethra of two mesenchymal bodies. The arterial supply of this purely masculine formation originates from a new vascularization and it is probably, developed under secretion of androgens (angiogenic substances in target tissues). CONCLUSION These findings allow us to suggest that the proximal hypospadias is due to avascular necrosis of the distal part ie. poorly vascularized part of the corpus spongiosum.
Collapse
Affiliation(s)
- Smail Acimi
- Department of Visceral Surgery, Children hospital Canastel, Faculty of Medicine, University of Oran, Oran, Algeria
| |
Collapse
|
17
|
Regulation of masculinization: androgen signalling for external genitalia development. Nat Rev Urol 2018; 15:358-368. [DOI: 10.1038/s41585-018-0008-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
18
|
Wang Y, Gong C, Qin M, Liu Y, Tian Y. Clinical and genetic features of 64 young male paediatric patients with congenital hypogonadotropic hypogonadism. Clin Endocrinol (Oxf) 2017; 87:757-766. [PMID: 28833369 DOI: 10.1111/cen.13451] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/27/2017] [Accepted: 08/11/2017] [Indexed: 11/26/2022]
Abstract
CONTEXT The diagnosis of congenital hypogonadotropic hypogonadism (CHH) in prepuberty has always been challenging. Here, we aimed at studying the clinical and genetic features of paediatric CHH, especially the phenotype of hypospadias and dual defects (patients showing hypothalamic and/or pituitary defects and testicular hypoplasia), so as to have a better understanding of CHH. DESIGN The clinical and genetic features of patients with CHH were analysed, and the relationships between hypospadias, dual defects and genetics were investigated. PATIENTS Patients who visited Beijing Children's Hospital and were positively diagnosed with CHH. MEASUREMENTS The collected data included sex hormones, MRI of the olfactory bulb, human chorionic gonadotrophin (hCG) test and genetic testing. We analysed clinical features and genetic results, especially hypospadias and dual defects, and compared the stimulated testosterone (T) levels in patients with and without cryptorchidism. RESULTS Sixty-four patients were positively diagnosed, and forty-seven (73.4%) had Kallmann syndrome (KS). Four patients (6.3%) had hypospadias, including 2 KS. Micropenis combined with cryptorchidism was the most common phenotype (39%). Approximately two-third of patients showed a poor response to hCG; 15 cases were diagnosed with dual defects, and there were no significant differences between those with and without cryptorchidism. Twenty-six cases (51%) of 51 patients were identified as having classical HH mutations, affecting 10 different genes, with oligogenic mutations in 5 cases (9.8%). The most common mutations were in PROKR2 (17.6%), FGFR1 (13.7%) and CHD7 (7.8%). The frequency of PROKR2 mutations was higher in dual HH when compared to other HH cases (6/15 vs 3/36, P = .021). CONCLUSIONS Micropenis and/or cryptorchidism can serve as important signs for the diagnosis of HH in paediatrics, and the coexistence of hypospadias does not exclude the diagnosis of CHH, including KS or normosmic isolated HH (nHH). Testicular function may be impaired earlier than expected, and PROKR2 mutations need to be evaluated to identify presumed dual defects.
Collapse
Affiliation(s)
- Yi Wang
- National Center for Children's Health, Capital Medical University, Beijing, China
- Department of Endocrinology, Genetics, Metabolism and Adolescent Medicine, Beijing Children's Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Chunxiu Gong
- National Center for Children's Health, Capital Medical University, Beijing, China
- Department of Endocrinology, Genetics, Metabolism and Adolescent Medicine, Beijing Children's Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Miao Qin
- National Center for Children's Health, Capital Medical University, Beijing, China
- Department of Endocrinology, Genetics, Metabolism and Adolescent Medicine, Beijing Children's Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Ying Liu
- National Center for Children's Health, Capital Medical University, Beijing, China
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yuanyuan Tian
- National Center for Children's Health, Capital Medical University, Beijing, China
- Department of Endocrinology, Genetics, Metabolism and Adolescent Medicine, Beijing Children's Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Children's Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Molecular diagnosis in hypospadias. Rev Urol 2017. [DOI: 10.1016/j.uroco.2017.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Abello A, Ayala P, Ortiz AM, Fernández N. Diagnóstico molecular en hipospadias. Rev Urol 2017. [DOI: 10.1016/j.uroco.2016.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
21
|
Hagan A, Amarillo IE. Small copy-number variations involving genes of the FGF pathway in differences in sex development. Hum Genome Var 2017; 4:17011. [PMID: 28446957 PMCID: PMC5390097 DOI: 10.1038/hgv.2017.11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/11/2016] [Accepted: 01/14/2017] [Indexed: 11/17/2022] Open
Abstract
Retrospective chromosome microarray analysis of 83 genes within the fibroblast growth factor signaling pathway in 52 patients with heterogeneous differences in sex development (DSD) revealed small copy-number variations (CNVs) in ~31% (n=26) of investigated genes. Roughly half of these genes (39/83) are ⩽50 kb. This study highlights the potential involvement of small CNVs in disrupting normal gene function and dysregulating genes of the FGF pathway associated with DSD.
Collapse
Affiliation(s)
- Andrew Hagan
- Department of Developmental Biology, Washington University in St Louis School of Medicine, Saint Louis, MO, USA
| | - Ina E Amarillo
- Cytogenetics and Molecular Pathology Laboratory, Division of Laboratory and Genomic Medicine, Department of Pathology and Immunology, Washington University in St Louis School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
22
|
de Graaf P, van der Linde EM, Rosier PFWM, Izeta A, Sievert KD, Bosch JLHR, de Kort LMO. Systematic Review to Compare Urothelium Differentiation with Urethral Epithelium Differentiation in Fetal Development, as a Basis for Tissue Engineering of the Male Urethra. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:257-267. [PMID: 27809709 DOI: 10.1089/ten.teb.2016.0352] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Tissue-engineered (TE) urethra is desirable in men with urethral disease (stricture or hypospadias) and shortage of local tissue. Although ideally a TE graft would contain urethral epithelium cells, currently, bladder epithelium (urothelium) is widely used, but morphologically different. Understanding the differences and similarities of urothelium and urethral epithelium could help design a protocol for in vitro generation of urethral epithelium to be used in TE grafts for the urethra. PURPOSE To understand the development toward urethral epithelium or urothelium to improve TE of the urethra. METHODS A literature search was done following PRISMA guidelines. Articles describing urethral epithelium and bladder urothelium development in laboratory animals and humans were selected. RESULTS Twenty-nine studies on development of urethral epithelium and 29 studies on development of urothelium were included. Both tissue linings derive from endoderm and although adult urothelium and urethral epithelium are characterized by different gene expression profiles, the signaling pathways underlying their development are similar, including Shh, BMP, Wnt, and FGF. The progenitor of the urothelium and the urethral epithelium is the early fetal urogenital sinus (UGS). The urethral plate and the urothelium are both formed from the p63+ cells of the UGS. Keratin 20 and uroplakins are exclusively expressed in urothelium, not in the urethral epithelium. Further research has to be done on unique markers for the urethral epithelium. CONCLUSION This review has summarized the current knowledge about embryonic development of urothelium versus urethral epithelium and especially focuses on the influencing factors that are potentially specific for the eventual morphological differences of both cell linings, to be a basis for developmental or tissue engineering of urethral tissue.
Collapse
Affiliation(s)
- Petra de Graaf
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands .,2 Regenerative Medicine Center Utrecht , Utrecht, The Netherlands
| | | | - Peter F W M Rosier
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands
| | - Ander Izeta
- 3 Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, Hospital Universitario Donostia , San Sebastián, Spain .,4 Department of Biomedical Engineering, School of Engineering, Tecnun-University of Navarra , San Sebastián, Spain
| | | | - J L H Ruud Bosch
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands
| | - Laetitia M O de Kort
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands
| |
Collapse
|
23
|
Computational modeling and simulation of genital tubercle development. Reprod Toxicol 2016; 64:151-61. [PMID: 27180093 DOI: 10.1016/j.reprotox.2016.05.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/13/2016] [Accepted: 05/07/2016] [Indexed: 11/22/2022]
Abstract
Hypospadias is a developmental defect of urethral tube closure that has a complex etiology involving genetic and environmental factors, including anti-androgenic and estrogenic disrupting chemicals; however, little is known about the morphoregulatory consequences of androgen/estrogen balance during genital tubercle (GT) development. Computer models that predictively model sexual dimorphism of the GT may provide a useful resource to translate chemical-target bipartite networks and their developmental consequences across the human-relevant chemical universe. Here, we describe a multicellular agent-based model of genital tubercle (GT) development that simulates urethrogenesis from the sexually-indifferent urethral plate stage to urethral tube closure. The prototype model, constructed in CompuCell3D, recapitulates key aspects of GT morphogenesis controlled by SHH, FGF10, and androgen pathways through modulation of stochastic cell behaviors, including differential adhesion, motility, proliferation, and apoptosis. Proper urethral tube closure in the model was shown to depend quantitatively on SHH- and FGF10-induced effects on mesenchymal proliferation and epithelial apoptosis-both ultimately linked to androgen signaling. In the absence of androgen, GT development was feminized and with partial androgen deficiency, the model resolved with incomplete urethral tube closure, thereby providing an in silico platform for probabilistic prediction of hypospadias risk across combinations of minor perturbations to the GT system at various stages of embryonic development.
Collapse
|
24
|
Fernández N, Pérez J, Zarante I. Is hypospadias a spectrum of different diseases? MAMLD1 gen: A new candidate gene for hypospadias. Rev Urol 2015. [DOI: 10.1016/j.uroco.2015.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
25
|
Fernández N, Pérez J, Zarante I. ¿Son las hipospadias la expresión de diferentes enfermedades? MAMLD1 : un nuevo gen candidato para hipospadias. UROLOGÍA COLOMBIANA 2015. [DOI: 10.1016/j.uroco.2015.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
26
|
Bouty A, Ayers KL, Pask A, Heloury Y, Sinclair AH. The Genetic and Environmental Factors Underlying Hypospadias. Sex Dev 2015; 9:239-259. [PMID: 26613581 DOI: 10.1159/000441988] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2015] [Indexed: 12/22/2022] Open
Abstract
Hypospadias results from a failure of urethral closure in the male phallus and affects 1 in 200-300 boys. It is thought to be due to a combination of genetic and environmental factors. The development of the penis progresses in 2 stages: an initial hormone-independent phase and a secondary hormone-dependent phase. Here, we review the molecular pathways that contribute to each of these stages, drawing on studies from both human and mouse models. Hypospadias can occur when normal development of the phallus is disrupted, and we provide evidence that mutations in genes underlying this developmental process are causative. Finally, we discuss the environmental factors that may contribute to hypospadias and their potential immediate and transgenerational epigenetic impacts.
Collapse
Affiliation(s)
- Aurore Bouty
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Melbourne, Vic., Australia.,Department of Surgery, Royal Children's Hospital, University of Melbourne, Melbourne, Vic., Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Vic., Australia
| | - Katie L Ayers
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Melbourne, Vic., Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Vic., Australia
| | - Andrew Pask
- Department of Zoology, University of Melbourne, Melbourne, Vic., Australia
| | - Yves Heloury
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Melbourne, Vic., Australia.,Department of Surgery, Royal Children's Hospital, University of Melbourne, Melbourne, Vic., Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Vic., Australia
| | - Andrew H Sinclair
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Melbourne, Vic., Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Vic., Australia
| |
Collapse
|
27
|
Gredler ML, Seifert AW, Cohn MJ. Tissue-specific roles of Fgfr2 in development of the external genitalia. Development 2015; 142:2203-12. [PMID: 26081573 DOI: 10.1242/dev.119891] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Congenital anomalies frequently occur in organs that undergo tubulogenesis. Hypospadias is a urethral tube defect defined by mislocalized, oversized, or multiple openings of the penile urethra. Deletion of Fgfr2 or its ligand Fgf10 results in severe hypospadias in mice, in which the entire urethral plate is open along the ventral side of the penis. In the genital tubercle, the embryonic precursor of the penis and clitoris, Fgfr2 is expressed in two epithelial populations: the endodermally derived urethral epithelium and the ectodermally derived surface epithelium. Here, we investigate the tissue-specific roles of Fgfr2 in external genital development by generating conditional deletions of Fgfr2 in each of these cell types. Conditional deletion of Fgfr2 results in two distinct phenotypes: endodermal Fgfr2 deletion causes mild hypospadias and inhibits maturation of a complex urethral epithelium, whereas loss of ectodermal Fgfr2 results in severe hypospadias and absence of the ventral prepuce. Although these cell type-specific mutants exhibit distinctive genital anomalies, cellular analysis reveals that Fgfr2 regulates epithelial maturation and cell cycle progression in the urethral endoderm and in the surface ectoderm. The unexpected finding that ectodermal deletion of Fgfr2 results in the most severe hypospadias highlights a major role for Fgfr2 in the developing genital surface epithelium, where epithelial maturation is required for maintenance of a closed urethral tube. These results demonstrate that urethral tubulogenesis, prepuce morphogenesis, and sexually dimorphic patterning of the lower urethra are controlled by discrete regions of Fgfr2 activity.
Collapse
Affiliation(s)
- Marissa L Gredler
- Department of Biology, UF Genetics Institute, University of Florida, PO Box 103610, Gainesville, FL 32611, USA
| | - Ashley W Seifert
- Department of Biology, UF Genetics Institute, University of Florida, PO Box 103610, Gainesville, FL 32611, USA
| | - Martin J Cohn
- Department of Biology, UF Genetics Institute, University of Florida, PO Box 103610, Gainesville, FL 32611, USA Howard Hughes Medical Institute, Department of Molecular Genetics and Microbiology, University of Florida, PO Box 103610, Gainesville, FL 32611, USA
| |
Collapse
|
28
|
Sequencing of the DKK1 gene in patients with anorectal malformations and hypospadias. Eur J Pediatr 2015; 174:583-7. [PMID: 25319845 DOI: 10.1007/s00431-014-2436-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 09/30/2014] [Accepted: 10/02/2014] [Indexed: 01/08/2023]
Abstract
UNLABELLED Anorectal malformations (ARM) are rare congenital malformations of the gastrointestinal tract. Approximately 60% of the patients have additional congenital malformations, such as hypospadias. A recently published article showed that deletion of one single gene, dickkopf WNT signaling pathway inhibitor-1 (Dkk1), resulted in an imperforate anus with rectourinary fistula and preputial hypospadias in mice. To determine whether DKK1 also plays a role in the etiology of ARM and hypospadias in humans, we sequenced the four exons of the DKK1 gene in 17 patients affected with both ARM and hypospadias. No new potential disease-causing variant was identified. However, we detected a known non-synonymous variant in one patient, which was predicted in silico to be damaging, and the corresponding unaffected amino acid is highly conserved. CONCLUSION In this human study, a potential interesting non-synonymous variant was found in the DKK1 gene. Whether this variant plays a contributory role in the genesis of ARM or hypospadias would require a much larger study.
Collapse
|
29
|
Svechnikov K, Stukenborg JB, Savchuck I, Söder O. Similar causes of various reproductive disorders in early life. Asian J Androl 2014; 16:50-9. [PMID: 24369133 PMCID: PMC3901882 DOI: 10.4103/1008-682x.122199] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
During the past few decades, scientific evidence has been accumulated concerning the possible adverse effects of the exposure to environmental chemicals on the well-being of wildlife and human populations. One large and growing group of such compounds of anthropogenic or natural origin is referred to as endocrine-disrupting chemicals (EDCs), due to their deleterious action on the endocrine system. This concern was first focused on the control of reproductive function particularly in males, but has later been expanded to include all possible endocrine functions. The present review describes the underlying physiology behind the cascade of developmental events that occur during sexual differentiation of males and the specific role of androgen in the masculinization process and proper organogenesis of the external male genitalia. The impact of the genetic background, environmental exposures and lifestyle factors in the etiology of hypospadias, cryptorchidism and testicular cancer are reviewed and the possible role of EDCs in the development of these reproductive disorders is discussed critically. Finally, the possible direct and programming effects of exposures in utero to widely use therapeutic compounds, environmental estrogens and other chemicals on the incidence of reproductive abnormalities and poor semen quality in humans are also highlighted.
Collapse
Affiliation(s)
| | | | | | - Olle Söder
- Department of Women's and Children's Health, Paediatric Endocrinology Unit, Karolinska Institutet and University Hospital, Stockholm, Sweden
| |
Collapse
|
30
|
The power and perils of animal models with urogenital anomalies: handle with care. J Pediatr Urol 2014; 10:699-705. [PMID: 24768568 PMCID: PMC4454504 DOI: 10.1016/j.jpurol.2014.03.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 03/04/2014] [Indexed: 11/24/2022]
Abstract
Congenital abnormalities of the urogenital tracts form a major part of clinical practice for paediatric urologists, but their knowledge of normal and abnormal development is often limited. Advances in understanding frequently come from studying experimental findings from animal models, however, most clinicians underestimate both the power and perils of extrapolating scientific knowledge from animals. In this review, the key issues that urologists need to understand in order to link animal studies to clinical practice are discussed. Urologists must avoid the traps of anthropomorphism (assuming humans are always the same as animal models) or anthropocentrism (assuming humans are too different from animal models). This review used two common disorders: hypospadias and undescended testes.
Collapse
|
31
|
Murashima A, Kishigami S, Thomson A, Yamada G. Androgens and mammalian male reproductive tract development. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:163-70. [PMID: 24875095 DOI: 10.1016/j.bbagrm.2014.05.020] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/28/2014] [Accepted: 05/19/2014] [Indexed: 12/31/2022]
Abstract
One of the main functions of androgen is in the sexually dimorphic development of the male reproductive tissues. During embryogenesis, androgen determines the morphogenesis of male specific organs, such as the epididymis, seminal vesicle, prostate and penis. Despite the critical function of androgens in masculinization, the downstream molecular mechanisms of androgen signaling are poorly understood. Tissue recombination experiments and tissue specific androgen receptor (AR) knockout mouse studies have revealed epithelial or mesenchymal specific androgen-AR signaling functions. These findings also indicate that epithelial-mesenchymal interactions are a key feature of AR specific activity, and paracrine growth factor action may mediate some of the effects of androgens. This review focuses on mouse models showing the interactions of androgen and growth factor pathways that promote the sexual differentiation of reproductive organs. Recent studies investigating context dependent AR target genes are also discussed. This article is part of a Special Issue entitled: Nuclear receptors in animal development.
Collapse
Affiliation(s)
- Aki Murashima
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Kimiidera 811-1, Wakayama 641-8509, Wakayama, Japan
| | - Satoshi Kishigami
- Faculty of Biology-Oriented Science and Technology, Kinki University, Kinokawa 649-6493, Wakayama, Japan
| | - Axel Thomson
- Department of Urology, McGill University Health Centre, 1650 Cedar Av, Montreal, Québec, H3A 1A4, Canada
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Kimiidera 811-1, Wakayama 641-8509, Wakayama, Japan.
| |
Collapse
|
32
|
Nur BG, Pehlivanoğlu S, Mıhçı E, Calışkan M, Demir D, Alper OM, Kayserili H, Lüleci G. Clinicogenetic study of Turkish patients with syndromic craniosynostosis and literature review. Pediatr Neurol 2014; 50:482-90. [PMID: 24656465 DOI: 10.1016/j.pediatrneurol.2014.01.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 12/28/2013] [Accepted: 01/03/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND Fibroblast growth factor receptor 2 mutations have been associated with the craniosynostotic conditions of Apert, Crouzon, Pfeiffer, Saethre-Chotzen, Jackson-Weiss, Beare-Stevenson cutis gyrata, and Antley-Bixler syndromes in various ethnic groups. METHODS Thirty-three unrelated Turkish patients (12 with Apert syndrome, 14 with Crouzon syndrome, six with Pfeiffer syndrome, and one with Saethre-Chotzen syndrome) and 67 nonsyndromic craniosynostosis patients were screened for mutations in exons IIIa and IIIc of the FGFR2 gene by denaturing high-performance liquid chromatography and confirmed by direct sequencing. RESULTS We detected several pathogenic mutations in 11/33 (33%) patients with Apert syndrome (four with p.Pro253Arg; seven with p.Ser252Trp) and 8/33 (24%) patients with Crouzon syndrome (three with p.Trp290Arg, one with p.Cys342Tyr, p.Cys278Phe, p.Gln289Pro, and a novel p.Tyr340Asn mutation) and five (15%) with Pfeiffer syndrome (p.Cys342Arg, p.Pro253Arg, p.Trp290Arg, and p.Ser351Cys). No FGFR2 gene mutation was detected in any of the patients with Saethre-Chotzen syndrome and nonsyndromic craniosynostosis. CONCLUSIONS Our results indicate that the majority of Turkish patients with syndromic craniosynostosis have detectable genetic changes with an overall frequency of 72.7%. Because this is the first molecular genetic report from a Turkish cohort, the identified spectrum profile of FGFR2 mutations of the syndromic craniosynostotic patients would be very helpful for understanding the genotype-phenotype relationship and has a great value for diagnosis, prognosis, and genetic counseling.
Collapse
Affiliation(s)
- Banu G Nur
- Department of Pediatric Genetics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Suray Pehlivanoğlu
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Ercan Mıhçı
- Department of Pediatric Genetics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Mualla Calışkan
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Durkadın Demir
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Ozgül M Alper
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya, Turkey.
| | - Hülya Kayserili
- Department of Medical Genetics, Institute of Children's Health, Faculty of Medicine, Istanbul University, Çapa, İstanbul, Turkey
| | - Güven Lüleci
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
33
|
Shih EM, Graham JM. Review of genetic and environmental factors leading to hypospadias. Eur J Med Genet 2014; 57:453-63. [PMID: 24657417 DOI: 10.1016/j.ejmg.2014.03.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/04/2014] [Indexed: 10/25/2022]
Abstract
Hypospadias is one of the most common congenital malformations, affecting about 4-6 males per 1000 male births, and ranging in severity from a urethral meatus that is slightly off-center to a meatus in the perineal area. Over the past three decades its prevalence may have increased due to changes in reporting of mild cases and/or increased survival of low birth weight infants due to improved neonatal care. However, despite the increasing numbers of males with hypospadias, the overall etiology remains unclear and likely multifactorial in nature. The purpose of this review article is to provide a comprehensive overview of the various factors implicated in hypospadias etiology, including genetic and environmental factors. In addition, we list syndromes in which hypospadias is a relatively common association and delineate the areas that require further investigation in an effort to understand this condition.
Collapse
Affiliation(s)
- Erin M Shih
- Center for Endocrinology, Diabetes, and Metabolism, Children's Hospital Los Angeles, 4650 Sunset Blvd, #61, Los Angeles, CA 90027, USA.
| | - John M Graham
- Medical Genetics Institute, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
34
|
Ipulan LA, Suzuki K, Matsushita S, Suzuki H, Okazawa M, Jacinto S, Hirai SI, Yamada G. Development of the external genitalia and their sexual dimorphic regulation in mice. Sex Dev 2014; 8:297-310. [PMID: 24503953 DOI: 10.1159/000357932] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The study of the external genitalia is divided into 2 developmental stages: the formation and growth of a bipotential genital tubercle (GT) and the sexual differentiation of the male and female GT. The sexually dimorphic processes, which occur during the second part of GT differentiation, are suggested to be governed by androgen signaling and more recently crosstalk with other signaling factors. The process of elucidating the regulatory mechanisms of hormone signaling towards other signaling networks in the GT is still in its early stages. Nevertheless, it is becoming a productive area of research. This review summarizes various studies on the development of the murine GT and the defining characteristics of a masculinized GT and presents the different signaling pathways possibly involved during masculinization.
Collapse
Affiliation(s)
- Lerrie Ann Ipulan
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Ohnesorg T, Vilain E, Sinclair AH. The genetics of disorders of sex development in humans. Sex Dev 2014; 8:262-72. [PMID: 24504012 DOI: 10.1159/000357956] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
One of the defining events during human embryonic development with the most far-reaching effects for the individual is whether the embryo develops as male or female. The crucial step in this process is the differentiation of the bipotential embryonic gonads into either testes or ovaries. If the embryo inherits X and Y sex chromosomes, the Y-linked SRY (sex determining region in Y) gene initiates a network of genes that results in a functional testis and ultimately a male phenotype. By contrast, in an embryo with 2 X chromosomes, the undifferentiated gonad develops as an ovary resulting in a female phenotype. Perturbation of any of the genes in either the testicular or ovarian developmental pathway can result in individuals with disorders of sex development. In this review, we provide a summary of known components of testicular or ovarian pathways and their antagonistic actions and give a brief overview of new technologies currently used to identify the missing pieces of the sex development network.
Collapse
Affiliation(s)
- Thomas Ohnesorg
- Murdoch Children's Research Institute and Department of Paediatrics, The University of Melbourne, Royal Children's Hospital, Melbourne, Vic., Australia
| | | | | |
Collapse
|
36
|
Ching ST, Cunha GR, Baskin LS, Basson MA, Klein OD. Coordinated activity of Spry1 and Spry2 is required for normal development of the external genitalia. Dev Biol 2013; 386:1-11. [PMID: 24361260 DOI: 10.1016/j.ydbio.2013.12.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 12/05/2013] [Accepted: 12/10/2013] [Indexed: 11/16/2022]
Abstract
Development of the mammalian external genitalia is controlled by a network of signaling molecules and transcription factors. Because FGF signaling plays a central role in this complicated morphogenetic process, we investigated the role of Sprouty genes, which are important intracellular modulators of FGF signaling, during embryonic development of the external genitalia in mice. We found that Sprouty genes are expressed by the urethral epithelium during embryogenesis, and that they have a critical function during urethral canalization and fusion. Development of the genital tubercle (GT), the anlage of the prepuce and glans penis in males and glans clitoris in females, was severely affected in male embryos carrying null alleles of both Spry1 and Spry2. In Spry1(-/-);Spry2(-/-) embryos, the internal tubular urethra was absent, and urothelial morphology and organization was abnormal. These effects were due, in part, to elevated levels of epithelial cell proliferation in Spry1(-/-);Spry2(-/-) embryos. Despite changes in overall organization, terminal differentiation of the urothelium was not significantly affected. Characterization of the molecular pathways that regulate normal GT development confirmed that deletion of Sprouty genes leads to elevated FGF signaling, whereas levels of signaling in other cascades were largely preserved. Together, these results show that levels of FGF signaling must be tightly regulated during embryonic development of the external genitalia in mice, and that this regulation is mediated in part through the activity of Sprouty gene products.
Collapse
Affiliation(s)
- Saunders T Ching
- Department of Orofacial Sciences, University of California, San Francisco, United States; Department of Urology, University of California, San Francisco, United States
| | - Gerald R Cunha
- Department of Urology, University of California, San Francisco, United States
| | - Laurence S Baskin
- Department of Urology, University of California, San Francisco, United States
| | - M Albert Basson
- Department of Craniofacial Development and Stem Cell Biology, King's College, London, UK
| | - Ophir D Klein
- Department of Orofacial Sciences, University of California, San Francisco, United States; Program in Craniofacial and Mesenchymal Biology, University of California, San Francisco, United States; Institute for Human Genetics, University of California, San Francisco, United States; Department of Pediatrics, University of California, San Francisco, United States.
| |
Collapse
|
37
|
Carmichael SL, Ma C, Choudhry S, Lammer EJ, Witte JS, Shaw GM. Hypospadias and genes related to genital tubercle and early urethral development. J Urol 2013; 190:1884-92. [PMID: 23727413 PMCID: PMC4103581 DOI: 10.1016/j.juro.2013.05.061] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2013] [Indexed: 11/19/2022]
Abstract
PURPOSE We determined whether variants in genes associated with genital tubercle (the anlage for the penis) and early urethral development were associated with hypospadias in humans. MATERIALS AND METHODS We examined 293 relatively common tag single nucleotide polymorphisms in BMP4, BMP7, FGF8, FGF10, FGFR2, HOXA13, HOXD13, HOXA4, HOXB6, SRY, WT1, WTAP, SHH, GLI1, GLI2 and GLI3. The analysis included 624 cases (81 mild, 319 moderate, 209 severe, 15 undetermined severity) and 844 population based nonmalformed male controls born in California from 1990 to 2003. RESULTS There were 28 single nucleotide polymorphisms for which any of the comparisons (ie overall or for a specific severity) had a p value of less than 0.01. The homozygous variant genotypes for 4 single nucleotide polymorphisms in BMP7 were associated with at least a twofold increased risk of hypospadias regardless of severity. Five single nucleotide polymorphisms for FGF10 were associated with threefold to fourfold increased risks, regardless of severity. For 4 of them the results were restricted to whites. For GLI1, GLI2 and GLI3 there were 12 associated single nucleotide polymorphisms but results were inconsistent by severity and race/ethnicity. For SHH 1 single nucleotide polymorphism was associated with a 2.4-fold increased risk of moderate hypospadias. For WT1 6 single nucleotide polymorphisms were associated with approximately a twofold increased risk, primarily for severe hypospadias. CONCLUSIONS This study provides evidence that single nucleotide polymorphisms in several genes that contribute to genital tubercle and early urethral development are associated with hypospadias risk.
Collapse
Affiliation(s)
- Suzan L Carmichael
- Department of Pediatrics, Division of Neonatology, Stanford University School of Medicine, Stanford, California.
| | | | | | | | | | | |
Collapse
|
38
|
Baxter RM, Vilain E. Translational genetics for diagnosis of human disorders of sex development. Annu Rev Genomics Hum Genet 2013; 14:371-92. [PMID: 23875799 DOI: 10.1146/annurev-genom-091212-153417] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Disorders of sex development (DSDs) are congenital conditions with discrepancies between the chromosomal, gonadal, and phenotypic sex of the individual. Such disorders have historically been difficult to diagnose and cause great stress to patients and their families. Genetic analysis of human samples has been instrumental in elucidating the molecules and pathways involved in the development of the bipotential gonad into a functioning testis or ovary. However, many DSD patients still do not receive a genetic diagnosis. New genetic and genomic technologies are expanding our knowledge of the underlying mechanism of DSDs and opening new avenues for clinical diagnosis. We review the genetic technologies that have elucidated the genes that are well established in sex determination in humans, discuss findings from more recent genomic technologies, and propose a new paradigm for clinical diagnosis of DSDs.
Collapse
|
39
|
Blaschko SD, Cunha GR, Baskin LS. Molecular mechanisms of external genitalia development. Differentiation 2012; 84:261-8. [PMID: 22790208 DOI: 10.1016/j.diff.2012.06.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 05/09/2012] [Accepted: 06/16/2012] [Indexed: 12/21/2022]
Abstract
External genitalia development occurs through a combination of hormone independent, hormone dependent, and endocrine pathways. Perturbation of these pathways can lead to abnormal external genitalia development. We review human and animal mechanisms of normal and abnormal external genitalia development, and we evaluate abnormal mechanisms that lead to hypospadias. We also discuss recent laboratory findings that further our understanding of animal models of hypospadias.
Collapse
Affiliation(s)
- Sarah D Blaschko
- University of California San Francisco, Department of Urology, 400 Parnassus Avenue, A610, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
40
|
Carmichael SL, Shaw GM, Lammer EJ. Environmental and genetic contributors to hypospadias: a review of the epidemiologic evidence. BIRTH DEFECTS RESEARCH. PART A, CLINICAL AND MOLECULAR TERATOLOGY 2012; 94:499-510. [PMID: 22678668 PMCID: PMC3393839 DOI: 10.1002/bdra.23021] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 03/22/2012] [Accepted: 03/28/2012] [Indexed: 12/28/2022]
Abstract
This review evaluates current knowledge related to trends in the prevalence of hypospadias, the association of hypospadias with endocrine-disrupting exposures, and the potential contribution of genetic susceptibility to its etiology. The review focuses on epidemiologic evidence. Increasing prevalence of hypospadias has been observed, but such increases tend to be localized to specific regions or time periods. Thus, generalized statements that hypospadias is increasing are unsupported. Owing to the limitations of study designs and inconsistent results, firm conclusions cannot be made regarding the association of endocrine-disrupting exposures with hypospadias. Studies with more rigorous study designs (e.g., larger and more detailed phenotypes) and exposure assessment that encompasses more breadth and depth (e.g., specific endocrine-related chemicals) will be critical to make better inferences about these important environmental exposures. Many candidate genes for hypospadias have been identified, but few of them have been examined to an extent that enables solid conclusions. Further study is needed that includes larger sample sizes, comparison groups that are more representative of the populations from which the cases were derived, phenotype-specific analyses, and more extensive exploration of variants. In conclusion, examining the associations of environmental and genetic factors with hypospadias remain important areas of inquiry, although our actual understanding of their contribution to hypospadias risk in humans is currently limited.
Collapse
Affiliation(s)
- Suzan L Carmichael
- Department of Pediatrics, Division of Neonatology, Stanford University School of Medicine, California, USA.
| | | | | |
Collapse
|
41
|
Mulatinho MV, de Carvalho Serao CL, Scalco F, Hardekopf D, Pekova S, Mrasek K, Liehr T, Weise A, Rao N, Llerena JC. Severe intellectual disability, omphalocele, hypospadia and high blood pressure associated to a deletion at 2q22.1q22.3: case report. Mol Cytogenet 2012; 5:30. [PMID: 22686481 PMCID: PMC3407782 DOI: 10.1186/1755-8166-5-30] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Accepted: 06/11/2012] [Indexed: 01/11/2023] Open
Abstract
Background Recently, array-comparative genomic hybridization (aCGH) platforms have significantly improved the resolution of chromosomal analysis allowing the identification of genomic copy number gains and losses smaller than 5 Mb. Here we report on a young man with unexplained severe mental retardation, autism spectrum disorder, congenital malformations comprising hypospadia and omphalocele, and episodes of high blood pressure. An ~ 6 Mb interstitial deletion that includes the causative genes is identified by oligonucleotide-based aCGH. Results Our index case exhibited a de novo chromosomal abnormality at 2q22 [del(2)(q22.1q22.3)dn] which was not visible at the 550 haploid band level. The deleted region includes eight genes: HNMT, SPOPL, NXPH2, LOC64702, LRP1B, KYNU, ARHGAP15 and GTDC1. Discussion aCGH revealed an ~ 6 Mb deletion in 2q22.1 to 2q22.3 in an as-yet unique clinical case associated with intellectual disability, congenital malformations and autism spectrum disorder. Interestingly, the deletion is co-localized with a fragile site (FRA2K), which could be involved in the formation of this chromosomal aberration. Further studies are needed to determine if deletions of 2q22.1 to 2q22.3 define a new microdeletion syndrome.
Collapse
Affiliation(s)
- Milene Vianna Mulatinho
- Instituto Fernandes Figueira, IFF/FIOCRUZ, Departamento de Genética Médica, Av, Rui Barbosa, 716, Flamengo, Rio de Janeiro, RJ 22250-020, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
van der Zanden LFM, van Rooij IALM, Feitz WFJ, Franke B, Knoers NVAM, Roeleveld N. Aetiology of hypospadias: a systematic review of genes and environment. Hum Reprod Update 2012; 18:260-83. [PMID: 22371315 DOI: 10.1093/humupd/dms002] [Citation(s) in RCA: 183] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Hypospadias is a common congenital malformation of the male external genitalia. Most cases have an unknown aetiology, which is probably a mix of monogenic and multifactorial forms, implicating both genes and environmental factors. This review summarizes current knowledge about the aetiology of hypospadias. METHODS Pubmed was used to identify studies on hypospadias aetiology published between January 1995 and February 2011. Reference lists of the selected manuscripts were also searched to identify additional studies, including those published before 1995. RESULTS The search provided 922 articles and 169 articles were selected for this review. Studies screening groups of patients with hypospadias for single gene defects found mutations in WT1, SF1, BMP4, BMP7, HOXA4, HOXB6, FGF8, FGFR2, AR, HSD3B2, SRD5A2, ATF3, MAMLD1, MID1 and BNC2. However, most investigators are convinced that single mutations do not cause the majority of isolated hypospadias cases. Indeed, associations were found with polymorphisms in FGF8, FGFR2, AR, HSD17B3, SRD5A2, ESR1, ESR2, ATF3, MAMLD1, DGKK, MID1, CYP1A1, GSTM1 and GSTT1. In addition, gene expression studies indentified CTGF, CYR61 and EGF as candidate genes. Environmental factors consistently implicated in hypospadias are low birthweight, maternal hypertension and pre-eclampsia, suggesting that placental insufficiency may play an important role in hypospadias aetiology. Exogenous endocrine-disrupting chemicals have the potential to induce hypospadias but it is unclear whether human exposure is high enough to exert this effect. Other environmental factors have also been associated with hypospadias but, for most, the results are inconsistent. CONCLUSIONS Although a number of contributors to the aetiology of hypospadias have been identified, the majority of risk factors remain unknown.
Collapse
Affiliation(s)
- L F M van der Zanden
- Department of Epidemiology, Biostatistics and HTA, Radboud University Nijmegen Medical Centre, 6500 HB Nijmegen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
43
|
Mutation analysis of NR5A1 encoding steroidogenic factor 1 in 77 patients with 46, XY disorders of sex development (DSD) including hypospadias. PLoS One 2011; 6:e24117. [PMID: 22028768 PMCID: PMC3197579 DOI: 10.1371/journal.pone.0024117] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 08/01/2011] [Indexed: 11/19/2022] Open
Abstract
Background Mutations of the NR5A1 gene encoding steroidogenic factor-1 have been reported in association with a wide spectrum of 46,XY DSD (Disorder of Sex Development) phenotypes including severe forms of hypospadias. Methodology/Principal Findings We evaluated the frequency of NR5A1 gene mutations in a large series of patients presenting with 46,XY DSD and hypospadias. Based on their clinical presentation 77 patients were classified either as complete or partial gonadal dysgenesis (uterus seen at genitography and/or surgery, n = 11), ambiguous external genitalia without uterus (n = 33) or hypospadias (n = 33). We identified heterozygous NR5A1 mutations in 4 cases of ambiguous external genitalia without uterus (12.1%; p.Trp279Arg, pArg39Pro, c.390delG, c140_141insCACG) and a de novo missense mutation in one case with distal hypospadias (3%; p.Arg313Cys). Mutant proteins showed reduced transactivation activity and mutants p.Arg39Pro and p.Arg313Cys did not synergize with the GATA4 cofactor to stimulate reporter gene activity, although they retained their ability to physically interact with the GATA4 protein. Conclusions/Significance Mutations in NR5A1 were observed in 5/77 (6.5%) cases of 46,XY DSD including hypospadias. Excluding the cases of 46,XY gonadal dysgenesis the incidence of NR5A1 mutations was 5/66 (7.6%). An individual with isolated distal hypopadias carried a de novo heterozygous missense mutation, thus extending the range of phenotypes associated with NR5A1 mutations and suggesting that this group of patients should be screened for NR5A1 mutations.
Collapse
|
44
|
Zhang LF, Qin C, Wei YF, Wang Y, Chang JK, Mi YY, Ma L, Jiang JT, Feng NH, Wang ZJ, Zhang W. Differential expression of the Wnt/β-catenin pathway in the genital tubercle (GT) of fetal male rat following maternal exposure to di-n-butyl phthalate (DBP). Syst Biol Reprod Med 2011; 57:244-50. [DOI: 10.3109/19396368.2011.577509] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
45
|
Kalfa N, Philibert P, Baskin LS, Sultan C. Hypospadias: interactions between environment and genetics. Mol Cell Endocrinol 2011; 335:89-95. [PMID: 21256920 DOI: 10.1016/j.mce.2011.01.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 01/03/2011] [Accepted: 01/07/2011] [Indexed: 10/18/2022]
Abstract
Hypospadias is one of the most common congenital malformations. It is considered to be a mild form of the 46,XY disorders of sex development (DSD), but its precise etiology remains to be elucidated. Compromised androgen synthesis or effects can cause this frequent malformation, although the mutational analyses of the genes involved in androgen actions have identified abnormalities in only a very small portion of patients. The overwhelming majority of cases remain unexplained and hypospadias may be a highly heterogeneous condition subject to multiple genetic and environmental factors. We here review the recent advances in this field and discuss the potential interactions between the environment and genetics.
Collapse
Affiliation(s)
- N Kalfa
- Service d'Hormonologie, Hôpital Lapeyronie, CHU de Montpellier et UM1, Montpellier, France
| | | | | | | |
Collapse
|
46
|
Toppari J, Virtanen HE, Main KM, Skakkebaek NE. Cryptorchidism and hypospadias as a sign of testicular dysgenesis syndrome (TDS): environmental connection. ACTA ACUST UNITED AC 2011; 88:910-9. [PMID: 20865786 DOI: 10.1002/bdra.20707] [Citation(s) in RCA: 159] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cryptorchidism and hypospadias are common genital birth defects that affect 2-9% and 0.2-1% of male newborns, respectively. The incidence of both defects shows large geographic variation, and in several countries increasing trends have been reported. The conditions share many risk factors, and they are also interlinked to the risk of testis cancer and poor semen quality. Testicular Dysgenesis Syndrome (TDS) may underlie many cases of all these male reproductive health problems. Genetic defects in androgen production or action can cause both cryptorchidism and hypospadias, but these are not common. A monogenic reason for cryptorchidism or hypospadias has been identified only in a small proportion of all cases. Environmental effects appear to play a major role in TDS. Exposure to several persistent chemicals has been found to be associated with the risk of cryptorchidism, and exposure to anti-androgenic phthalates has been shown to be associated with hormonal changes predisposing to male reproductive problems. Despite progress in identification of endocrine-disrupting substances, we are still far from knowing all the risk factors for these birth defects, and advice for prevention must be based on precautionary principles.
Collapse
Affiliation(s)
- Jorma Toppari
- Departments of Physiology and Pediatrics, University of Turku, Kiinamyllynkatu 10, Turku, Finland.
| | | | | | | |
Collapse
|
47
|
Identification of de novo copy number variants associated with human disorders of sexual development. PLoS One 2010; 5:e15392. [PMID: 21048976 PMCID: PMC2964326 DOI: 10.1371/journal.pone.0015392] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 09/01/2010] [Indexed: 11/19/2022] Open
Abstract
Disorders of sexual development (DSD), ranging in severity from genital abnormalities to complete sex reversal, are among the most common human birth defects with incidence rates reaching almost 3%. Although causative alterations in key genes controlling gonad development have been identified, the majority of DSD cases remain unexplained. To improve the diagnosis, we screened 116 children born with idiopathic DSD using a clinically validated array-based comparative genomic hybridization platform. 8951 controls without urogenital defects were used to compare with our cohort of affected patients. Clinically relevant imbalances were found in 21.5% of the analyzed patients. Most anomalies (74.2%) evaded detection by the routinely ordered karyotype and were scattered across the genome in gene-enriched subtelomeric loci. Among these defects, confirmed de novo duplication and deletion events were noted on 1p36.33, 9p24.3 and 19q12-q13.11 for ambiguous genitalia, 10p14 and Xq28 for cryptorchidism and 12p13 and 16p11.2 for hypospadias. These variants were significantly associated with genitourinary defects (P = 6.08×10(-12)). The causality of defects observed in 5p15.3, 9p24.3, 22q12.1 and Xq28 was supported by the presence of overlapping chromosomal rearrangements in several unrelated patients. In addition to known gonad determining genes including SRY and DMRT1, novel candidate genes such as FGFR2, KANK1, ADCY2 and ZEB2 were encompassed. The identification of risk germline rearrangements for urogenital birth defects may impact diagnosis and genetic counseling and contribute to the elucidation of the molecular mechanisms underlying the pathogenesis of human sexual development.
Collapse
|
48
|
Genetic pathway of external genitalia formation and molecular etiology of hypospadias. J Pediatr Urol 2010; 6:346-54. [PMID: 19995686 DOI: 10.1016/j.jpurol.2009.11.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Accepted: 11/10/2009] [Indexed: 11/23/2022]
Abstract
Hypospadias is one of the most common congenital disorders in males. Impaired fetal androgen action interferes with masculinization, including external genitalia formation, and can result in this anomaly; however, the molecular etiology remains unknown. Recent molecular approaches, including gene-targeting approaches in mice and single nucleotide polymorphisms analyses in humans, might provide an opportunity to identify the causative and risk factors of this anomaly. Several genes, such as sonic hedgehog, fibroblast growth factors, bone morphogenetic proteins, homeobox genes, and the Wnt family regulate external genitalia formation. Mastermind-like domain containing 1/chromosome X open reading frame 6 mutation and activating transcription factor 3 variants have been shown to be associated with the incidence of isolated hypospadias. In addition, this anomaly may be associated with a specific haplotype of the gene for estrogen receptor alpha, which mediates the estrogenic effects of environmental endocrine disruptors, and the effects of these disruptors on external genitalia formation might depend on individual genetic susceptibility. These molecular studies will refine our knowledge of the genetic mechanism involved in external genitalia formation, and lead to new strategies for the clinical management of hypospadias.
Collapse
|
49
|
Chiu HS, Szucsik JC, Georgas KM, Jones JL, Rumballe BA, Tang D, Grimmond SM, Lewis AG, Aronow BJ, Lessard JL, Little MH. Comparative gene expression analysis of genital tubercle development reveals a putative appendicular Wnt7 network for the epidermal differentiation. Dev Biol 2010; 344:1071-87. [PMID: 20510229 DOI: 10.1016/j.ydbio.2010.05.495] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 04/23/2010] [Accepted: 05/15/2010] [Indexed: 02/08/2023]
Abstract
Here we describe the first detailed catalog of gene expression in the developing lower urinary tract (LUT), including epithelial and mesenchymal portions of the developing bladder, urogenital sinus, urethra, and genital tubercle (GT) at E13 and E14. Top compartment-specific genes implicated by the microarray data were validated using whole-mount in situ hybridization (ISH) over the entire LUT. To demonstrate the potential of this resource to implicate developmentally critical features, we focused on gene expression patterns and pathways in the sexually indeterminate, androgen-independent GT. GT expression patterns reinforced the proposed similarities between development of GT, limb, and craniofacial prominences. Comparison of spatial expression patterns predicted a network of Wnt7a-associated GT-enriched epithelial genes, including Gjb2, Dsc3, Krt5, and Sostdc1. Known from other contexts, these genes are associated with normal epidermal differentiation, with disruptions in Dsc3 and Gjb2 showing palmo-plantar keratoderma in the limb. We propose that this gene network contributes to normal foreskin, scrotum, and labial development. As several of these genes are known to be regulated by, or contain cis elements responsive to retinoic acid, estrogen, or androgen, this implicates this pathway in the later androgen-dependent development of the GT.
Collapse
Affiliation(s)
- Han Sheng Chiu
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia 4072, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
van der Zanden LFM, van Rooij IALM, Feitz WFJ, Vermeulen SHHM, Kiemeney LALM, Knoers NVAM, Roeleveld N, Franke B. Genetics of hypospadias: are single-nucleotide polymorphisms in SRD5A2, ESR1, ESR2, and ATF3 really associated with the malformation? J Clin Endocrinol Metab 2010; 95:2384-90. [PMID: 20215396 DOI: 10.1210/jc.2009-2101] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
CONTEXT Hypospadias is a common congenital malformation of the male external genitalia with a multifactorial etiology. Little is known about the genes involved in hypospadias. A few genetic associations have been reported but mainly in studies of small sample size. Most of these associations have not been replicated. OBJECTIVE The aim of this study was to investigate whether previously reported associations for four single-nucleotide polymorphisms (SNPs) in genes involved in hormonal pathways could be replicated in a large Dutch hypospadias sample. The SNPs investigated are rs523349 in steroid-5 alpha-reductase (SRD5A2), rs6932902 in estrogen receptor 1 (ESR1), rs2987983 in ESR2, and rs11119982 in activating transcription factor 3 (ATF3). DESIGN, PARTICIPANTS, AND METHODS We genotyped 620 Caucasian hypospadias cases and 596 controls for these SNPs using TaqMan-based genotyping. RESULTS We did not replicate the associations of the SNPs in SRD5A2 and ESR1 with hypospadias. The SNPs in ESR2 and ATF3 were borderline associated with hypospadias [odds ratios 0.9 (95% confidence interval 0.7-1.0) and 1.2 (95% confidence interval 1.0-1.4), respectively] but in the opposite direction compared with earlier publications. Stratification according to localization of the urethral opening produced comparable results in the subgroups. CONCLUSIONS The lack of consistency between our and previously performed studies might represent spurious results or chance findings in our or the earlier studies, differences in criteria used to select the study populations, or a real difference between populations, i.e. different genes contributing to disease risk. These results once again confirm the importance of replication in genetic association approaches.
Collapse
Affiliation(s)
- Loes F M van der Zanden
- Department of Epidemiology, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|