1
|
Naghi P, Zaha IA, Stefan L, Sorian A, Marcu A, Sachelarie L, Huniadi A. Teratozoospermia and Embryo Development: The Significance of Sperm Selection in In Vitro Fertilization Success. J Clin Med 2025; 14:3763. [PMID: 40507525 PMCID: PMC12156187 DOI: 10.3390/jcm14113763] [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: 03/19/2025] [Revised: 04/26/2025] [Accepted: 05/24/2025] [Indexed: 06/16/2025] Open
Abstract
Background: Sperm morphology is a key factor influencing fertilization and embryo development in assisted reproductive technology (ART). However, the predictive value of sperm deformity indices and selection techniques remains debated. This study evaluated the impact of teratozoospermia on fertilization, blastocyst formation, and embryo quality, comparing conventional and microfluidic sperm selection methods. Methods: A retrospective analysis was conducted on ART cycles involving patients with teratozoospermia. Sperm selection was performed using density gradient centrifugation (DGC) or microfluidic sperm sorting (MFSS). The correlations between the Sperm Deformity Index (SDI), Multiple Anomalies Index (MAI), and Teratozoopermia Index (TZI) with fertilization rates, blastocyst formation, and embryo quality were assessed. Statistical analysis included correlation tests, receiver operating characteristic (ROC) curves, and independent samples t-tests. Results: Patients with severe teratozoospermia exhibited lower fertilization rates (p < 0.01) and reduced blastocyst formation (p = 0.02). The SDI and MAI showed moderate negative correlations with fertilization (r = -0.15 and r = -0.25, respectively) and blastocyst development (r = -0.20 and r = -0.30, respectively), while the TZI had only weak associations (r = -0.10 and r = -0.15, respectively). ROC analysis demonstrated that the SDI and MAI were moderate predictors of embryo viability (AUC = 0.70 and 0.75, respectively). Patients who underwent microfluidic sperm selection had higher fertilization rates (p = 0.03) and improved blastocyst quality (p = 0.04) than those processed with DGC. Conclusions: Severe teratozoospermia negatively affects fertilization and blastocyst formation, with the SDI and MAI showing moderate predictive value for embryo development. The use of microfluidic sperm selection significantly improved embryo quality, supporting its clinical relevance in ART.
Collapse
Affiliation(s)
- Petronela Naghi
- Calla—Infertility Diagnostic and Treatment Center, Constantin A. Rosetti Street, 410103 Oradea, Romania; (P.N.); (I.A.Z.); (A.S.); (A.M.); (A.H.)
| | - Ioana Alexandra Zaha
- Calla—Infertility Diagnostic and Treatment Center, Constantin A. Rosetti Street, 410103 Oradea, Romania; (P.N.); (I.A.Z.); (A.S.); (A.M.); (A.H.)
- Faculty of Medicine and Pharmacy, University of Oradea, 1st December Square 10, 410073 Oradea, Romania
- Pelican Clinical Hospital, Corneliu Coposu Street 2, 410450 Oradea, Romania
| | - Liana Stefan
- Calla—Infertility Diagnostic and Treatment Center, Constantin A. Rosetti Street, 410103 Oradea, Romania; (P.N.); (I.A.Z.); (A.S.); (A.M.); (A.H.)
- Faculty of Medicine and Pharmacy, University of Oradea, 1st December Square 10, 410073 Oradea, Romania
- Oradea County Hospital, Gheorghe Doja Street 65–67, 410169 Oradea, Romania
| | - Andrea Sorian
- Calla—Infertility Diagnostic and Treatment Center, Constantin A. Rosetti Street, 410103 Oradea, Romania; (P.N.); (I.A.Z.); (A.S.); (A.M.); (A.H.)
| | - Adelin Marcu
- Calla—Infertility Diagnostic and Treatment Center, Constantin A. Rosetti Street, 410103 Oradea, Romania; (P.N.); (I.A.Z.); (A.S.); (A.M.); (A.H.)
| | - Liliana Sachelarie
- Department of Clinical Discipline, Apollonia University, 700511 Iasi, Romania
| | - Anca Huniadi
- Calla—Infertility Diagnostic and Treatment Center, Constantin A. Rosetti Street, 410103 Oradea, Romania; (P.N.); (I.A.Z.); (A.S.); (A.M.); (A.H.)
- Faculty of Medicine and Pharmacy, University of Oradea, 1st December Square 10, 410073 Oradea, Romania
- Pelican Clinical Hospital, Corneliu Coposu Street 2, 410450 Oradea, Romania
- Oradea County Hospital, Gheorghe Doja Street 65–67, 410169 Oradea, Romania
| |
Collapse
|
2
|
Sethi S, Andrabi W, Mitra K, Rajender S. Case Report: A homozygous mutation in the SPAG17 gene in a case with oligoasthenoteratozoospermic infertility. FRONTIERS IN REPRODUCTIVE HEALTH 2025; 7:1554027. [PMID: 40330001 PMCID: PMC12053157 DOI: 10.3389/frph.2025.1554027] [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: 12/31/2024] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Background Defects in sperm size and form, known as teratozoospermia, can adversely impair sperm motility and its ability to fertilize an oocyte. Teratozoospermia has been most often linked with genetic abnormalities with close to 100 genes known. Objective The primary objective of this study was to investigate the genetic basis of oligoasthenoteratozoospermic infertility in an infertile man. Methods We performed the whole exome sequencing, followed by in silico filtration of observed genetic variations. Filtered rare variants were assessed for their pathogenic nature on the basis of scores assigned by various in-silico tools and their biological relevance to sperm structural development. The potential pathogenic mutation was validated by Sanger sequencing. Results Our study identified a homozygous substitution, c.4511A > G, in the SPAG17 gene as a potential pathogenic mutation associated with oligoasthenoteratozoospermic infertility in the case under investigation. The mutation resulted in the substitution of asparagine with serine at the 1504th amino acid position in a protein of 2,223 amino acids. This mutation shows a minor allele frequency of 0.0004671 in the gnomAD database. ACMG classification suggested this mutation to be likely pathogenic. Conclusion Our study identified a homozygous likely pathogenic mutation (c.4511A > G, Asn1504Ser) in the SPAG17 gene that explains oligoasthenoteratozoospermic infertility in the present case.
Collapse
Affiliation(s)
- Shruti Sethi
- Division of Endocrinology, Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Industrial Research (AcSIR), Ghaziabad, India
| | | | - Kalyan Mitra
- Division of Endocrinology, Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Industrial Research (AcSIR), Ghaziabad, India
| | - Singh Rajender
- Division of Endocrinology, Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Industrial Research (AcSIR), Ghaziabad, India
| |
Collapse
|
3
|
Milewska AJ, Kuczyńska A, Pawłowski M, Martynowicz I, Deluga-Białowarczuk S, Sieczyński P, Kuczyński W, Milewski R. The Influence of Cryopreservation on Sperm Morphology and Its Implications in Terms of Fractions of Higher-Quality Sperm. J Clin Med 2024; 13:7562. [PMID: 39768485 PMCID: PMC11727669 DOI: 10.3390/jcm13247562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Background/Objectives: Male infertility is a significant global health issue, comprising approx. 50% of all infertility cases. Semen cryopreservation, a critical component of assisted reproductive technologies (ARTs), is a method commonly used in a wide range of situations, including gonadotoxic treatments such as radiation or chemotherapy, hazardous occupational exposures, and various medical conditions. Although historically viewed as potentially damaging to sperm, recent findings suggest that cryopreservation, when performed with appropriate techniques, may in fact enhance semen quality by improving the proportion of healthy spermatozoa, particularly in terms of their morphological parameters. The aim of this study was to evaluate the impact of cryopreservation on sperm morphology and viability, utilizing advanced morphological assessments pre- and post-freezing. Methods: Semen samples were collected from 97 patients undergoing infertility treatment at the KRIOBANK clinic (Białystok, Poland). The semen was liquefied and prepared in the form of slides. Sperm morphology was then assessed using an OLYMPUS BX40 microscope at 60× magnification. Results: The findings of the study revealed significant improvements in sperm morphology, with increased percentages of normal sperm and reductions in deformation indices post-thaw. Conclusions: The findings indicate that optimized cryopreservation protocols may support the selection of higher-quality sperm, offering valuable benefits for ART applications. These results challenge certain past assumptions regarding the impact of cryopreservation and underscore the need for refined freezing techniques to maintain and potentially enhance semen quality for reproductive use.
Collapse
Affiliation(s)
- Anna Justyna Milewska
- Department of Biostatistics and Medical Informatics, Medical University of Bialystok, 15-295 Bialystok, Poland; (M.P.); (R.M.)
| | - Agnieszka Kuczyńska
- Kriobank Infertility Treatment Center, 15-879 Bialystok, Poland (I.M.); (S.D.-B.)
| | - Michał Pawłowski
- Department of Biostatistics and Medical Informatics, Medical University of Bialystok, 15-295 Bialystok, Poland; (M.P.); (R.M.)
| | - Iwo Martynowicz
- Kriobank Infertility Treatment Center, 15-879 Bialystok, Poland (I.M.); (S.D.-B.)
| | | | - Piotr Sieczyński
- Kriobank Infertility Treatment Center, 15-879 Bialystok, Poland (I.M.); (S.D.-B.)
| | - Waldemar Kuczyński
- Kriobank Infertility Treatment Center, 15-879 Bialystok, Poland (I.M.); (S.D.-B.)
| | - Robert Milewski
- Department of Biostatistics and Medical Informatics, Medical University of Bialystok, 15-295 Bialystok, Poland; (M.P.); (R.M.)
| |
Collapse
|
4
|
Arora M, Mehta P, Sethi S, Anifandis G, Samara M, Singh R. Genetic etiological spectrum of sperm morphological abnormalities. J Assist Reprod Genet 2024; 41:2877-2929. [PMID: 39417902 PMCID: PMC11621285 DOI: 10.1007/s10815-024-03274-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
PURPOSE Male infertility manifests in the form of a reduction in sperm count, sperm motility, or the loss of fertilizing ability. While the loss of sperm production can have mixed reasons, sperm structural defects, cumulatively known as teratozoospermia, have predominantly genetic bases. The aim of the present review is to undertake a comprehensive analysis of the genetic mutations leading to sperm morphological deformities/teratozoospermia. METHODS We undertook literature review for genes involved in sperm morphological abnormalities. The genes were classified according to the type of sperm defects they cause and on the basis of the level of evidence determined by the number of human studies and the availability of a mouse knockout. RESULTS Mutations in the SUN5, CEP112, BRDT, DNAH6, PMFBP1, TSGA10, and SPATA20 genes result in acephalic sperm; mutations in the DPY19L2, SPATA16, PICK1, CCNB3, CHPT1, PIWIL4, and TDRD9 genes cause globozoospermia; mutations in the AURKC gene cause macrozoospermia; mutations in the WDR12 gene cause tapered sperm head; mutations in the RNF220 and ADCY10 genes result in small sperm head; mutations in the AMZ2 gene lead to vacuolated head formation; mutations in the CC2D1B and KIAA1210 genes lead to pyriform head formation; mutations in the SEPT14, ZPBP1, FBXO43, ZCWPW1, KATNAL2, PNLDC1, and CCIN genes cause amorphous head; mutations in the SEPT12, RBMX, and ACTL7A genes cause deformed acrosome formation; mutations in the DNAH1, DNAH2, DNAH6, DNAH17, FSIP2, CFAP43, AK7, CHAP251, CFAP65, ARMC2 and several other genes result in multiple morphological abnormalities of sperm flagella (MMAF). CONCLUSIONS Altogether, mutations in 31 genes have been reported to cause head defects and mutations in 62 genes are known to cause sperm tail defects.
Collapse
Affiliation(s)
- Manvi Arora
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Poonam Mehta
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shruti Sethi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - George Anifandis
- Department of Obstetrics and Gynaecology, School of Health Sciences, Faculty of Medicine, University of Thessaly, Larisa, Greece
| | - Mary Samara
- Department of Obstetrics and Gynaecology, School of Health Sciences, Faculty of Medicine, University of Thessaly, Larisa, Greece
| | - Rajender Singh
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
5
|
Sethi S, Mehta P, Andrabi W, Mitra K, Rajender S. SPEM1 Gene Mutation in a Case with Sperm Morphological Defects Leading to Male Infertility. Reprod Sci 2024; 31:3102-3111. [PMID: 38886283 DOI: 10.1007/s43032-024-01612-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/29/2024] [Indexed: 06/20/2024]
Abstract
The present study aimed at identifying the genetic mutation responsible for teratozoospermic infertility in a case with coiled sperm tails. A 33-year-old infertile male was diagnosed with teratozoospermic infertility, with sperm head in coiled (HIC) tail as the most common deformity. We employed whole exome sequencing to identify the genetic cause in this case. Exome sequencing data was filtered using the following criteria: MAF (< 0.003), ALFA project (< 0.001), 1000 Genomes (< 0.003), Granthem (> 50), Polyphen-2 (> 0.70), SIFT (< 0.03), and PhyloP (> = 0) scores. Shortlisted variants were looked in the in-house 29 exomes data available with us, and the variants that affected conserved amino acid residues or led to insertion/deletion or to protein-truncation with a Combined Annotation Dependent Depletion (CADD) score ≥ 10 were shortlisted. The variants thus populated were prioritized according to their roles in spermiogenesis. The study identified a heterozygous mutation c.826C > T (Arg276Trp) in the SPEM1 gene as a potential pathogenic variant that led to teratozoospermic infertility in the case under investigation. The mutation had a minor allele frequency of 0.00008176 in the gnomAd database and was absent in the Indian Genome Variations database. This is the first human study reporting a mutation in the SPEM1 gene as a cause of coiled sperm tails.
Collapse
Affiliation(s)
- Shruti Sethi
- Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Industrial Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Poonam Mehta
- Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Industrial Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | | | - Kalyan Mitra
- Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Industrial Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Singh Rajender
- Central Drug Research Institute, Lucknow, Uttar Pradesh, India.
- Academy of Scientific and Industrial Research (AcSIR), Ghaziabad, Uttar Pradesh, India.
| |
Collapse
|
6
|
Wang H, Iida-Norita R, Mashiko D, Pham AH, Miyata H, Ikawa M. Golgi associated RAB2 interactor protein family contributes to murine male fertility to various extents by assuring correct morphogenesis of sperm heads. PLoS Genet 2024; 20:e1011337. [PMID: 38935810 PMCID: PMC11236154 DOI: 10.1371/journal.pgen.1011337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 07/10/2024] [Accepted: 06/10/2024] [Indexed: 06/29/2024] Open
Abstract
Sperm heads contain not only the nucleus but also the acrosome which is a distinctive cap-like structure located anterior to the nucleus and is derived from the Golgi apparatus. The Golgi Associated RAB2 Interactors (GARINs; also known as FAM71) protein family shows predominant expression in the testis and all possess a RAB2-binding domain which confers binding affinity to RAB2, a small GTPase that is responsible for membrane transport and vesicle trafficking. Our previous study showed that GARIN1A and GARIN1B are important for acrosome biogenesis and that GARIN1B is indispensable for male fertility in mice. Here, we generated KO mice of other Garins, namely Garin2, Garin3, Garin4, Garin5a, and Garin5b (Garin2-5b). Using computer-assisted morphological analysis, we found that the loss of each Garin2-5b resulted in aberrant sperm head morphogenesis. While the fertilities of Garin2-/- and Garin4-/- males are normal, Garin5a-/- and Garin5b-/- males are subfertile, and Garin3-/- males are infertile. Further analysis revealed that Garin3-/- males exhibited abnormal acrosomal morphology, but not as severely as Garin1b-/- males; instead, the amounts of membrane proteins, particularly ADAM family proteins, decreased in Garin3 KO spermatozoa. Moreover, only Garin4 KO mice exhibit vacuoles in the sperm head. These results indicate that GARINs assure correct head morphogenesis and some members of the GARIN family function distinctively in male fertility.
Collapse
Affiliation(s)
- Haoting Wang
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Rie Iida-Norita
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Daisuke Mashiko
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Anh Hoang Pham
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Haruhiko Miyata
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Masahito Ikawa
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
7
|
Wang S, Zeng X, Liu S, Hoque SAM, Min L, Ding N, Zhu Z. Vibration Emissions Reduce Boar Sperm Quality via Disrupting Its Metabolism. BIOLOGY 2024; 13:370. [PMID: 38927250 PMCID: PMC11200616 DOI: 10.3390/biology13060370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/24/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024]
Abstract
Artificial insemination (AI) with liquid-preserved semen has recently become common in pig breeding. The semen doses are produced in a centralized manner at the boar stud and then subsequently distributed and transported to sow farms. However, vibration emissions during transportation by logistic vehicles may adversely affect the quality of boar sperm. Therefore, this study aimed to explore the impact of vibration-induced emissions on sperm quality and function under simulated transportation conditions. Each time, ejaculates from all 15 boars were collected and then pooled together to minimize individual variations, and the sample was split using an extender for dilution. Different rotational speeds (0 rpm, 80 rpm, 140 rpm, 200 rpm) were utilized to simulate varying intensities of vibration exposure using an orbital shaker, considering different transportation times (0 h, 3 h, and 6 h). Subsequently, evaluations were conducted regarding sperm motility, plasma membrane integrity, acrosome integrity, mitochondrial function, adenosine triphosphate (ATP) levels, mitochondrial reactive oxygen species (ROS) levels, pH, glycolytic pathway enzyme activities, and capacitation following exposure to vibration emissions. Both vibration time and intensity impact sperm motility, plasma membrane integrity, and acrosomal integrity. Vibration exposure significantly reduced sperm ATP levels, mitochondrial membrane potential, and the levels of mitochondria-encoded proteins (MT-ND1, MT-ND6) (p < 0.05). After vibration emission treatment, the pH value and mitochondrial ROS levels significantly increased (p < 0.05). Inhibition of sperm glycolysis was observed, with reduced activities of hexokinase (HK), pyruvate kinase (PK), and lactate dehydrogenase (LDH), along with decreased lactate levels (p < 0.05). Additionally, sperm tyrosine phosphorylation levels were significantly reduced by vibration emissions compared to the control group (p < 0.05). After the vibration emission treatment, the number of sperm bound to each square millimeter of oviduct explants decreased significantly compared to the control group (p < 0.05). Similarly, compared to the control group, using semen subjected to vibration stress for AI results in significantly reduced pregnancy rates, total born litter size, live-born litter size, and healthy born litter size (p < 0.05).
Collapse
Affiliation(s)
- Shanpeng Wang
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China (L.M.)
| | - Xuejun Zeng
- Fujian Aoxin Biotechnology Group Co., Ltd., Zhangzhou 363000, China;
- Ji’an Aobao Biotechnology Group Co., Ltd., Ji’an 343000, China
| | - Shenao Liu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China (L.M.)
| | - S. A. Masudul Hoque
- Department of Animal Breeding and Genetics, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh;
| | - Lingjiang Min
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China (L.M.)
| | - Nengshui Ding
- Fujian Aoxin Biotechnology Group Co., Ltd., Zhangzhou 363000, China;
- Ji’an Aobao Biotechnology Group Co., Ltd., Ji’an 343000, China
- State Key Laboratory for Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Zhendong Zhu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China (L.M.)
| |
Collapse
|
8
|
Cheng GP, Wang YF, Li YY, Guo SM, Li HG, Ji DM, Yi NH, Zhou LQ. Deficiency of nucleosome-destabilizing factor GLYR1 dampens spermatogenesis in mice. Mol Cell Endocrinol 2024; 586:112194. [PMID: 38395189 DOI: 10.1016/j.mce.2024.112194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/13/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
Aberrant sperm morphology hinders sperm motility and causes male subfertility. Spermatogenesis, a complex process in male germ cell development, necessitates precise regulation of numerous developmental genes. However, the regulatory pathways involved in this process remain partially understood. We have observed the widespread expression of Glyr1, the gene encoding a nucleosome-destabilizing factor, in mouse testicular cells. Our study demonstrates that mice experiencing Glyr1 depletion in spermatogenic cells exhibit subfertility characterized by a diminished count and motility of spermatozoa. Furthermore, the rate of sperm malformation significantly increases in the absence of Glyr1, with a predominant occurrence of head and neck malformation in spermatozoa within the cauda epididymis. Additionally, a reduction in spermatocyte numbers across different meiotic stages is observed, accompanied by diminished histone acetylation in spermatogenic cells upon Glyr1 depletion. Our findings underscore the crucial roles of Glyr1 in mouse spermiogenesis and unveil novel insights into the etiology of male reproductive diseases.
Collapse
Affiliation(s)
- Gui-Ping Cheng
- Department of Women Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Fan Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan-Yuan Li
- Department of Gynecology and Obstetrics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi-Meng Guo
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Gang Li
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong-Mei Ji
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Anhui, China.
| | - Nian-Hua Yi
- Department of Women Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Li-Quan Zhou
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Anhui, China.
| |
Collapse
|
9
|
Graziani A, Rocca MS, Vinanzi C, Masi G, Grande G, De Toni L, Ferlin A. Genetic Causes of Qualitative Sperm Defects: A Narrative Review of Clinical Evidence. Genes (Basel) 2024; 15:600. [PMID: 38790229 PMCID: PMC11120687 DOI: 10.3390/genes15050600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Several genes are implicated in spermatogenesis and fertility regulation, and these genes are presently being analysed in clinical practice due to their involvement in male factor infertility (MFI). However, there are still few genetic analyses that are currently recommended for use in clinical practice. In this manuscript, we reviewed the genetic causes of qualitative sperm defects. We distinguished between alterations causing reduced sperm motility (asthenozoospermia) and alterations causing changes in the typical morphology of sperm (teratozoospermia). In detail, the genetic causes of reduced sperm motility may be found in the alteration of genes associated with sperm mitochondrial DNA, mitochondrial proteins, ion transport and channels, and flagellar proteins. On the other hand, the genetic causes of changes in typical sperm morphology are related to conditions with a strong genetic basis, such as macrozoospermia, globozoospermia, and acephalic spermatozoa syndrome. We tried to distinguish alterations approved for routine clinical application from those still unsupported by adequate clinical studies. The most important aspect of the study was related to the correct identification of subjects to be tested and the correct application of genetic tests based on clear clinical data. The correct application of available genetic tests in a scenario where reduced sperm motility and changes in sperm morphology have been observed enables the delivery of a defined diagnosis and plays an important role in clinical decision-making. Finally, clarifying the genetic causes of MFI might, in future, contribute to reducing the proportion of so-called idiopathic MFI, which might indeed be defined as a subtype of MFI whose cause has not yet been revealed.
Collapse
Affiliation(s)
- Andrea Graziani
- Department of Medicine, University of Padova, 35128 Padova, Italy; (A.G.); (G.M.); (L.D.T.)
| | - Maria Santa Rocca
- Unit of Andrology and Reproductive Medicine, University Hospital of Padova, 35128 Padova, Italy; (M.S.R.); (C.V.); (G.G.)
| | - Cinzia Vinanzi
- Unit of Andrology and Reproductive Medicine, University Hospital of Padova, 35128 Padova, Italy; (M.S.R.); (C.V.); (G.G.)
| | - Giulia Masi
- Department of Medicine, University of Padova, 35128 Padova, Italy; (A.G.); (G.M.); (L.D.T.)
| | - Giuseppe Grande
- Unit of Andrology and Reproductive Medicine, University Hospital of Padova, 35128 Padova, Italy; (M.S.R.); (C.V.); (G.G.)
| | - Luca De Toni
- Department of Medicine, University of Padova, 35128 Padova, Italy; (A.G.); (G.M.); (L.D.T.)
| | - Alberto Ferlin
- Department of Medicine, University of Padova, 35128 Padova, Italy; (A.G.); (G.M.); (L.D.T.)
- Unit of Andrology and Reproductive Medicine, University Hospital of Padova, 35128 Padova, Italy; (M.S.R.); (C.V.); (G.G.)
| |
Collapse
|
10
|
Zhou H, Zhang Z, Qu R, Zhu H, Luo Y, Li Q, Mu J, Yu R, Zeng Y, Chen B, Sang Q, Wang L. CCDC28A deficiency causes sperm head defects, reduced sperm motility and male infertility in mice. Cell Mol Life Sci 2024; 81:174. [PMID: 38597936 PMCID: PMC11006775 DOI: 10.1007/s00018-024-05184-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 04/11/2024]
Abstract
Mature spermatozoa with normal morphology and motility are essential for male reproduction. The epididymis has an important role in the proper maturation and function of spermatozoa for fertilization. However, factors related to the processes involved in spermatozoa modifications are still unclear. Here we demonstrated that CCDC28A, a member of the CCDC family proteins, is highly expressed in testes and the CCDC28A deletion leads to male infertility. We found CCDC28A deletion had a mild effect on spermatogenesis. And epididymal sperm collected from Ccdc28a-/- mice showed bent sperm heads, acrosomal defects, reduced motility and decreased in vitro fertilization competence whereas their axoneme, outer dense fibers, and fibrous sheath were all normal. Furthermore, we found that CCDC28A interacted with sperm acrosome membrane-associated protein 1 (SPACA1) and glycogen synthase kinase 3a (GSK3A), and deficiencies in both proteins in mice led to bent heads and abnormal acrosomes, respectively. Altogether, our results reveal the essential role of CCDC28A in regulating sperm morphology and motility and suggesting a potential marker for male infertility.
Collapse
Affiliation(s)
- Hongbin Zhou
- Institute of Pediatrics, The Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital of Fudan University, Fudan University, Shanghai, 200032, China
| | - Zhihua Zhang
- Institute of Pediatrics, The Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital of Fudan University, Fudan University, Shanghai, 200032, China
| | - Ronggui Qu
- Institute of Pediatrics, The Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital of Fudan University, Fudan University, Shanghai, 200032, China
| | - Hongying Zhu
- Institute of Pediatrics, The Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital of Fudan University, Fudan University, Shanghai, 200032, China
| | - Yuxi Luo
- Institute of Pediatrics, The Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital of Fudan University, Fudan University, Shanghai, 200032, China
| | - Qun Li
- Institute of Pediatrics, The Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital of Fudan University, Fudan University, Shanghai, 200032, China
| | - Jian Mu
- Institute of Pediatrics, The Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital of Fudan University, Fudan University, Shanghai, 200032, China
| | - Ran Yu
- Institute of Pediatrics, The Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital of Fudan University, Fudan University, Shanghai, 200032, China
| | - Yang Zeng
- Institute of Pediatrics, The Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital of Fudan University, Fudan University, Shanghai, 200032, China
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, 200032, China
| | - Qing Sang
- Institute of Pediatrics, The Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital of Fudan University, Fudan University, Shanghai, 200032, China.
| | - Lei Wang
- Institute of Pediatrics, The Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital of Fudan University, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
11
|
Sayegh L, Shah R, Shmoury M, Depret-Bixio L, Fakih M. Sperm morphology by strict criteria does not predict clinical pregnancy rate following intra-uterine insemination. Arab J Urol 2024; 22:195-205. [PMID: 39355794 PMCID: PMC11441082 DOI: 10.1080/20905998.2024.2327194] [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: 11/29/2023] [Accepted: 03/03/2024] [Indexed: 10/03/2024] Open
Abstract
Objective To determine the impact of abnormal sperm morphology of the pre-washed semen sample on the day of intrauterine insemination (IUI) on clinical pregnancy rates (CPR). Design Cross sectional retrospective chart review. Setting Academic fertility center. Patients Couples undergoing (IUI) from May 2014 to March 2022. Interventions Sperm morphology, by strict criteria, on the pre-washed IUI sample. Main outcomes Measures To determine the association of sperm morphology with CPR. Results Semen analysis reports, including Kruger strict criteria for morphology from the pre-washed IUI sample, were reviewed for 1,059 cycles, comprising 825 total treated couples.Of the total 1,059 cycles,15.1% resulted in clinical pregnancy. When categorized by strict morphology ≥4% (normal morphology), (3%-2%) [mild-moderate teratozoospermia (TZS)], and ≤1% (severe TZS), the CPR was 16%, 13%, and 10%, respectively (p value 0.30). Early spontaneous miscarriage rate was 4% and when stratified by morphology ≥4% (3%-2%), and ≤1%, was 3%, 1%, and 0%, respectively (p value 0.20).In couples with isolated TZS, the pregnancy rate was 16% in the normal morphology group, 14% in the mild-moderate group, and 8% in the severe group. (p value 0.30).In the multivariate logistic regression, sperm morphology, mild/moderate TZS vs normal forms (OR = 0.99, 95% CI [0.94-1.1]), severe TZS vs normal forms (OR = 0.98, 95% CI [0.0.83-1.1]), was not a predictor of CPR. The Pre-wash TMSC (OR = 1.0, 95% CI [0.996-1.00]) was also not predictive of CPR.The only predictive factor of CPR in IUI was the PWTMSC (OR = 1.03, 95%CI [1.00-1.06). Conclusions The morphology of the pre-washed sample on the day of IUI did not find a difference in CPR, neither in miscarriage rate following IUI, in couples with normal or abnormal sperm morphology, including severe TZS.Mild, moderate, or severe TZS in the semen sample should not exclude couples to attempt an IUI procedure.
Collapse
Affiliation(s)
- Lamia Sayegh
- Department of Infertility, Fakih IVF Fertility Center, Dubai, United Arab Emirates
| | - Rupin Shah
- Department of Infertility, Fakih IVF Fertility Center, Dubai, United Arab Emirates
| | - Mohamad Shmoury
- Department of Infertility, Fakih IVF Fertility Center, Dubai, United Arab Emirates
| | - Leyla Depret-Bixio
- Department of Infertility, Fakih IVF Fertility Center, Dubai, United Arab Emirates
| | - Michael Fakih
- Department of Infertility, Fakih IVF Fertility Center, Dubai, United Arab Emirates
| |
Collapse
|
12
|
Gajabe G, More A, Shrivastava J, Choudhary N, Dutta S, Kadu KS, Jadhav R. Application of Intracytoplasmic Morphologically Selected Sperm Injection (IMSI) on a Teratozoospermic Patient and Its Effect on the In-Vitro Fertilization (IVF) Outcome. Cureus 2024; 16:e53268. [PMID: 38435865 PMCID: PMC10905051 DOI: 10.7759/cureus.53268] [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: 01/15/2024] [Accepted: 01/30/2024] [Indexed: 03/05/2024] Open
Abstract
Infertility, defined as the inability to conceive after 12 months of unprotected sexual activity, affects millions globally. Approximately 80% of cases have identifiable causes, including endometriosis, tubal obstruction, ovulatory dysfunction, and male sperm abnormalities. Lifestyle factors, such as smoking and obesity, also impact fertility. Sperm morphology, a key factor in male infertility, often presents as teratozoospermia, with defects in the head, midpiece, or tail. Poor ovarian reserve, indicated by low anti-mullerine hormone (AMH) and antra-follicular count (AFC) values, contributes to female infertility, often exacerbated by age-related factors. Elevated follicle-stimulating hormone (FSH) levels further diminish oocyte quantity and quality. Intracytoplasmic Sperm Injection (ICSI), a micromanipulation technique aiding infertile couples, may face challenges in detecting subtle sperm morphology defects. Advanced methods like Motile Sperm Organelle Morphological Examination (MSOME) and Intracytoplasmic Morphologically Selected Sperm Injection (IMSI) under high magnification enhance sperm selection accuracy. We present the case of a 36-year-old woman and her 42-year-old husband who sought assistance after seven years of infertility. Previous Intrauterine injection (IUI) and ICSI attempts failed due to the wife's low ovarian reserve and elevated FSH, compounded by the husband's teratozoospermia. Their earlier In-Vitro Fertilization (IVF) experience yielded a single poor-quality oocyte, hindering blastocyst formation. Investigations revealed the wife's poor AFC, AMH of 0.033ng/ml, and FSH at 24IU/L. Her medical history included hypertension and gallbladder removal. The husband exhibited 98% defective sperm, devoid of a substance abuse history. The wife's family had a polycystic ovarian syndrome (PCOS) history, and her low AMH and AFC yielded only three poor-quality oocytes during the current assessment. Oocytes were retrieved, and sperm were selected with the help of IMSI. After ICSI, the patient successfully conceived.
Collapse
Affiliation(s)
- Gauri Gajabe
- Clinical Embryology, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Akash More
- Clinical Embryology, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Jarul Shrivastava
- Clinical Embryology, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Namrata Choudhary
- Clinical Embryology, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Shilpa Dutta
- Clinical Embryology, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Krushnali S Kadu
- Clinical Embryology, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Ritesh Jadhav
- Clinical Embryology, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| |
Collapse
|
13
|
Samare-Najaf M, Neisy A, Samareh A, Moghadam D, Jamali N, Zarei R, Zal F. The constructive and destructive impact of autophagy on both genders' reproducibility, a comprehensive review. Autophagy 2023; 19:3033-3061. [PMID: 37505071 PMCID: PMC10621263 DOI: 10.1080/15548627.2023.2238577] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 07/08/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
Reproduction is characterized by a series of massive renovations at molecular, cellular, and tissue levels. Recent studies have strongly tended to reveal the involvement of basic molecular pathways such as autophagy, a highly conserved eukaryotic cellular recycling, during reproductive processes. This review comprehensively describes the current knowledge, updated to September 2022, of autophagy contribution during reproductive processes in males including spermatogenesis, sperm motility and viability, and male sex hormones and females including germ cells and oocytes viability, ovulation, implantation, fertilization, and female sex hormones. Furthermore, the consequences of disruption in autophagic flux on the reproductive disorders including oligospermia, azoospermia, asthenozoospermia, teratozoospermia, globozoospermia, premature ovarian insufficiency, polycystic ovarian syndrome, endometriosis, and other disorders related to infertility are discussed as well.Abbreviations: AKT/protein kinase B: AKT serine/threonine kinase; AMPK: AMP-activated protein kinase; ATG: autophagy related; E2: estrogen; EDs: endocrine disruptors; ER: endoplasmic reticulum; FSH: follicle stimulating hormone; FOX: forkhead box; GCs: granulosa cells; HIF: hypoxia inducible factor; IVF: in vitro fertilization; IVM: in vitro maturation; LCs: Leydig cells; LDs: lipid droplets; LH: luteinizing hormone; LRWD1: leucine rich repeats and WD repeat domain containing 1; MAP1LC3: microtubule associated protein 1 light chain 3; MAPK: mitogen-activated protein kinase; MTOR: mechanistic target of rapamycin kinase; NFKB/NF-kB: nuclear factor kappa B; P4: progesterone; PCOS: polycystic ovarian syndrome; PDLIM1: PDZ and LIM domain 1; PI3K: phosphoinositide 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate; PtdIns3K: class III phosphatidylinositol 3-kinase; POI: premature ovarian insufficiency; ROS: reactive oxygen species; SCs: Sertoli cells; SQSTM1/p62: sequestosome 1; TSGA10: testis specific 10; TST: testosterone; VCP: vasolin containing protein.
Collapse
Affiliation(s)
- Mohammad Samare-Najaf
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Kerman Regional Blood Transfusion Center, Kerman, Iran
| | - Asma Neisy
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Samareh
- Department of Biochemistry, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Delaram Moghadam
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Navid Jamali
- Department of Laboratory Sciences, Sirjan School of Medical Sciences, Sirjan, Iran
| | - Reza Zarei
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Zal
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
14
|
Salehi N, Totonchi M. The construction of a testis transcriptional cell atlas from embryo to adult reveals various somatic cells and their molecular roles. J Transl Med 2023; 21:859. [PMID: 38012716 PMCID: PMC10680190 DOI: 10.1186/s12967-023-04722-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND The testis is a complex organ that undergoes extensive developmental changes from the embryonic stage to adulthood. The development of germ cells, which give rise to spermatozoa, is tightly regulated by the surrounding somatic cells. METHODS To better understand the dynamics of these changes, we constructed a transcriptional cell atlas of the testis, integrating single-cell RNA sequencing data from over 26,000 cells across five developmental stages: fetal germ cells, infants, childhood, peri-puberty, and adults. We employed various analytical techniques, including clustering, cell type assignments, identification of differentially expressed genes, pseudotime analysis, weighted gene co-expression network analysis, and evaluation of paracrine cell-cell communication, to comprehensively analyze this transcriptional cell atlas of the testis. RESULTS Our analysis revealed remarkable heterogeneity in both somatic and germ cell populations, with the highest diversity observed in Sertoli and Myoid somatic cells, as well as in spermatogonia, spermatocyte, and spermatid germ cells. We also identified key somatic cell genes, including RPL39, RPL10, RPL13A, FTH1, RPS2, and RPL18A, which were highly influential in the weighted gene co-expression network of the testis transcriptional cell atlas and have been previously implicated in male infertility. Additionally, our analysis of paracrine cell-cell communication supported specific ligand-receptor interactions involved in neuroactive, cAMP, and estrogen signaling pathways, which support the crucial role of somatic cells in regulating germ cell development. CONCLUSIONS Overall, our transcriptional atlas provides a comprehensive view of the cell-to-cell heterogeneity in the testis and identifies key somatic cell genes and pathways that play a central role in male fertility across developmental stages.
Collapse
Affiliation(s)
- Najmeh Salehi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.
| | - Mehdi Totonchi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
15
|
Redouane S, Charoute H, Harmak H, Malki A, Barakat A, Rouba H. Computational study of the potential impact of AURKC missense SNPs on AURKC-INCENP interaction and their correlation to macrozoospermia. J Biomol Struct Dyn 2023; 41:9503-9522. [PMID: 36326488 DOI: 10.1080/07391102.2022.2142846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
Aurora Kinase C (AURKC) is considered an important element in Chromosome Passenger Complex (CPC), its interaction with Inner Centromere Protein (INCENP) plays a critical role in the establishment and the recruitment of a stable CPC during spermatogenesis. Genetic variations of AURKC gene are susceptible to impact AURKC-INCENP interaction, which may affect CPC stability and predispose male subjects to macrozoospermia. In this study, we systematically applied computational approaches using different bioinformatic tools to predict the effect of missense SNPs reported on AURKC gene, we selected the deleterious ones and we introduced their corresponding amino acid substitutions on AURKC protein structure. Then we did a protein-protein docking between AURKC variants and INCENP followed by a structural assessment of each resulting complex using PRODIGY server, Yassara view, Ligplot + and we choose the complexes of the most impactful variants for molecular dynamics (MD) simulation study. Seventeen missense SNPs of AURKC were identified as deleterious between all reported ones. All of them were located on relatively conserved positions on AURKC protein according to Consurf server. Only the four missense SNPs; E91K, D166V, D221Y and G235V were ranked as the most impactful ones and were chosen for MD simulation. D221Y and G235V were responsible for the most remarkable changes on AURKC-INCENP structural stability, therefore, they were selected as the most deleterious ones. Experimental studies are recommended to test the actual effect of these two variants and their actual impact on the morphology of sperm cells.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Salaheddine Redouane
- Laboratory of Genomics and Human Genetics, Institut Pasteur du Maroc, Casablanca, Morocco
- Laboratory of Physiopathology and Molecular Genetics, Department of Biology, Faculty of Sciences Ben M'Sik, Hassan II University, Casablanca, Morocco
| | - Hicham Charoute
- Research Unit of Epidemiology, Biostatistics and Bioinformatics, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Houda Harmak
- Laboratory of Genomics and Human Genetics, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Abderrahim Malki
- Laboratory of Physiopathology and Molecular Genetics, Department of Biology, Faculty of Sciences Ben M'Sik, Hassan II University, Casablanca, Morocco
| | - Abdelhamid Barakat
- Laboratory of Genomics and Human Genetics, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Hassan Rouba
- Laboratory of Genomics and Human Genetics, Institut Pasteur du Maroc, Casablanca, Morocco
| |
Collapse
|
16
|
Andone BA, Handrea-Dragan IM, Botiz I, Boca S. State-of-the-art and future perspectives in infertility diagnosis: Conventional versus nanotechnology-based assays. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 54:102709. [PMID: 37717928 DOI: 10.1016/j.nano.2023.102709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/27/2023] [Accepted: 09/07/2023] [Indexed: 09/19/2023]
Abstract
According to the latest World Health Organization statistics, around 50 to 80 million people worldwide suffer from infertility, amongst which male factors are responsible for around 20 to 30 % of all infertility cases while 50 % were attributed to the female ones. As it is becoming a recurrent health problem worldwide, clinicians require more accurate methods for the improvement of both diagnosis and treatment schemes. By emphasizing the potential use of innovative methods for the rapid identification of the infertility causes, this review presents the news from this dynamic domain and highlights the benefits brought by emerging research fields. A systematic description of the standard techniques used in clinical protocols for diagnosing infertility in both genders is firstly provided, followed by the presentation of more accurate and comprehensive nanotechnology-related analysis methods such as nanoscopic-resolution imaging, biosensing approaches and assays that employ nanomaterials in their design. Consequently, the implementation of nanotechnology related tools in clinical practice, as recently demonstrated in the selection of spermatozoa, the detection of key proteins in the fertilization process or the testing of DNA integrity or the evaluation of oocyte quality, might confer excellent advantages both for improving the assessment of infertility, and for the success of the fertilization process.
Collapse
Affiliation(s)
- Bianca-Astrid Andone
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, 42 T. Laurian Str., 400271 Cluj-Napoca, Romania; Faculty of Physics, Babes-Bolyai University, 1 M. Kogalniceanu Str., 400084 Cluj-Napoca, Romania
| | - Iuliana M Handrea-Dragan
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, 42 T. Laurian Str., 400271 Cluj-Napoca, Romania; Faculty of Physics, Babes-Bolyai University, 1 M. Kogalniceanu Str., 400084 Cluj-Napoca, Romania
| | - Ioan Botiz
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, 42 T. Laurian Str., 400271 Cluj-Napoca, Romania
| | - Sanda Boca
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, 42 T. Laurian Str., 400271 Cluj-Napoca, Romania; National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat Str., 400293 Cluj-Napoca, Romania.
| |
Collapse
|
17
|
Kyrgiafini MA, Giannoulis T, Chatziparasidou A, Christoforidis N, Mamuris Z. Unveiling the Genetic Complexity of Teratozoospermia: Integrated Genomic Analysis Reveals Novel Insights into lncRNAs' Role in Male Infertility. Int J Mol Sci 2023; 24:15002. [PMID: 37834450 PMCID: PMC10573971 DOI: 10.3390/ijms241915002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023] Open
Abstract
Male infertility is a global health issue, affecting over 20 million men worldwide. Genetic factors are crucial in various male infertility forms, including teratozoospermia. Nonetheless, the genetic causes of male infertility remain largely unexplored. In this study, we employed whole-genome sequencing and RNA expression analysis to detect differentially expressed (DE) long-noncoding RNAs (lncRNAs) in teratozoospermia, along with mutations that are exclusive to teratozoospermic individuals within these DE lncRNAs regions. Bioinformatic tools were used to assess variants' impact on lncRNA structure, function, and lncRNA-miRNA interactions. Our analysis identified 1166 unique mutations in teratozoospermic men within DE lncRNAs, distinguishing them from normozoospermic men. Among these, 64 variants in 23 lncRNAs showed potential regulatory roles, 7 variants affected 4 lncRNA structures, while 37 variants in 17 lncRNAs caused miRNA target loss or gain. Pathway Enrichment and Gene Ontology analyses of the genes targeted by the affected miRNAs revealed dysregulated pathways in teratozoospermia and a link between male infertility and cancer. This study lists novel variants and lncRNAs associated for the first time with teratozoospermia. These findings pave the way for future studies aiming to enhance diagnosis and therapy in the field of male infertility.
Collapse
Affiliation(s)
- Maria-Anna Kyrgiafini
- Laboratory of Genetics, Comparative and Evolutionary Biology, Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, Mezourlo, 41500 Larissa, Greece
| | - Themistoklis Giannoulis
- Laboratory of Biology, Genetics and Bioinformatics, Department of Animal Sciences, University of Thessaly, Gaiopolis, 41336 Larissa, Greece
| | - Alexia Chatziparasidou
- Embryolab IVF Unit, St. 173-175 Ethnikis Antistaseos, Kalamaria, 55134 Thessaloniki, Greece
| | | | - Zissis Mamuris
- Laboratory of Genetics, Comparative and Evolutionary Biology, Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, Mezourlo, 41500 Larissa, Greece
| |
Collapse
|
18
|
Adriansyah RF, Margiana R, Supardi S, Narulita P. Current Progress in Stem Cell Therapy for Male Infertility. Stem Cell Rev Rep 2023; 19:2073-2093. [PMID: 37440145 DOI: 10.1007/s12015-023-10577-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2023] [Indexed: 07/14/2023]
Abstract
Infertility has become one of the most common issues worldwide, which has negatively affected society and infertile couples. Meanwhile, male infertility is responsible for about 50% of infertility. Accordingly, a great number of researchers have focused on its treatment during the last few years; however, current therapies such as assisted reproductive technology (ART) are not effective enough in treating male infertility. Because of their self-renewal and differentiation capabilities and unlimited sources, stem cells have recently raised great hope in the treatment of reproductive system disorders. Stem cells are undifferentiated cells that can induce different numbers of specific cells, such as male and female gametes, demonstrating their potential application in the treatment of infertility. The present review aimed at identifying the causes and potential factors that influence male fertility. Besides, we highlighted the recent studies that investigated the efficiency of stem cells such as spermatogonial stem cells (SSCs), embryonic stem cells (ESCs), very small embryonic-like stem cells (VSELs), induced pluripotent stem cells (iPSCs), and mesenchymal stem cells (MSCs) in the treatment of various types of male infertility.
Collapse
Affiliation(s)
| | - Ria Margiana
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
- Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
- Indonesia General Academic Hospital, Depok, Indonesia.
- Ciptomangunkusumo General Academic Hospital, Jakarta, Indonesia.
| | - Supardi Supardi
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Pety Narulita
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| |
Collapse
|
19
|
Wang Y, Su M, Chen Y, Huang X, Ruan L, Lv Q, Li L. Research progress on the role and mechanism of DNA damage repair in germ cell development. Front Endocrinol (Lausanne) 2023; 14:1234280. [PMID: 37529603 PMCID: PMC10390305 DOI: 10.3389/fendo.2023.1234280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 06/28/2023] [Indexed: 08/03/2023] Open
Abstract
In the complex and dynamic processes of replication, transcription, and translation of DNA molecules, a large number of replication errors or damage can occur which lead to obstacles in the development process of germ cells and result in a decreased reproductive rate. DNA damage repair has attracted widespread attention due to its important role in the maintenance and regulation of germ cells. This study reports on a systematic review of the role and mechanism of DNA damage repair in germline development. First, the causes, detection methods, and repair methods of DNA damage, and the mechanism of DNA damage repair are summarized. Second, a summary of the causes of abnormal DNA damage repair in germ cells is introduced along with common examples, and the relevant effects of germ cell damage. Third, we introduce the application of drugs related to DNA damage repair in the treatment of reproductive diseases and related surgical treatment of abnormal DNA damage, and summarize various applications of DNA damage repair in germ cells. Finally, a summary and discussion is given of the current deficiencies in DNA damage repair during germ cell development and future research development. The purpose of this paper is to provide researchers engaged in relevant fields with a further systematic understanding of the relevant applications of DNA damage repair in germ cells and to gain inspiration from it to provide new research ideas for related fields.
Collapse
Affiliation(s)
- Yan Wang
- College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
- College of Biology & Pharmacy, Yulin Normal University, Yulin, China
| | - Mengrong Su
- College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
- College of Biology & Pharmacy, Yulin Normal University, Yulin, China
| | - Yujie Chen
- College of Biology & Pharmacy, Yulin Normal University, Yulin, China
| | - Xinyu Huang
- College of Biology & Pharmacy, Yulin Normal University, Yulin, China
| | - Lian Ruan
- College of Biology & Pharmacy, Yulin Normal University, Yulin, China
| | - Qizhuang Lv
- College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
- College of Biology & Pharmacy, Yulin Normal University, Yulin, China
| | - Li Li
- College of Biology & Pharmacy, Yulin Normal University, Yulin, China
| |
Collapse
|
20
|
Abstract
In recent years, the incidence of teratospermia has been increasing, and it has become a very important factor leading to male infertility. The research on the molecular mechanism of teratospermia is also progressing rapidly. This article briefly summarizes the clinical incidence of teratozoospermia, and makes a retrospective summary of related studies reported in recent years. Specifically discussing the relationship between gene status and spermatozoa, the review aims to provide the basis for the genetic diagnosis and gene therapy of teratozoospermia.
Collapse
|
21
|
Fattahi M, Maghsudlu M, Hasan Sheikhha M. Is sperm telomere length altered in teratozoospermia specimens? A case-control study. Int J Reprod Biomed 2023; 21:229-236. [PMID: 37122888 PMCID: PMC10133734 DOI: 10.18502/ijrm.v21i3.13198] [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: 03/08/2022] [Revised: 07/19/2022] [Accepted: 02/02/2023] [Indexed: 05/02/2023] Open
Abstract
Background Male factor infertility is a multifactorial defect, and many of its etiologies are unknown. Teratozoospermia is determined by the existence of over 85% morphologically abnormal spermatozoa in semen which are almost incompetent in fertilization function. One of the most novel issues in genetic alterations studies is the variation of sperm telomere lengths (STL) and its collaboration with male infertility. The present study has been focused on STL alterations in teratozoospermia. Objective Investigation of differences in telomere length of teratozoospermia specimens and sperms with normal parameters. Materials and Methods In this case-control study, 60 men referred to Arak Fertility Clinic, Markazi province, Iran from November 2017 to February 2018 were categorized into teratozoospermia and normozoospermic groups. Sperm genomic DNA extraction was conducted, and STL were evaluated using quantitative polymerase chain reaction. Results Statistical evaluation of relative telomere length was calculated by the ratio of telomere to single-copy gene for teratozoospermia and normal specimens. Results significantly demonstrated that relative telomere length in teratozoospermia samples is nearly 3 times shorter than in normal samples (p > 0.001). Conclusion Our results represent the reduction of telomeres length in teratozoospermia and suggest that this alteration might be one of the factors contributing to the sperm fertility potential of this kind of specimen. However, defining relevant molecular processes requires further detailed investigations.
Collapse
Affiliation(s)
- Maryam Fattahi
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohadese Maghsudlu
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hasan Sheikhha
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
22
|
Shah R, Agarwal A, Kavoussi P, Rambhatla A, Saleh R, Cannarella R, Harraz AM, Boitrelle F, Kuroda S, Hamoda TAAAM, Zini A, Ko E, Calik G, Toprak T, Kandil H, Gül M, Bakırcıoğlu ME, Parekh N, Russo GI, Tadros N, Kadioglu A, Arafa M, Chung E, Rajmil O, Dimitriadis F, Malhotra V, Salvio G, Henkel R, Le TV, Sogutdelen E, Vij S, Alarbid A, Gudeloglu A, Tsujimura A, Calogero AE, El Meliegy A, Crafa A, Kalkanli A, Baser A, Hazir B, Giulioni C, Cho CL, Ho CCK, Salzano C, Zylbersztejn DS, Tien DMB, Pescatori E, Borges E, Serefoglu EC, Saïs-Hamza E, Huyghe E, Ceyhan E, Caroppo E, Castiglioni F, Bahar F, Gokalp F, Lombardo F, Gadda F, Duarsa GWK, Pinggera GM, Busetto GM, Balercia G, Cito G, Blecher G, Franco G, Liguori G, Elbardisi H, Keskin H, Lin H, Taniguchi H, Park HJ, Ziouziou I, de la Rosette J, Hotaling J, Ramsay J, Molina JMC, Lo KL, Bocu K, Khalafalla K, Bowa K, Okada K, Nagao K, Chiba K, Hakim L, Makarounis K, Hehemann M, Rodriguez Peña M, Falcone M, Bendayan M, Martinez M, Timpano M, Altan M, Fode M, Al-Marhoon MS, Sadighi Gilani MA, Soebadi MA, Gherabi N, Sofikitis N, Kahraman O, Birowo P, et alShah R, Agarwal A, Kavoussi P, Rambhatla A, Saleh R, Cannarella R, Harraz AM, Boitrelle F, Kuroda S, Hamoda TAAAM, Zini A, Ko E, Calik G, Toprak T, Kandil H, Gül M, Bakırcıoğlu ME, Parekh N, Russo GI, Tadros N, Kadioglu A, Arafa M, Chung E, Rajmil O, Dimitriadis F, Malhotra V, Salvio G, Henkel R, Le TV, Sogutdelen E, Vij S, Alarbid A, Gudeloglu A, Tsujimura A, Calogero AE, El Meliegy A, Crafa A, Kalkanli A, Baser A, Hazir B, Giulioni C, Cho CL, Ho CCK, Salzano C, Zylbersztejn DS, Tien DMB, Pescatori E, Borges E, Serefoglu EC, Saïs-Hamza E, Huyghe E, Ceyhan E, Caroppo E, Castiglioni F, Bahar F, Gokalp F, Lombardo F, Gadda F, Duarsa GWK, Pinggera GM, Busetto GM, Balercia G, Cito G, Blecher G, Franco G, Liguori G, Elbardisi H, Keskin H, Lin H, Taniguchi H, Park HJ, Ziouziou I, de la Rosette J, Hotaling J, Ramsay J, Molina JMC, Lo KL, Bocu K, Khalafalla K, Bowa K, Okada K, Nagao K, Chiba K, Hakim L, Makarounis K, Hehemann M, Rodriguez Peña M, Falcone M, Bendayan M, Martinez M, Timpano M, Altan M, Fode M, Al-Marhoon MS, Sadighi Gilani MA, Soebadi MA, Gherabi N, Sofikitis N, Kahraman O, Birowo P, Kothari P, Sindhwani P, Javed Q, Ambar RF, Kosgi R, Ghayda RA, Adriansjah R, Condorelli RA, La Vignera S, Micic S, Kim SHK, Fukuhara S, Ahn ST, Mostafa T, Ong TA, Takeshima T, Amano T, Barrett T, Arslan U, Karthikeyan VS, Atmoko W, Yumura Y, Yuan Y, Kato Y, Jezek D, Cheng BKC, Hatzichristodoulou G, Dy J, Castañé ER, El-Sakka AI, Nguyen Q, Sarikaya S, Boeri L, Tan R, Moussa MA, El-Assmy A, Alali H, Alhathal N, Osman Y, Perovic D, Sajadi H, Akhavizadegan H, Vučinić M, Kattan S, Kattan MS, Mogharabian N, Phuoc NHV, Ngoo KS, Alkandari MH, Alsuhaibani S, Sokolakis I, Babaei M, King MS, Diemer T, Gava MM, Henrique R, Silva RSE, Paul GM, Mierzwa TC, Glina S, Siddiqi K, Wu H, Wurzacher J, Farkouh A, Son H, Minhas S, Lee J, Magsanoc N, Capogrosso P, Albano GJ, Lewis SEM, Jayasena CN, Alvarez JG, Teo C, Smith RP, Chua JBM, Jensen CFS, Parekattil S, Finelli R, Durairajanayagam D, Karna KK, Ahmed A, Evenson D, Umemoto Y, Puigvert A, Çeker G, Colpi GM. Consensus and Diversity in the Management of Varicocele for Male Infertility: Results of a Global Practice Survey and Comparison with Guidelines and Recommendations. World J Mens Health 2023; 41:164-197. [PMID: 35791302 PMCID: PMC9826919 DOI: 10.5534/wjmh.220048] [Show More Authors] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/15/2022] [Accepted: 03/20/2022] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Varicocele is a common problem among infertile men. Varicocele repair (VR) is frequently performed to improve semen parameters and the chances of pregnancy. However, there is a lack of consensus about the diagnosis, indications for VR and its outcomes. The aim of this study was to explore global practice patterns on the management of varicocele in the context of male infertility. MATERIALS AND METHODS Sixty practicing urologists/andrologists from 23 countries contributed 382 multiple-choice-questions pertaining to varicocele management. These were condensed into an online questionnaire that was forwarded to clinicians involved in male infertility management through direct invitation. The results were analyzed for disagreement and agreement in practice patterns and, compared with the latest guidelines of international professional societies (American Urological Association [AUA], American Society for Reproductive Medicine [ASRM], and European Association of Urology [EAU]), and with evidence emerging from recent systematic reviews and meta-analyses. Additionally, an expert opinion on each topic was provided based on the consensus of 16 experts in the field. RESULTS The questionnaire was answered by 574 clinicians from 59 countries. The majority of respondents were urologists/uro-andrologists. A wide diversity of opinion was seen in every aspect of varicocele diagnosis, indications for repair, choice of technique, management of sub-clinical varicocele and the role of VR in azoospermia. A significant proportion of the responses were at odds with the recommendations of AUA, ASRM, and EAU. A large number of clinical situations were identified where no guidelines are available. CONCLUSIONS This study is the largest global survey performed to date on the clinical management of varicocele for male infertility. It demonstrates: 1) a wide disagreement in the approach to varicocele management, 2) large gaps in the clinical practice guidelines from professional societies, and 3) the need for further studies on several aspects of varicocele management in infertile men.
Collapse
Affiliation(s)
- Rupin Shah
- Division of Andrology, Department of Urology, Lilavati Hospital and Research Centre, Mumbai, India
| | - Ashok Agarwal
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA.
| | - Parviz Kavoussi
- Austin Fertility & Reproductive Medicine/Westlake IVF, TX, USA
| | - Amarnath Rambhatla
- Department of Urology, Vattikuti Urology Institute, Henry Ford Health System, Detroit, MI, USA
| | - Ramadan Saleh
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Ahmed M Harraz
- Mansoura University Urology and Nephrology Center, Mansoura, Egypt
- Department of Surgery, Urology Unit, Farwaniya Hospital, Farwaniya, Kuwait
- Department of Urology, Sabah Al Ahmad Urology Center, Kuwait City, Kuwait
| | - Florence Boitrelle
- Department of Reproductive Biology, Fertility Preservation, Andrology, CECOS, Poissy Hospital, Poissy, France
- Department of Biology, Reproduction, Epigenetics, Environment and Development, Paris Saclay University, UVSQ, INRAE, BREED, Jouy-en-Josas, France
| | - Shinnosuke Kuroda
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Taha Abo-Almagd Abdel-Meguid Hamoda
- Department of Urology, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Urology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Armand Zini
- Division of Urology, Department of Surgery, McGill University, Montreal, QC, Canada
| | - Edmund Ko
- Department of Urology, Loma Linda University Health, Loma Linda, CA, USA
| | - Gokhan Calik
- Department of Urology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Tuncay Toprak
- Department of Urology, Fatih Sultan Mehmet Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | | | - Murat Gül
- Department of Urology, Selcuk University School of Medicine, Konya, Turkey
| | | | - Neel Parekh
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Nicholas Tadros
- Division of Urology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Ates Kadioglu
- Section of Andrology, Department of Urology, Istanbul University, Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Mohamed Arafa
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
- Department of Urology, Hamad Medical Corporation, Doha, Qatar
- Department of Urology, Weill Cornell Medical-Qatar, Doha, Qatar
| | - Eric Chung
- Department of Urology, Princess Alexandra Hospital, University of Queensland, Brisbane, Australia
| | - Osvaldo Rajmil
- Department of Andrology, Fundacio Puigvert, Barcelona, Spain
| | - Fotios Dimitriadis
- Department of Urology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Gianmaria Salvio
- Department of Endocrinology, Polytechnic University of Marche, Ancona, Italy
| | - Ralf Henkel
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Medical Bioscience, University of the Western Cape, Bellville, South Africa
| | - Tan V Le
- Department of Andrology, Binh Dan Hospital, Ho Chi Minh City, Vietnam
- Department of Urology and Andrology, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam
| | | | - Sarah Vij
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Abdullah Alarbid
- Department of Surgery, Urology Unit, Farwaniya Hospital, Farwaniya, Kuwait
| | - Ahmet Gudeloglu
- Department of Urology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Akira Tsujimura
- Department of Urology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Amr El Meliegy
- Department of Andrology, Sexology and STIs, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Arif Kalkanli
- Department of Urology, Taksim Education and Research Hospital, Istanbul, Turkey
| | - Aykut Baser
- Department of Urology, Faculty of Medicine, Bandırma Onyedi Eylül University, Balıkesir, Turkey
| | - Berk Hazir
- Reproductive medicine, Clinical Research Centre, Lund University, Malmö, Sweden
| | - Carlo Giulioni
- Department of Urology, Polytechnic University of Marche Region, Ancona, Italy
| | - Chak-Lam Cho
- S. H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Christopher C K Ho
- Department of Urology, Universiti Teknologi Mara (UiTM) Specialist Centre, Selangor, Malaysia
| | - Ciro Salzano
- PO San Giovanni Bosco, ASL Napoli 1 Centro, Napoli, Italy
| | | | - Dung Mai Ba Tien
- Department of Andrology, Binh Dan Hospital, Ho Chi Minh City, Vietnam
| | - Edoardo Pescatori
- Andrology and Reproductive Medicine Unit, Gynepro Medical, Bologna, Italy
| | | | - Ege Can Serefoglu
- Department of Urology, Biruni University School of Medicine, Istanbul, Turkey
| | - Emine Saïs-Hamza
- Department of Reproductive Biology, Fertility Preservation, Andrology, CECOS, Poissy Hospital, Poissy, France
- Department of Biology, Reproduction, Epigenetics, Environment and Development, Paris Saclay University, UVSQ, INRAE, BREED, Jouy-en-Josas, France
| | - Eric Huyghe
- Department of Urology and Andrology, University Hospital of Toulouse, France
| | - Erman Ceyhan
- Department of Urology, School of Medicine, Baskent University, Konya, Turkey
| | - Ettore Caroppo
- Asl Bari, PTA "F Jaia", Reproductive and IVF unit, Andrology Outpatients Clinic, Asl Bari, Conversano (Ba), Italy
| | | | - Fahmi Bahar
- Andrology Section, Siloam Sriwijaya Hospital, Palembang, Indonesia
| | - Fatih Gokalp
- Department of Urology, Hatay Mustafa Kemal University, Antakya, Turkey
| | - Francesco Lombardo
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, University of Rome "Sapienza", Rome, Italy
| | - Franco Gadda
- Department of Urology, IRCCS Fondazione Ca Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | | | - Gian Maria Busetto
- Department of Urology and Organ Transplantation, University of Foggia, Ospedali Riuniti of Foggia, Foggia, Italy
| | - Giancarlo Balercia
- Department of Endocrinology, Polytechnic University of Marche, Ancona, Italy
| | - Gianmartin Cito
- Department of Urology, University of Florence, Florence, Italy
| | - Gideon Blecher
- Department of Surgery, School of Clinical Sciences, Monash University, Melbourne, Australia
- Department of Urology, The Alfred Hospital, Melbourne, Australia
| | - Giorgio Franco
- Department of Urology, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | | | - Haitham Elbardisi
- Department of Urology, Hamad Medical Corporation, Doha, Qatar
- Department of Urology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Hakan Keskin
- Department of Urology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Haocheng Lin
- Department of Urology, Peking University Third Hospital, Peking University, Beijing, China
| | - Hisanori Taniguchi
- Department of Urology and Andrology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Hyun Jun Park
- Department of Urology, Pusan National University School of Medicine, Busan, Korea
- Medical Research Institute of Pusan National University Hospital, Busan, Korea
| | - Imad Ziouziou
- Department of Urology, College of medicine and pharmacy, Ibn Zohr University, Agadir, Morocco
| | - Jean de la Rosette
- Department of urology, Faculty of Medicine, Istanbul Medipol Mega University Hospital, Istanbul, Turkey
| | - Jim Hotaling
- Department of Surgery (Urology), University of Utah, Salt Lake City, UT, USA
| | - Jonathan Ramsay
- Department of Andrology, Hammersmith Hospital, Imperial, London, UK
| | | | - Ka Lun Lo
- Division of Urology, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Kadir Bocu
- Department of Urology, Silopi State Hospital, Sirnak, Turkey
| | - Kareim Khalafalla
- Department of Urology, Hamad Medical Corporation, Doha, Qatar
- Department of Urology, University of Illinois, Chicago, IL, USA
| | - Kasonde Bowa
- Department of Urology, School of Medicine and Health Sciences, University of Lusaka, Lusaka, Zambia
| | - Keisuke Okada
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Koichi Nagao
- Department of Urology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Koji Chiba
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Lukman Hakim
- Department of Urology, Universitas Airlangga/Rumah Sakit Universitas Airlangga Teaching Hospital, Surabaya, Indonesia
| | | | - Marah Hehemann
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Marcelo Rodriguez Peña
- Instituto de Ginecología y Fertilidad (IFER), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marco Falcone
- Department of Urology, Molinette Hospital, A.O.U. Città della Salute e della Scienza, University of Turin, Torino, Italy
| | - Marion Bendayan
- Department of Reproductive Biology, Fertility Preservation, Andrology, CECOS, Poissy Hospital, Poissy, France
- Department of Biology, Reproduction, Epigenetics, Environment and Development, Paris Saclay University, UVSQ, INRAE, BREED, Jouy-en-Josas, France
| | - Marlon Martinez
- Section of Urology, Department of Surgery, University of Santo Tomas Hospital, Manila, Philippines
| | - Massimiliano Timpano
- Department of Urology, Molinette Hospital, A.O.U. Città della Salute e della Scienza, University of Turin, Torino, Italy
| | - Mesut Altan
- Department of Urology, Haceppete University, Ankara, Turkey
| | - Mikkel Fode
- Department of Urology, Herlev and Gentofte University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Mohammad Ali Sadighi Gilani
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | | | - Nazim Gherabi
- Andrology Committee of the Algerian Association of Urology, Algiers, Algeria
| | - Nikolaos Sofikitis
- Department of Urology, Ioannina University School of Medicine, Ioannina, Greece
| | - Oğuzhan Kahraman
- Department of Urology, School of Medicine, Baskent University, Konya, Turkey
| | - Ponco Birowo
- Department of Urology, Cipto Mangunkusumo General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | | | - Puneet Sindhwani
- Department of Urology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Qaisar Javed
- Department of Urology, Al - Ahlia Hospital Abu Dhabi UAE, Abu Dhabi, UAE
| | - Rafael F Ambar
- Department of Urology, Centro Universitario em Saude do ABC, Santo André, Brazil
- Andrology Group at Ideia Fertil Institute of Human Reproduction, Santo André, Brazil
| | - Raghavender Kosgi
- Department of Urology and Andrology, AIG Hospitals, Hyderabad, Telangana, India
| | - Ramy Abou Ghayda
- Urology Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, USA
| | - Ricky Adriansjah
- Department of Urology, Faculty of Medicine Universitas Padjadjaran, Hasan Sadikin General Hospital, Banding, Indonesia
| | | | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sava Micic
- Department of Andrology, Uromedica Polyclinic, Belgrade, Serbia
| | - Shannon Hee Kyung Kim
- IVF Australia, Sydney, New South Wales, Australia
- Macquarie School of Medicine, Macquaire University, Sydney, New South Wales, Australia
| | - Shinichiro Fukuhara
- Department of Urology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Sun Tae Ahn
- Department of Urology, Korea University College of Medicine, Seoul, Korea
| | - Taymour Mostafa
- Department of Andrology, Sexology and STIs, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Teng Aik Ong
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Teppei Takeshima
- Department of Urology, Reproduction Center, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Toshiyasu Amano
- Department of Urology, Nagano Red Cross Hospital, Nagano, Japan
| | | | - Umut Arslan
- Department of Urology, Fatih Sultan Mehmet Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | | | - Widi Atmoko
- Department of Urology and Andrology, AIG Hospitals, Hyderabad, Telangana, India
| | - Yasushi Yumura
- Department of Urology, Reproduction Center, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Yiming Yuan
- Andrology Center & Urology Department, Peking University First Hospital, Peking University, Beijing, China
| | - Yuki Kato
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Davor Jezek
- Department for Transfusion Medicine and Transplantation Biology, Reproductive Tissue Bank, University of Zagreb, School of Medicine, University Hospital Zagreb, Zagreb, Croatia
| | | | | | - Jun Dy
- Stone and Prostate Treatment Services/Pelvic Floor Center and Anorectal Diseases, St. Luke's Medical Center, Quezon City, Metro Manila, Philippines
| | - Eduard Ruiz Castañé
- Department of Andrology, Fundació Puigvert, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ahmed I El-Sakka
- Department of Urology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Quang Nguyen
- Center for Andrology and Sexual Medicine, Viet Duc University Hospital, Hanoi, Vietnam
- Department of Urology, Andrology and Sexual Medicine, University of Medicine and Pharmacy, Vietnam National University, Hanoi, Vietnam
| | - Selcuk Sarikaya
- Department of Urology, Gülhane Research and Training Hospital, University of Health Sciences, Ankara, Turkey
| | - Luca Boeri
- Department of Urology, IRCCS Fondazione Ca Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ronny Tan
- Advanced Urology Associates, Singapore
- Mount Elizabeth Hospital, Singapore
| | - Mohamad A Moussa
- Department of Urology, Lebanese University, Beirut, Lebanon
- Department of Urology, Al Zahraa Hospital, UMC, Lebanon
| | - Ahmed El-Assmy
- Mansoura University Urology and Nephrology Center, Mansoura, Egypt
| | - Hamed Alali
- King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Naif Alhathal
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Yasser Osman
- Mansoura University Urology and Nephrology Center, Mansoura, Egypt
| | - Dragoljub Perovic
- Urology and Andrology Center, CODRA Hospital, University of Montenegro, Podgorica, Montenegro
| | | | - Hamed Akhavizadegan
- Department of Urology, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Said Kattan
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mohamed S Kattan
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Nasser Mogharabian
- Sexual Health and Fertility Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | | | - Kay Seong Ngoo
- Urology Unit, Department of Surgery, Hospital Angkatan Tentera Tuanku Mizan, Kuala Lumpur, Malaysia
| | - Mohammad H Alkandari
- Department of Urology, Mubarak Al-Kabeer Teaching Hospital, Kuwait University, Jabriya, Kuwait
| | - Shaheed Alsuhaibani
- Department of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Ioannis Sokolakis
- Department of Urology, Martha-Maria Hospital Nuremberg, Nuremberg, Germany
| | - Mehdi Babaei
- Department of Andrology, Shariati Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mak Siu King
- Division of Urology, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Thorsten Diemer
- Department of Urology, Pediatric Urology and Andrology, Justus-Liebig-University Giessen (GER), University Hospital Giessen and Marburg GmbH, Campus Giessen, Giessen, Germany
| | - Marcelo M Gava
- Department of Urology, Centro Universitario em Saude do ABC, Santo André, Brazil
- Andrology Group at Ideia Fertil Institute of Human Reproduction, Santo André, Brazil
| | | | - Rodrigo Spinola E Silva
- Department of Urology, Centro Universitario em Saude do ABC, Santo André, Brazil
- Andrology Group at Ideia Fertil Institute of Human Reproduction, Santo André, Brazil
| | - Gustavo Marquesine Paul
- Department of Urology, Hospital de Clínicas of the Federal University of Paraná, Curitiba, Brazil
| | | | - Sidney Glina
- Department of Urology, Centro Universitario em Saude do ABC, Santo André, Brazil
| | | | - Han Wu
- Department of Andrology, PKU 3rd Hospital Reproductive Medicine Center, Beijing, China
| | - Jana Wurzacher
- Department of Urology, University Hospital Innsbruck, Innsbruck, Austria
| | - Ala'a Farkouh
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Hwancheol Son
- Department of Urology, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Suks Minhas
- Division of Surgery, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Joe Lee
- Department of Urology, National University Hospital, Singapore
| | - Nikko Magsanoc
- Department of Surgery, University of the Philippines College of Medicine, Manila, Philippines
| | | | - German Jose Albano
- Section of Urology, Department of Surgery, University of Santo Tomas Hospital, Manila, Philippines
| | | | - Channa N Jayasena
- Department of Reproductive Endocrinology and Andrology, Imperial College London, London, UK
- Department of Andrology, Hammersmith & St. Mary's Hospitals, London, UK
| | - Juan G Alvarez
- Department of Andrology, Centro ANDROGEN, La Coruña, Spain
| | - Colin Teo
- Department of Urology, Gleneagles Hospital, Singapore
| | - Ryan P Smith
- Department of Urology, University of Virginia School of Medicine, Virginia, USA
| | - Jo Ben M Chua
- Department of Urology, East Avenue Medical Center, Quezon City, Philippines
| | | | - Sijo Parekattil
- Avant Concierge Urology & University of Central Florida, Winter Garden, FL, USA
| | - Renata Finelli
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Damayanthi Durairajanayagam
- Department of Physiology, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Keshab Kumar Karna
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Abdelkareem Ahmed
- Department of Urology, Sabah Al Ahmad Urology Center, Kuwait City, Kuwait
| | | | - Yukihiro Umemoto
- Department of Nephro-Urology, Nagoya City West Medical Center, Nagoya, Japan
| | - Ana Puigvert
- Urological and Human Reproduction Unit, Quiron Hospital, Barcelona, Spain
| | - Gökhan Çeker
- Department of Urology, Başakşehir Çam and Sakura City Hospital, Istanbul, Turkey
- Department of Embryology and Histology, Zonguldak Bülent Ecevit University Institute of Health Sciences, Zonguldak, Turkey
| | | |
Collapse
|
23
|
Huang S, Huang Y, Li S, He Y. Chromosome 17 translocation affects sperm morphology: Two case studies and literature review. Andrologia 2022; 54:e14620. [PMID: 36270636 DOI: 10.1111/and.14620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/14/2022] [Accepted: 09/28/2022] [Indexed: 11/30/2022] Open
Abstract
We present two cases of infertile males with teratozoospermia stemming from chromosome 17 translocation. The patients present karyotypes that have not been previously reported. Genes located on breakpoints (17p11.2, 9q31, and 11p15) were analysed to find the probable mechanism affecting sperm morphology. Our results suggest that ALKBH5, TOP3A, and LLGL1 interactions may be an underlying cause of abnormal sperm head morphology. Translocation of chromosome 17 occurred in conjunction with chromosome 9 and chromosome 11 translocation in the two cases, resulting in oligozoospermia and asthenozoospermia, respectively. These abnormal phenotypes may involve meiosis- and motility-related genes such as LDHC, DNHD1, UBQLN3, and NUP98. Translocation is thus a risk factor for sperm morphological abnormalities and motility deficiency. The interaction network of 22 genes on breakpoints suggests that they contribute to spermatogenesis as a group. In conclusion, this study highlighted the importance of investigating genes linked to sperm morphology, together with chromosome 17 translocation and reproductive risks. For patients interested in screening before a future pregnancy, we recommend preimplantation genetic diagnosis to reduce the risk of karyotypically unbalanced foetuses and birth defects.
Collapse
Affiliation(s)
- Shan Huang
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yingting Huang
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shan Li
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yu He
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
24
|
Whole-Genome Profile of Greek Patients with Teratozοοspermia: Identification of Candidate Variants and Genes. Genes (Basel) 2022; 13:genes13091606. [PMID: 36140773 PMCID: PMC9498395 DOI: 10.3390/genes13091606] [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: 07/26/2022] [Revised: 08/23/2022] [Accepted: 08/30/2022] [Indexed: 01/09/2023] Open
Abstract
Male infertility is a global health problem that affects a large number of couples worldwide. It can be categorized into specific subtypes, including teratozoospermia. The present study aimed to identify new variants associated with teratozoospermia in the Greek population and to explore the role of genes on which these were identified. For this reason, whole-genome sequencing (WGS) was performed on normozoospermic and teratozoospermic individuals, and after selecting only variants found in teratozoospermic men, these were further prioritized using a wide range of tools, functional and predictive algorithms, etc. An average of 600,000 variants were identified, and of them, 61 were characterized as high impact and 153 as moderate impact. Many of these are mapped in genes previously associated with male infertility, yet others are related for the first time to teratozoospermia. Furthermore, pathway enrichment analysis and Gene ontology (GO) analyses revealed the important role of the extracellular matrix in teratozoospermia. Therefore, the present study confirms the contribution of genes studied in the past to male infertility and sheds light on new molecular mechanisms by providing a list of variants and candidate genes associated with teratozoospermia in the Greek population.
Collapse
|
25
|
Moretti E, Signorini C, Noto D, Corsaro R, Collodel G. The relevance of sperm morphology in male infertility. FRONTIERS IN REPRODUCTIVE HEALTH 2022; 4:945351. [PMID: 36303645 PMCID: PMC9580829 DOI: 10.3389/frph.2022.945351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/11/2022] [Indexed: 11/23/2022] Open
Abstract
This brief report concerns the role of human sperm morphology assessment in different fields of male infertility: basic research, genetics, assisted reproduction technologies, oxidative stress. One of the best methods in studying sperm morphology is transmission electron microscopy (TEM) that enables defining the concept of sperm pathology and classifying alterations in non-systematic and systematic. Non-systematic sperm defects affect head and tail in variable ratio, whereas the rare systematic defects are characterized by a particular anomaly that marks most sperm of an ejaculate. TEM analysis and fluorescence in situ hybridization represent outstanding methods in the study of sperm morphology and cytogenetic in patients with altered karyotype characterizing their semen quality before intracytoplasmic sperm injection. In recent years, the genetic investigations on systematic sperm defects, made extraordinary progress identifying candidate genes whose mutations induce morphological sperm anomalies. The question if sperm morphology has an impact on assisted fertilization outcome is debated. Nowadays, oxidative stress represents one of the most important causes of altered sperm morphology and function and can be analyzed from two points of view: 1) spermatozoa with cytoplasmic residue produce reactive oxygen species, 2) the pathologies with inflammatory/oxidative stress background cause morphological alterations. Finally, sperm morphology is also considered an important endpoint in in vitro experiments where toxic substances, drugs, antioxidants are tested. We think that the field of sperm morphology is far from being exhausted and needs other research. This parameter can be still considered a valuable indicator of sperm dysfunction both in basic and clinical research.
Collapse
|
26
|
Delessard M, Stalin L, Rives-Feraille A, Moutard L, Saulnier J, Dumont L, Rives N, Rondanino C. Achievement of complete in vitro spermatogenesis in testicular tissues from prepubertal mice exposed to mono- or polychemotherapy. Sci Rep 2022; 12:7407. [PMID: 35523907 PMCID: PMC9076692 DOI: 10.1038/s41598-022-11286-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/18/2022] [Indexed: 02/08/2023] Open
Abstract
The assessment of the impact of chemotherapies on in vitro spermatogenesis in experimental models is required before considering the application of this fertility restoration strategy to prepubertal boys who received these treatments before testicular tissue cryopreservation. The present work investigated the effects of exposure of prepubertal mice to mono- (vincristine or cyclophosphamide) and polychemotherapy (a combination of vincristine and cyclophosphamide) on the first wave of in vitro spermatogenesis. When testicular tissue exposed to monochemotherapy was preserved, polychemotherapy led to severe alterations of the seminiferous epithelium and increased apoptosis in prepubertal testes prior in vitro maturation, suggesting a potential additive gonadotoxic effect. These alterations were also found in the testicular tissues of polychemotherapy-treated mice after 30 days of organotypic culture and were associated with a reduction in the germ cell/Sertoli cell ratio. The different treatments neither altered the ability of spermatogonia to differentiate in vitro into spermatozoa nor the yield of in vitro spermatogenesis. However, more spermatozoa with morphological abnormalities and fragmented DNA were produced after administration of polychemotherapy. This work therefore shows for the first time the possibility to achieve a complete in vitro spermatogenesis after an in vivo exposure of mice to a mono- or polychemotherapy before meiotic entry.
Collapse
Affiliation(s)
- Marion Delessard
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Laura Stalin
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Aurélie Rives-Feraille
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Laura Moutard
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Justine Saulnier
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Ludovic Dumont
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Nathalie Rives
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Christine Rondanino
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France.
| |
Collapse
|
27
|
Sáez-Espinosa P, Robles-Gómez L, Ortega-López L, Aizpurua J, Gómez-Torres MJ. Immunofluorescence and High-Resolution Microscopy Reveal New Insights in Human Globozoospermia. Int J Mol Sci 2022; 23:ijms23031729. [PMID: 35163651 PMCID: PMC8836125 DOI: 10.3390/ijms23031729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/25/2022] [Accepted: 01/31/2022] [Indexed: 12/23/2022] Open
Abstract
Globozoospermia is a rare and severe type of teratozoospermia characterized by the presence of round-headed, acrosomeless spermatozoa with cytoskeleton defects. Current data support a negative relationship between globozoospermia and intracytoplasmic sperm injection (ICSI) outcomes, revealing the need to perform exhaustive studies on this type of sperm disorder. The aim of this study was to evaluate different structural, functional and molecular sperm biomarkers in total globozoospermia with proper embryo development after ICSI. The combination of field-emission scanning electron microscopy (FE-SEM) and transmission electron microscopy (TEM) allowed us to identify and correlate eight morphological patterns with both types of microscopy. Additionally, results reported a high percentage of coiled forms, with cytoplasmic retentions around the head and midpiece. By fluorescent microscopy, we detected that most of the sperm showed tubulin in the terminal piece of the flagellum and less than 1% displayed tyrosine phosphorylation in the flagellum. Moreover, we did not detect chaperone Heat shock-related 70 kDa protein 2 (HSPA2) in 85% of the cells. Overall, these findings provide new insights into globozoospermia, which could have potential implications in improving sperm selection methods for assisted reproductive techniques.
Collapse
Affiliation(s)
- Paula Sáez-Espinosa
- Departamento de Biotecnología, Facultad de Ciencias, Universidad de Alicante, 03690001 Alicante, Spain; (P.S.-E.); (L.R.-G.)
| | - Laura Robles-Gómez
- Departamento de Biotecnología, Facultad de Ciencias, Universidad de Alicante, 03690001 Alicante, Spain; (P.S.-E.); (L.R.-G.)
| | - Leonor Ortega-López
- IVF Spain, Reproductive Medicine, 03540001 Alicante, Spain; (L.O.-L.); (J.A.)
| | - Jon Aizpurua
- IVF Spain, Reproductive Medicine, 03540001 Alicante, Spain; (L.O.-L.); (J.A.)
| | - María José Gómez-Torres
- Departamento de Biotecnología, Facultad de Ciencias, Universidad de Alicante, 03690001 Alicante, Spain; (P.S.-E.); (L.R.-G.)
- Cátedra Human Fertility, Facultad de Ciencias, Universidad de Alicante, 03690001 Alicante, Spain
- Correspondence: ; Tel.:+34-965-903-878
| |
Collapse
|
28
|
Porumb IG, Coricovac AM, Raica II, Zărnescu O, Didilescu AC, Didilescu AC, Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Romania, Department of Embryology, Gynera Fertility Clinic, Bucharest, Romania; Department of Embryology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania, Department of Embryology, Gynera Fertility Clinic, Bucharest, Romania, Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Romania, Department of Embryology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania. Healthy birth in a case of total globozoospermia after intracytoplasmic sperm injection and assisted oocyte activation. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2022; 63:459-463. [PMID: 36374151 PMCID: PMC9801671 DOI: 10.47162/rjme.63.2.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Globozoospermia is a rare (incidence <0.1%) and very severe disorder, with major implications in male fertility. Total globozoospermia is represented by the presence of spermatozoa with 100% rounded heads and a lack of acrosomes. These specific morphological modifications seem to be connected to defects occurring in the last stage of spermatogenesis, spermiogenesis, and will result in anomalies of the acrosomal reaction and a defective adherence of the spermatozoa to the oocytes zona pellucida. This will result in a failure of natural fertilization. This article aims to present the case of a couple diagnosed and successfully treated for primary male infertility. The 26-year-old male partner underwent two semen analyses that revealed the presence of fully rounded spermatozoa heads (morphological abnormality) and consequently was proposed for in vitro fertilization treatment. Semen preparation and the use of assisted reproductive techniques, intracytoplasmic injection of sperm cells into the assisted oocyte activation, have resulted in the conceivement of a healthy child. The particularities of this case lie in the early recognition of the total abnormal globozoospermia morphology. This is the first case reported in Romania where specific assisted reproductive techniques and treatments have resulted in a successful pregnancy for a couple with male total globozoospermia.
Collapse
Affiliation(s)
- Ionuţ George Porumb
- Department of Embryology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; ; Department of Embryology, Gynera Fertility Clinic, Bucharest, Romania;
| | - Anca Magdalena Coricovac
- Department of Embryology, Gynera Fertility Clinic, Bucharest, Romania,Department of Embryology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Ioana Iulia Raica
- Department of Embryology, Gynera Fertility Clinic, Bucharest, Romania
| | - Otilia Zărnescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Romania
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Yeast Two-Hybrid Screen Identifies PKA-Riα Interacting Proteins during Mouse Spermiogenesis. Genes (Basel) 2021; 12:genes12121941. [PMID: 34946890 PMCID: PMC8700991 DOI: 10.3390/genes12121941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/26/2022] Open
Abstract
cAMP-dependent protein kinase (PKA) signaling plays various roles during mammalian spermatogenesis, ranging from the regulation of gene expression to the modulation of sperm motility. However, the molecular mechanisms that govern the multifaceted functions of PKA during spermatogenesis remain largely unclear. We previously found that PKA regulatory subunit I α (RIα) and catalytic subunit α (Cα) co-sediment with polyribosomal fractions of mouse testis lysate on sucrose gradient and the stimulation of PKA activity facilitates protein synthesis in post-meiotic elongating spermatids, indicating that type I PKA is intricately associated with protein translation machinery and regulates protein synthesis during mouse spermiogenesis. Since PKA activity is often regulated by interacting proteins that form complexes with its regulatory subunits, the identification of PKA-RIα interacting proteins in post-meiotic spermatogenic cells will facilitate our understanding of its regulatory roles in protein synthesis and spermiogenesis. In the present study, we applied a yeast two-hybrid screen to identify PKA-Riα-binding proteins using a cDNA library generated from mouse round and elongating spermatids. Numerous proteins were found to potentially interact with PKA-RIα, including proteostasis modulators, metabolic enzymes, cytoskeletal regulators, and mitochondrial proteins, many of which are specifically expressed in testes. Consistently, the examination of MENA (mouse ENA/VASP homolog) in developing mouse testes suggested that post-meiotic spermatogenic cells express a short isoform of MENA that interacts with PKA-RIα in yeast two-hybrid assay. The identification of PKA-RIα interacting proteins provides us solid basis to further explore how PKA signaling regulates protein synthesis and cellular morphogenesis during mouse spermatogenesis.
Collapse
|
30
|
Asadpour R, Mofidi Chelan E. Using microRNAs as molecular biomarkers for the evaluation of male infertility. Andrologia 2021; 54:e14298. [PMID: 34738652 DOI: 10.1111/and.14298] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 09/25/2021] [Accepted: 10/23/2021] [Indexed: 12/16/2022] Open
Abstract
Infertility is a multiplex disorder in the reproductive system, and men are responsible for more than half of the cases. Nowadays, semen analysis has been considered the critical assessment test to diagnose infertile men; however, it has limitations so that the cause behind infertility in 40% of infertile men is unrevealed. Weaknesses of semen assessment indicate a global need for novel and better diagnostic tools and biomarkers. MicroRNAs are short (about 18-22 nucleotide length) non-coding RNAs that control most (>60%) of our protein-coding genes post-transcriptionally. These molecules are aberrant in the body fluids, and abnormal alterations in their expression level can signify a specific disease such as infertility. Therefore, microRNAs can be novel candidate biomarkers that can diagnose different types of male infertility, including azoospermia, oligozoospermia, asthenozoospermia and teratozoospermia. This narrative review aimed to collect and sum up new papers published about the significant role of microRNAs in different male infertility categories.
Collapse
Affiliation(s)
- Reza Asadpour
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ehsan Mofidi Chelan
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
31
|
Abstract
Sperm selection in the female reproductive tract (FRT) is sophisticated. Only about 1,000 sperm out of millions in an ejaculate reach the fallopian tube and thus have a chance of fertilizing an oocyte. In assisted reproduction techniques, sperm are usually selected using their density or motility, characteristics that do not reflect their fertilization competence and, therefore, might result in failure to fertilize the oocyte. Although sperm processing in in vitro fertilization (IVF) and intrauterine insemination (IUI) bypasses many of the selection processes in the FRT, selection by the cumulus mass and the zona pellucida remain intact. By contrast, the direct injection of a sperm into an oocyte in intracytoplasmic sperm injection (ICSI) bypasses all natural selection barriers and, therefore, increases the risk of transferring paternal defects such as fragmented DNA and genomic abnormalities in sperm to the resulting child. Research into surrogate markers of fertilization potential and into simulating the natural sperm selection processes has progressed. However, methods of sperm isolation - such as hyaluronic acid-based selection and microfluidic isolation based on sperm tactic responses - use only one or two parameters and are not comparable with the multistep sperm selection processes naturally occurring within the FRT. Fertilization-competent sperm require a panel of molecules, including zona pellucida-binding proteins and ion channel proteins, that enable them to progress through the FRT to achieve fertilization. The optimal artificial sperm selection method will, therefore, probably need to use a multiparameter tool that incorporates the molecular signature of sperm with high fertilization potential, and their responses to external cues, within a microfluidic system that can replicate the physiological processes of the FRT in vitro.
Collapse
|
32
|
Integration and gene co-expression network analysis of scRNA-seq transcriptomes reveal heterogeneity and key functional genes in human spermatogenesis. Sci Rep 2021; 11:19089. [PMID: 34580317 PMCID: PMC8476490 DOI: 10.1038/s41598-021-98267-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/27/2021] [Indexed: 02/07/2023] Open
Abstract
Spermatogenesis is a complex process of cellular division and differentiation that begins with spermatogonia stem cells and leads to functional spermatozoa production. However, many of the molecular mechanisms underlying this process remain unclear. Single-cell RNA sequencing (scRNA-seq) is used to sequence the entire transcriptome at the single-cell level to assess cell-to-cell variability. In this study, more than 33,000 testicular cells from different scRNA-seq datasets with normal spermatogenesis were integrated to identify single-cell heterogeneity on a more comprehensive scale. Clustering, cell type assignments, differential expressed genes and pseudotime analysis characterized 5 spermatogonia, 4 spermatocyte, and 4 spermatid cell types during the spermatogenesis process. The UTF1 and ID4 genes were introduced as the most specific markers that can differentiate two undifferentiated spermatogonia stem cell sub-cellules. The C7orf61 and TNP can differentiate two round spermatid sub-cellules. The topological analysis of the weighted gene co-expression network along with the integrated scRNA-seq data revealed some bridge genes between spermatogenesis's main stages such as DNAJC5B, C1orf194, HSP90AB1, BST2, EEF1A1, CRISP2, PTMS, NFKBIA, CDKN3, and HLA-DRA. The importance of these key genes is confirmed by their role in male infertility in previous studies. It can be stated that, this integrated scRNA-seq of spermatogenic cells offers novel insights into cell-to-cell heterogeneity and suggests a list of key players with a pivotal role in male infertility from the fertile spermatogenesis datasets. These key functional genes can be introduced as candidates for filtering and prioritizing genotype-to-phenotype association in male infertility.
Collapse
|
33
|
Molecular Drivers of Developmental Arrest in the Human Preimplantation Embryo: A Systematic Review and Critical Analysis Leading to Mapping Future Research. Int J Mol Sci 2021; 22:ijms22158353. [PMID: 34361119 PMCID: PMC8347543 DOI: 10.3390/ijms22158353] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/28/2021] [Accepted: 07/31/2021] [Indexed: 12/14/2022] Open
Abstract
Developmental arrest of the preimplantation embryo is a multifactorial condition, characterized by lack of cellular division for at least 24 hours, hindering the in vitro fertilization cycle outcome. This systematic review aims to present the molecular drivers of developmental arrest, focusing on embryonic and parental factors. A systematic search in PubMed/Medline, Embase and Cochrane-Central-Database was performed in January 2021. A total of 76 studies were included. The identified embryonic factors associated with arrest included gene variations, mitochondrial DNA copy number, methylation patterns, chromosomal abnormalities, metabolic profile and morphological features. Parental factors included, gene variation, protein expression levels and infertility etiology. A valuable conclusion emerging through critical analysis indicated that genetic origins of developmental arrest analyzed from the perspective of parental infertility etiology and the embryo itself, share common ground. This is a unique and long-overdue contribution to literature that for the first time presents an all-inclusive methodological report on the molecular drivers leading to preimplantation embryos’ arrested development. The variety and heterogeneity of developmental arrest drivers, along with their inevitable intertwining relationships does not allow for prioritization on the factors playing a more definitive role in arrested development. This systematic review provides the basis for further research in the field.
Collapse
|
34
|
TSGA10 as a Potential Key Factor in the Process of Spermatid Differentiation/Maturation: Deciphering Its Association with Autophagy Pathway. Reprod Sci 2021; 28:3228-3240. [PMID: 34232471 DOI: 10.1007/s43032-021-00648-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 05/30/2021] [Indexed: 10/20/2022]
Abstract
Testis-specific gene antigen 10 (TSGA10) plays an important role in spermatogenesis. However, the exact TSGA10 role and its relationship with the autophagy pathway in the process of spermatids differentiation/maturation is still not clear. Therefore, the present study evaluates the role of TSGA10 gene in the spermatid differentiation/maturation through its effect on autophagy and explores possible underlying pathway(s). Sperm samples from patients with teratospermia were collected. The mRNA and protein level of TSGA10 in these samples were assessed by real-time PCR and western blotting. Using the ingenuity pathway analysis (IPA) software, the gene network and interactions of TSGA10 involved in sperm maturation and autophagy were investigated. Based on these analyses, the expression levels of identified genes in patient's samples and healthy controls were further evaluated. Moreover, using flow cytometry analysis, the levels of reactive oxygen species (ROS( production in teratospermic sperm samples were evaluated. The results showed that the expression levels of TSGA10 mRNA and protein decreased significantly in the teratospermic patients compared to controls (P < 0.05). Moreover, a significant reduction in the expression of the important genes involved in sperm maturation and autophagy was observed (P < 0.05). Also, the levels of ROS production in teratospermic sperm samples were shown to be significantly higher compared to those in normal sperms (P < 0.05). Our findings provide new evidence that simultaneous decrease in TSGA10 and autophagy beside the increased level of ROS production in sperm cells might be associated with the abnormalities in the spermatids differentiation/maturation and the formation of sperms with abnormal morphology.
Collapse
|
35
|
Faja F, Pallotti F, Cargnelutti F, Senofonte G, Carlini T, Lenzi A, Lombardo F, Paoli D. Molecular Analysis of DPY19L2, PICK1 and SPATA16 in Italian Unrelated Globozoospermic Men. Life (Basel) 2021; 11:life11070641. [PMID: 34209343 PMCID: PMC8307282 DOI: 10.3390/life11070641] [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: 05/17/2021] [Revised: 06/21/2021] [Accepted: 06/25/2021] [Indexed: 12/24/2022] Open
Abstract
This study aims to evaluate genetic contribution and sperm DNA fragmentation (SDF) in a cohort of 18 unrelated globozoospermic Italian men (Group G). Semen samples were assessed according to the WHO 2010 Laboratory Manual and compared with 31 fertile controls. We focused our genetic analysis on the exons of the main globozoospermia-associated genes, performing qualitative PCR to assess deletion of DPY19L2 and sequencing to detect mutations of SPATA16 and PICK1. SDF was evaluated using the TUNEL assay. In Group G, 10 patients had a complete form of globozoospermia, whereas 8 patients had a partial form. Molecular analysis revealed deletion of DPY19L2 in six of the patients, all of them with complete globozoospermia, while no mutations were found in the examined exons of PICK1 and SPATA16. TUNEL analysis showed a higher SDF% in Group G. Our findings confirm DPY19L2 defects as the most frequent genetic alteration in Italian patients contributing to globozoospermic phenotypes. Furthermore, spermatozoa with acrosomal defects could also display high levels of SDF as a possible consequence of abnormally remodeled chromatin. The possible effect on offspring of chromatin structure abnormalities and altered DNA integrity should be carefully evaluated by clinicians, especially regarding the feasibility and safety of artificial reproductive techniques, which represent the only treatment that allows these patients to conceive.
Collapse
|
36
|
Azhar M, Altaf S, Uddin I, Cheng J, Wu L, Tong X, Qin W, Bao J. Towards Post-Meiotic Sperm Production: Genetic Insight into Human Infertility from Mouse Models. Int J Biol Sci 2021; 17:2487-2503. [PMID: 34326689 PMCID: PMC8315030 DOI: 10.7150/ijbs.60384] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/16/2021] [Indexed: 02/06/2023] Open
Abstract
Declined quality and quantity of sperm is currently the major cause of patients suffering from infertility. Male germ cell development is spatiotemporally regulated throughout the whole developmental process. While it has been known that exogenous factors, such as environmental exposure, diet and lifestyle, et al, play causative roles in male infertility, recent progress has revealed abundant genetic mutations tightly associated with defective male germline development. In mammals, male germ cells undergo dramatic morphological change (i.e., nuclear condensation) and chromatin remodeling during post-meiotic haploid germline development, a process termed spermiogenesis; However, the molecular machinery players and functional mechanisms have yet to be identified. To date, accumulated evidence suggests that disruption in any step of haploid germline development is likely manifested as fertility issues with low sperm count, poor sperm motility, aberrant sperm morphology or combined. With the continually declined cost of next-generation sequencing and recent progress of CRISPR/Cas9 technology, growing studies have revealed a vast number of disease-causing genetic variants associated with spermiogenic defects in both mice and humans, along with mechanistic insights partially attained and validated through genetically engineered mouse models (GEMMs). In this review, we mainly summarize genes that are functional at post-meiotic stage. Identification and characterization of deleterious genetic variants should aid in our understanding of germline development, and thereby further improve the diagnosis and treatment of male infertility.
Collapse
Affiliation(s)
- Muhammad Azhar
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Saba Altaf
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Islam Uddin
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Jinbao Cheng
- The 901th hospital of Joint logistics support Force of PLA, Anhui, China
| | - Limin Wu
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Xianhong Tong
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Weibing Qin
- NHC Key Laboratory of Male Reproduction and Genetics, Family Planning Research Institute of Guangdong Province, China
| | - Jianqiang Bao
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| |
Collapse
|
37
|
Automatic Microscopy Analysis with Transfer Learning for Classification of Human Sperm. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11125369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Infertility is a global problem that affects many couples. Sperm analysis plays an essential role in the clinical diagnosis of human fertility. The examination of sperm morphology is an essential technique because sperm morphology is a proven indicator of biological functions. At present, the morphological classification of human sperm is conducted manually by medical experts. However, manual classification is laborious and highly dependent on the experience and capability of clinicians. To address these limitations, we propose a transfer learning method based on AlexNet to automatically classify the sperms into four different categories in terms of the World Health Organization (WHO) standards by analyzing their morphology. We adopt the feature extraction architecture of AlexNet as well as its pre-training parameters. Besides, we redesign the classification network by adding the Batch Normalization layers to improve the performance. The proposed method achieves an average accuracy of 96.0% and an average precision of 96.4% in the freely-available HuSHeM dataset, which exceeds the performance of previous algorithms. Our method shows that automatic sperm classification has great potential to replace manual sperm classification in the future.
Collapse
|
38
|
Candela L, Boeri L, Capogrosso P, Cazzaniga W, Pozzi E, Belladelli F, Baudo A, Ravizzoli A, Ventimiglia E, Viganò P, Alfano M, Abbate C, Cornelius J, Mattei A, Montorsi F, Salonia A. Correlation among isolated teratozoospermia, sperm DNA fragmentation and markers of systemic inflammation in primary infertile men. PLoS One 2021; 16:e0251608. [PMID: 34097690 PMCID: PMC8184012 DOI: 10.1371/journal.pone.0251608] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 04/28/2021] [Indexed: 01/03/2023] Open
Abstract
Aim To assess the prevalence of isolated teratozoospermia (iTZS) in a cohort of infertile and fertile men; explore the relationship between iTZS, inflammatory parameters and sperm DNA fragmentation index (SDF) in the same cohort. Materials and methods 1824 infertile men and 103 fertile controls. Semen analysis, the neutrophil-to-lymphocyte ratio (NLR) and serum hormones were investigated. DFI was tested in infertile men only. According to 2010 WHO semen analysis, patients were categorized in 3 sub-groups of isolated sperm defects: isolated oligozoospermia (iOZS), isolated asthenozoospermia (iAZS) and iTZS. Descriptive statistics and linear regression models tested the association between clinical variables and inflammatory markers. Results Among infertile men, iAZS, iTZS, and iOZS were found in 13.9%, 11.9% and 4.1% participants, respectively. iTZS was found in 37 (35.9%) fertile men. Infertile men with iTZS had higher NLR values than those with iOZS, iAZS and men with normal semen parameters (all p<0.001). FSH and LH were higher and inhibin B lower in iOZS infertile men compared to all other groups (p≤0.001). Hormonal characteristics were similar between iTZS infertile and fertile men. Similarly, iTZS infertile men had higher SDF than all other groups (all p<0.001). Infertile men with iTZS had higher NLR values than fertile men with iTZS (p<0.01). Linear regression analysis showed that, in infertile men, iTZS was associated with SDF and NLR (all p≤0.01). Conclusions iTZS was found in 11.9% of infertile men but it was even more prevalent in fertile controls. Infertile men with iTZS had higher NLR than fertile controls and increased SDF values than infertile participant with iAZS, iOZS, or normal semen parameters. No differences in hormonal characteristics were found between infertile and fertile men with iTZS.
Collapse
Affiliation(s)
- Luigi Candela
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Luca Boeri
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- Department of Urology, Foundation IRCCS Ca’ Granda–Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Paolo Capogrosso
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- Department of Urology and Andrology, Ospedale di Circolo and Macchi Foundation,Varese, Italy
| | - Walter Cazzaniga
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Edoardo Pozzi
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Federico Belladelli
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Baudo
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Ravizzoli
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Eugenio Ventimiglia
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Paola Viganò
- Obstetrics and Gynaecology Department, IRCCS Ospedale San Raffaele, Milan, Italy
- Division of Genetics and Cell Biology, Reproductive Sciences Laboratory, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Massimo Alfano
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Costantino Abbate
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Julian Cornelius
- Department of Urology, Luzerner Kantonsspital, Lucerne, Switzerland
| | - Agostino Mattei
- Department of Urology, Luzerner Kantonsspital, Lucerne, Switzerland
| | - Francesco Montorsi
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Salonia
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
- * E-mail:
| |
Collapse
|
39
|
Tripp JA, Berrio A, McGraw LA, Matz MV, Davis JK, Inoue K, Thomas JW, Young LJ, Phelps SM. Comparative neurotranscriptomics reveal widespread species differences associated with bonding. BMC Genomics 2021; 22:399. [PMID: 34058981 PMCID: PMC8165761 DOI: 10.1186/s12864-021-07720-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/20/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Pair bonding with a reproductive partner is rare among mammals but is an important feature of human social behavior. Decades of research on monogamous prairie voles (Microtus ochrogaster), along with comparative studies using the related non-bonding meadow vole (M. pennsylvanicus), have revealed many of the neural and molecular mechanisms necessary for pair-bond formation in that species. However, these studies have largely focused on just a few neuromodulatory systems. To test the hypothesis that neural gene expression differences underlie differential capacities to bond, we performed RNA-sequencing on tissue from three brain regions important for bonding and other social behaviors across bond-forming prairie voles and non-bonding meadow voles. We examined gene expression in the amygdala, hypothalamus, and combined ventral pallidum/nucleus accumbens in virgins and at three time points after mating to understand species differences in gene expression at baseline, in response to mating, and during bond formation. RESULTS We first identified species and brain region as the factors most strongly associated with gene expression in our samples. Next, we found gene categories related to cell structure, translation, and metabolism that differed in expression across species in virgins, as well as categories associated with cell structure, synaptic and neuroendocrine signaling, and transcription and translation that varied among the focal regions in our study. Additionally, we identified genes that were differentially expressed across species after mating in each of our regions of interest. These include genes involved in regulating transcription, neuron structure, and synaptic plasticity. Finally, we identified modules of co-regulated genes that were strongly correlated with brain region in both species, and modules that were correlated with post-mating time points in prairie voles but not meadow voles. CONCLUSIONS These results reinforce the importance of pre-mating differences that confer the ability to form pair bonds in prairie voles but not promiscuous species such as meadow voles. Gene ontology analysis supports the hypothesis that pair-bond formation involves transcriptional regulation, and changes in neuronal structure. Together, our results expand knowledge of the genes involved in the pair bonding process and open new avenues of research in the molecular mechanisms of bond formation.
Collapse
Affiliation(s)
- Joel A Tripp
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, 78712, USA
| | - Alejandro Berrio
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, 78712, USA
- Present Address: Department of Biology, Duke University, Durham, NC, 27708, USA
| | - Lisa A McGraw
- Center for Translational Social Neuroscience, Department of Psychiatry and Behavioral Sciences, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Mikhail V Matz
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, 78712, USA
| | - Jamie K Davis
- Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - Kiyoshi Inoue
- Center for Translational Social Neuroscience, Department of Psychiatry and Behavioral Sciences, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - James W Thomas
- National Institutes of Health Intramural Sequencing Center, National Human Genome Research Institute, National Institutes of Health, Rockville, MD, USA
| | - Larry J Young
- Center for Translational Social Neuroscience, Department of Psychiatry and Behavioral Sciences, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Steven M Phelps
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
40
|
Han B, Wang L, Yu S, Ge W, Li Y, Jiang H, Shen W, Sun Z. One potential biomarker for teratozoospermia identified by in-depth integrative analysis of multiple microarray data. Aging (Albany NY) 2021; 13:10208-10224. [PMID: 33819193 PMCID: PMC8064145 DOI: 10.18632/aging.202781] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/16/2021] [Indexed: 12/27/2022]
Abstract
Teratozoospermia is a common category of male infertility and with the increase in clinical patients and the increasing sophistication of assisted reproductive technology, there is an urgent need for an accurate semen diagnostic biomarker to accomplish rapid diagnosis of patients with teratozoospermia and accurately assess the success rate of assisted reproductive technologies. In this study, we performed gene differential expression analysis on two publicly available DNA microarray datasets (GSE6872 and GSE6967), followed by GSEA analysis to parse their enriched KEGG pathways, and WGCNA analysis to obtain the most highly correlated modules. Subsequent in-depth comparative analysis of the modules screened into the two datasets resulted in a gene set containing the identical expression trend, and then the differentially expressed genes in the set were screened using the corresponding criteria. Finally, three differentially expressed genes common to both datasets were selected. In addition, we validated the expression changes of this gene using another dataset (GSE6968) and in vitro experiments, and only screened one potential semen biomarker gene whose expression trend was identical to those in other datasets, which will also provide an important theoretical basis for the diagnosis and treatment of teratozoospermia.
Collapse
Affiliation(s)
- Baoquan Han
- Urology Department, Peking University Shenzhen Hospital, Shenzhen Peking University and The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Lu Wang
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Shuai Yu
- Urology Department, Peking University Shenzhen Hospital, Shenzhen Peking University and The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Wei Ge
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Yaqi Li
- Urology Department, Zaozhuang Hospital of Zaozhuang Mining Group, Zaozhuang 277100, China
| | - Hui Jiang
- Department of Urology, Department of Andrology, Department of Human Sperm Bank, Peking University Third Hospital, Beijing 100191, China
| | - Wei Shen
- Urology Department, Peking University Shenzhen Hospital, Shenzhen Peking University and The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China.,College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Zhongyi Sun
- Urology Department, Peking University Shenzhen Hospital, Shenzhen Peking University and The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| |
Collapse
|
41
|
Foroozan-Boroojeni S, Tavalaee M, Zakeri Z, Lockshin RA, Nasr-Esfahani MH. Assessment of Atg7 and LC3II/LC3, as The Markers of Autophagy, in Sperm of Infertile Men with Globozoospermia: A Case-Control Study. CELL JOURNAL 2021; 23:70-74. [PMID: 33650822 PMCID: PMC7944124 DOI: 10.22074/cellj.2021.7023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/11/2019] [Indexed: 12/01/2022]
Abstract
Objective Assessment of relationship between LC3II/LC3 and Autophagy-related 7 (Atg7) proteins, as markers of autophagy,
as well as evaluating the sperm parameters and DNA fragmentation in spermatozoa of infertile men with globozoospermia.
Materials and Methods In this case-control study, 10 semen samples from infertile men with globozoospermia and 10
fertile individuals were collected, and the sperm parameters, sperm DNA fragmentation, and main autophagy markers
(Atg7 and LC3II/LC3) were assessed according to World Health Organization (WHO) criteria, TUNEL assay, and
western blot technique, respectively.
Results The mean of sperm concentration and motility were significantly lower, while the percentage of abnormal
spermatozoa and DNA fragmentation were significantly higher in infertile men with globozoospermia compared to
fertile individuals (P<0.01). Unlike the relative expression of LC3II/LC3 that did not significantly differ between the two
groups, the relative expression of ATG7 was significantly higher in infertile men with globozoospermia compared to
fertile individuals (P<0.05). There was a significantly negative correlation between the sperm concentration (r=-0.679;
P=0.005) and motility (r=-0.64; P=0.01) with the expression of ATG7, while a significantly positive association was founf
between the percentage of DNA fragmentation and expression of ATG7 (0.841; P =0.018).
Conclusion The increased expression of ATG7 and unaltered expression of LC3II/LC3 may indicate that the
autophagy pathway is initiated but not completely executed in spermatozoa of individuals with globozoospermia. A
significant correlation of ATG7 expression with increased sperm DNA fragmentation, reduced sperm concentration, and
sperm motility may associate with the activation of a compensatory mechanism for promoting deficient spermatozoa to
undergo cell death by the autophagy pathway. Therfore, this pathway could act as a double-edged sword that, at the
physiological level, is involved in acrosome biogenesis, while, at the pathological level, such as globozoospermia, could
act as a compensatory mechanism.
Collapse
Affiliation(s)
- Shaghayegh Foroozan-Boroojeni
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Marziyeh Tavalaee
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Zahra Zakeri
- Department of Biology, Queens College and Graduate Center of The City University of New York, Flushing, NY, USA
| | - Richard A Lockshin
- Department of Biology, Queens College and Graduate Center of The City University of New York, Flushing, NY, USA.,Department of Biological Sciences, St. John's University, Jamaica, NY, USA
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran. .,Isfahan Fertility and Infertility Center, Isfahan, Iran
| |
Collapse
|
42
|
Greither T, Schumacher J, Dejung M, Behre HM, Zischler H, Butter F, Herlyn H. Fertility Relevance Probability Analysis Shortlists Genetic Markers for Male Fertility Impairment. Cytogenet Genome Res 2020; 160:506-522. [PMID: 33238277 DOI: 10.1159/000511117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/26/2020] [Indexed: 12/27/2022] Open
Abstract
Impairment of male fertility is one of the major public health issues worldwide. Nevertheless, genetic causes of male sub- and infertility can often only be suspected due to the lack of reliable and easy-to-use routine tests. Yet, the development of a marker panel is complicated by the large quantity of potentially predictive markers. Actually, hundreds or even thousands of genes could have fertility relevance. Thus, a systematic method enabling a selection of the most predictive markers out of the many candidates is required. As a criterion for marker selection, we derived a gene-specific score, which we refer to as fertility relevance probability (FRP). For this purpose, we first categorized 2,753 testis-expressed genes as either candidate markers or non-candidates, according to phenotypes in male knockout mice. In a parallel approach, 2,502 genes were classified as candidate markers or non-candidates based on phenotypes in men. Subsequently, we conducted logistic regression analyses with evolutionary rates of genes (dN/dS), transcription levels in testis relative to other organs, and connectivity of the encoded proteins in a protein-protein interaction network as covariates. In confirmation of the procedure, FRP values showed the expected pattern, thus being overall higher in genes with known relevance for fertility than in their counterparts without corresponding evidence. In addition, higher FRP values corresponded with an increased dysregulation of protein abundance in spermatozoa of 37 men with normal and 38 men with impaired fertility. Present analyses resulted in a ranking of genes according to their probable predictive power as candidate markers for male fertility impairment. Thus, AKAP4, TNP1, DAZL, BRDT, DMRT1, SPO11, ZPBP, HORMAD1, and SMC1B are prime candidates toward a marker panel for male fertility impairment. Additional candidate markers are DDX4, SHCBP1L, CCDC155, ODF1, DMRTB1, ASZ1, BOLL, FKBP6, SLC25A31, PRSS21, and RNF17. FRP inference additionally provides clues for potential new markers, thereunder TEX37 and POU4F2. The results of our logistic regression analyses are freely available at the PreFer Genes website (https://prefer-genes.uni-mainz.de/).
Collapse
Affiliation(s)
- Thomas Greither
- Center for Reproductive Medicine and Andrology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Julia Schumacher
- Anthropology, Institute of Organismic and Molecular Evolution (iomE), Johannes Gutenberg University Mainz, Mainz, Germany
| | - Mario Dejung
- Quantitative Proteomics, Institute of Molecular Biology (IMB) Mainz, Mainz, Germany
| | - Hermann M Behre
- Center for Reproductive Medicine and Andrology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Hans Zischler
- Anthropology, Institute of Organismic and Molecular Evolution (iomE), Johannes Gutenberg University Mainz, Mainz, Germany
| | - Falk Butter
- Quantitative Proteomics, Institute of Molecular Biology (IMB) Mainz, Mainz, Germany
| | - Holger Herlyn
- Anthropology, Institute of Organismic and Molecular Evolution (iomE), Johannes Gutenberg University Mainz, Mainz, Germany,
| |
Collapse
|
43
|
Lin YH, Huang CY, Ke CC, Wang YY, Lai TH, Liu HC, Ku WC, Chan CC, Lin YH. ACTN4 Mediates SEPT14 Mutation-Induced Sperm Head Defects. Biomedicines 2020; 8:biomedicines8110518. [PMID: 33228246 PMCID: PMC7699536 DOI: 10.3390/biomedicines8110518] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 12/22/2022] Open
Abstract
Septins (SEPTs) are highly conserved GTP-binding proteins and the fourth component of the cytoskeleton. Polymerized SEPTs participate in the modulation of various cellular processes, such as cytokinesis, cell polarity, and membrane dynamics, through their interactions with microtubules, actin, and other cellular components. The main objective of this study was to dissect the molecular pathological mechanism of SEPT14 mutation-induced sperm head defects. To identify SEPT14 interactors, co-immunoprecipitation (co-IP) and nano-liquid chromatography-mass spectrometry/mass spectrometry were applied. Immunostaining showed that SEPT14 was significantly localized to the manchette structure. The SEPT14 interactors were identified and classified as (1) SEPT-, (2) microtubule-, (3) actin-, and (4) sperm structure-related proteins. One interactor, ACTN4, an actin-holding protein, was selected for further study. Co-IP experiments showed that SEPT14 interacts with ACTN4 in a male germ cell line. SEPT14 also co-localized with ACTN4 in the perinuclear and manchette regions of the sperm head in early elongating spermatids. In the cell model, mutated SEPT14 disturbed the localization pattern of ACTN4. In a clinical aspect, sperm with mutant SEPT14, SEPT14A123T (p.Ala123Thr), and SEPT14I333T (p.Ile333Thr), have mislocalized and fragmented ACTN4 signals. Sperm head defects in donors with SEPT14 mutations are caused by disruption of the functions of ACTN4 and actin during sperm head formation.
Collapse
Affiliation(s)
- Yu-Hua Lin
- Department of Chemistry, Fu Jen Catholic University, New Taipei City 242, Taiwan;
- Division of Urology, Department of Surgery, Cardinal Tien Hospital, New Taipei City 231, Taiwan
| | - Chia-Yen Huang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 300, Taiwan;
- Gynecologic Cancer Center, Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei 106, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan; (T.-H.L.); (W.-C.K.)
| | - Chih-Chun Ke
- PhD Program in Nutrition & Food Science, Fu Jen Catholic University, New Taipei City 242, Taiwan;
- Department of Urology, En Chu Kong Hospital, New Taipei City 237, Taiwan
| | - Ya-Yun Wang
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City 242, Taiwan; (Y.-Y.W.); (H.-C.L.)
| | - Tsung-Hsuan Lai
- School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan; (T.-H.L.); (W.-C.K.)
- Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei 106, Taiwan
| | - Hsuan-Che Liu
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City 242, Taiwan; (Y.-Y.W.); (H.-C.L.)
| | - Wei-Chi Ku
- School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan; (T.-H.L.); (W.-C.K.)
| | - Chying-Chyuan Chan
- Department of Obstetrics and Gynecology, Taipei City Hospital, Renai Branch, Taipei 106, Taiwan;
| | - Ying-Hung Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City 242, Taiwan; (Y.-Y.W.); (H.-C.L.)
- Correspondence:
| |
Collapse
|
44
|
Hamza L, Gaitch N, Sallem A, Boucekkine N, Girodon E, Oumeziane A, Attal N, Wolf JP, Bienvenu T. Two frequent loss-of-function mutations in Aurora Kinase C gene in Algerian infertile men with macrozoospermia. Andrologia 2020; 52:e13868. [PMID: 33118205 DOI: 10.1111/and.13868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/06/2020] [Accepted: 09/10/2020] [Indexed: 11/29/2022] Open
Abstract
Macrozoospermia is associated with severe male infertility. To date, the only gene implicated in this phenotype is the Aurora Kinase C gene. We report in this work the genetic screening of AURKC mutations in 34 patients with macrozoospermia among 3,536 Algerian infertile men. Nineteen patients (56%) were homozygotes for the c.144delC mutation, eight (23.52%) homozygotes for the c.744C>G (p.Y248*) mutation and two (5.88%) compound heterozygotes. No AURKC mutation was identified in five patients (14.7%). Interestingly and although it is generally accepted that nearly all positive mutated AURKC patients have close to 100% large-head spermatozoa, our results showed that 11 patients with AURKC mutations (32.35%) had large-headed spermatozoa lower than 70% (7 with c.144delC and 4 with p.Y248*), and no mutation was found in 2 patients who had >70% of macrocephalic spermatozoa. Twenty ICSI attempts were performed before genetic screening resulting in 39 embryos but no pregnancy was obtained. The sequencing of AURKC exons 3 and 6 is appropriate as a first-line genetic exploration in these patients to avoid unsuccessful ICSI attempts. A percentage of large head spermatozoa beyond 25% and a percentage of multiflagellar spermatozoa beyond 10% are predictive of a positive mutation diagnosis.
Collapse
Affiliation(s)
- Loubna Hamza
- Faculté des Sciences Biologiques, Université de Science et de Technologie Houari Boumediane (USTHB), Bab Ezzouar, Algeria.,Centre d'Assistance Médicale à la Procréation Tiziri, Alger, Algeria
| | - Natacha Gaitch
- Assistance Publique - Hôpitaux de Paris, Site Cochin, Laboratoire de Génétique et Biologie Moléculaires, Groupe Universitaire Paris Centre, Paris, France
| | - Amira Sallem
- Assistance Publique - Hôpitaux de Paris, Site Cochin, Service d'Histologie, Embryologie, Biologie de la Reproduction - CECOS, Groupe Universitaire Paris Centre, Paris, France.,Inserm, U1016, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Laboratoire d'Histologie-Embryologie et de Cytogénétique (LR18ES40), Faculté de Médecine de Monastir, Université de Monastir, Monastir, Tunisie
| | - Nadjia Boucekkine
- Centre d'Assistance Médicale à la Procréation Tiziri, Alger, Algeria
| | - Emmanuelle Girodon
- Assistance Publique - Hôpitaux de Paris, Site Cochin, Laboratoire de Génétique et Biologie Moléculaires, Groupe Universitaire Paris Centre, Paris, France
| | - Amina Oumeziane
- Centre d'Assistance Médicale à la Procréation Tiziri, Alger, Algeria
| | - Nabila Attal
- Institut Pasteur d'Algérie ; service d'Immunologie, Dely Ibrahim, Alger, Algeria
| | - Jean Philippe Wolf
- Assistance Publique - Hôpitaux de Paris, Site Cochin, Service d'Histologie, Embryologie, Biologie de la Reproduction - CECOS, Groupe Universitaire Paris Centre, Paris, France.,Inserm, U1016, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,CNRS, UMR8104, Paris, France
| | - Thierry Bienvenu
- Assistance Publique - Hôpitaux de Paris, Site Cochin, Laboratoire de Génétique et Biologie Moléculaires, Groupe Universitaire Paris Centre, Paris, France.,Inserm, U1016, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,CNRS, UMR8104, Paris, France
| |
Collapse
|
45
|
Aitken RJ. The Male Is Significantly Implicated as the Cause of Unexplained Infertility. Semin Reprod Med 2020; 38:3-20. [PMID: 33086406 DOI: 10.1055/s-0040-1718941] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Male infertility is recognized as a relatively common, complex condition, generated by a broad array of environmental and genetic factors. Historical reliance on the conventional semen profile has tended to underestimate the true contribution of "the male factor" to human infertility. This review highlights the importance of genetic and epigenetic factors in the etiology of male infertility, identifying a range of mutations responsible for primary testicular failure and impaired fertilizing potential. More than three quarters of all de novo mutations arise in the male germline via mechanisms that involve the inefficient or defective repair of DNA damage. Understanding the range of factors capable of creating genetic turmoil in the paternal germline is essential, if we are to gain a deep understanding of the causes of male infertility, rather than just the symptoms that characterize its presence. High levels of DNA fragmentation induced by oxidative stress are part of this equation. Oxidative stress is, in turn, driven by biological (age, ejaculation frequency, varicocele, infection), lifestyle (smoking, obesity), and environmental factors (heat, other forms of electromagnetic radiation, and toxins) that can impair the fertilizing potential of the spermatozoa and influence the incidence of spontaneous mutations that may cause infertility in the offspring.
Collapse
Affiliation(s)
- Robert John Aitken
- Priority Research Centre in Reproductive Science, Faculty of Science and Faculty of Health and Medicine, University of Newcastle, Newcastle, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| |
Collapse
|
46
|
Al Zoubi MS, Bataineh H, Rashed M, Al-Trad B, Aljabali AAA, Al-Zoubi RM, Al Hamad M, Issam AbuAlArjah M, Batiha O, Al-Batayneh KM. CAG Repeats in the androgen receptor gene is associated with oligozoospermia and teratozoospermia in infertile men in Jordan. Andrologia 2020; 52:e13728. [PMID: 32583490 DOI: 10.1111/and.13728] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/09/2020] [Accepted: 05/28/2020] [Indexed: 01/06/2023] Open
Abstract
CAG trinucleotide repeats are coded for the polyglutamine tract in the N-terminal of the androgen receptor (AR) gene which varies in normal individuals from 6 to 36 residues. In this study, we inspected the impact of the CAG repeats on the spermatogenic defects by measuring the size of AR-CAG repeats length in a cohort of 260infertile and 169 fertile Jordanian men. The infertile group included three subgroups of a zoospermic, oligozoospermic and teratozoospermia men. The CAG allele size was determined by direct sequencing. The results showed a significant association between the length of the AR-CAG repeats and men's infertility (p = .001). In particular, the current cohort demonstrated a significant association between the AR-CAG length polymorphism and oligozoospermia (p < .001) and teratozoospermia (p < .001) but not azoospermia. According to distributions of allele frequency, the risk of oligozoospermia was 5.5-fold greater than normal when alleles frequency > 20 repeats, while the risk of teratozoospermia was > 10.6 folds greater than normal when allele frequency > 22 repeats. In conclusion, our results underscored that the long repeats of the AR-CAG polymorphism within the normal range might be associated with abnormal spermatogenesis such as teratozoospermia and oligozoospermia and contributing to infertility in Jordanian men.
Collapse
Affiliation(s)
- Mazhar Salim Al Zoubi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Hamzah Bataineh
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid, Jordan
| | - Mitri Rashed
- King Hussein Medical Centre, Royal Medical Services, Amman, Jordan
| | - Bahaa Al-Trad
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid, Jordan
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Raed M Al-Zoubi
- Department of Chemistry, Jordan University of Science & Technology, Irbid, Jordan
| | - Mohammad Al Hamad
- Department of Pathology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Manal Issam AbuAlArjah
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Osamah Batiha
- Department of applied biological sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Khalid M Al-Batayneh
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid, Jordan
| |
Collapse
|
47
|
Macrozoospermia associated with mutations of AURKC gene: First case report in Latin America and literature review. Rev Int Androl 2020; 18:159-163. [DOI: 10.1016/j.androl.2019.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 10/28/2018] [Accepted: 04/15/2019] [Indexed: 11/20/2022]
|
48
|
Li YZ, Wu RF, Zhu XS, Liu WS, Ye YY, Lu ZX, Li N. Identification of a novel deletion mutation in DPY19L2 from an infertile patient with globozoospermia: a case report. Mol Cytogenet 2020; 13:24. [PMID: 32582379 PMCID: PMC7310204 DOI: 10.1186/s13039-020-00495-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/10/2020] [Indexed: 12/26/2022] Open
Abstract
Background Male infertility is an increasing medical concern worldwide. In most cases, genetic factors are considered as the main cause of the disease. Globozoospermia (MIM102530) (also known as round-headed sperm) is a rare and severe malformed spermatospermia caused by acrosome deficiency or severe malformation. A subset of genetic mutations, such as DNAH6, SPATA16, DPY19L2, PICK1, and CCIN related to globozoospermia, have been reported in the past few years. The DPY19L2 mutation is commonly found in patients with globozoospermia. Herein, a 180-kbp homozygote deletion at 12q14.2 (g.63950001–64130000) was identified by copy number variation sequencing (CNVseq) in a patient with a globozoospermia, including the complete deletion of DPY19L2. Case presentation A 27-year-old patient at the First Affiliated Hospital of Xiamen University was diagnosed with infertility because, despite normal sexual activity for 4 years, his wife did not conceive. The patient was in good health with no obvious discomfort, no history of adverse chemical exposure, and no vices, such as smoking and drinking. The physical examination revealed normal genital development. However, semen tests showed a normal sperm count of 0% and the morphology was the round head. Sperm cytology showed that acrosomal enzyme was lower than normal. Reproductive hormones were in the normal range. B ultrasound did not show any abnormal seminal vesicle, prostate, bilateral testis, epididymis, and spermatic veins. The karyotype was normal, 46, XY, and no microdeletion of Y chromosome was detected. However, a homozygous deletion mutation was found in DPY19L2, which was further diagnosed as globozoospermia. Conclusions The present study reported a male infertility patient who was diagnosed with globozoospermia. The analysis of gene mutations revealed that DPY19L2 had a homozygous mutation, which was the primary cause of globozoospermia.
Collapse
Affiliation(s)
- You-Zhu Li
- Reproductive Medicine Center, The First Affiliated Hospital of Xiamen University, No. 6 Guchengxi Road, Si Ming, Xiamen, 361003 China
| | - Rong-Feng Wu
- Reproductive Medicine Center, The First Affiliated Hospital of Xiamen University, No. 6 Guchengxi Road, Si Ming, Xiamen, 361003 China
| | - Xing-Shen Zhu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361005 Fujian China
| | - Wen-Sheng Liu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361005 Fujian China
| | - Yuan-Yuan Ye
- Reproductive Medicine Center, The First Affiliated Hospital of Xiamen University, No. 6 Guchengxi Road, Si Ming, Xiamen, 361003 China
| | - Zhong-Xian Lu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361005 Fujian China
| | - Na Li
- Intensive Care Unit, Fujian Medical University Xiamen Humanity Hospital, No.3777 Xianyue Road, Huli, Xiamen, 361009 China
| |
Collapse
|
49
|
Bai S, Hu X, Zhao Y, Li W, Wan Y, Jin R, Wang Y, Guo T, Tong X, Xu B. Compound heterozygosity for novel AURKC mutations in an infertile man with macrozoospermia. Andrologia 2020; 52:e13663. [PMID: 32478938 DOI: 10.1111/and.13663] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/25/2020] [Accepted: 05/05/2020] [Indexed: 01/23/2023] Open
Abstract
Among causes of infertility, teratozoospermia is characterised by a percentage of morphologically abnormal spermatozoa >4%. Macrozoospermia, one form of monomorphic teratozoospermia, is observed in <1% of cases of male infertility and is described as approximately 100% large-headed and/or multitailed spermatozoa. This study reports that an infertile man with large-head spermatozoa presenting compound heterozygosity aurora kinase C (AURKC) mutations (c.382C>T, c.572C>T) by whole-exome sequencing. Consequently, both two novel AURKC mutations had high probability of damage-causing and conserved across species and extremely low allele frequency in the population. Flow cytometry analysis revealed a high ratio of sperm DNA fragmentation. Two intracytoplasmic sperm injection (ICSI) procedures were attempted for the patient, but all were unsuccessful. These results indicate that sequence analysis should be performed for the variants of AURKC in Chinese patients with macrozoospermia.
Collapse
Affiliation(s)
- Shun Bai
- Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xuechun Hu
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yun Zhao
- Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wei Li
- Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yangyang Wan
- Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Rentao Jin
- Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yanshi Wang
- Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Tonghang Guo
- Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xianhong Tong
- Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Bo Xu
- Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
50
|
Absence of murine CFAP61 causes male infertility due to multiple morphological abnormalities of the flagella. Sci Bull (Beijing) 2020; 65:854-864. [PMID: 36659204 DOI: 10.1016/j.scib.2020.01.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/08/2019] [Accepted: 01/03/2020] [Indexed: 01/21/2023]
Abstract
Impaired flagellar development and impaired motility of sperm is a cause of infertility in males. Several genes, including those of the AKAP, CCDC, CFAP, and DNAH families, among others, are involved in the "multiple morphological abnormalities of the flagella" (MMAF) phenotype; these are the most common causes of male infertility. The Cilia-and flagella-associated protein (CFAP) family includes six members reported to cause MMAF phenotypes: CFAP43, CFAP44, CFAP69, CFAP65, CFAP70, and CFAP251. Here, we found that cilia-and flagella-associated protein 61 (Cfap61) is highly expressed specifically in murine testes and show that the Cfap61-knockout male mice demonstrate MMAF phenotype, including sperm with short, coiled, and irregular flagella. Deletion of Cfap61 resulted in severe morphological and behavior abnormalities in sperm, reduced total sperm counts, impaired sperm motility, and led to male infertility. Notably, absence of Cfap61 impaired sperm flagella ultrastructural abnormalities on account of numerous distortions in multiple flagellum components. Immunostaining experiments in wild-type mice and healthy adult humans indicated that Cfap61 is initially localized at the neck of sperm, where it potentially functions in flagellum formation, and is later localized to the midpiece of the sperm. Thus, our study provides compelling evidence that dysregulation of Cfap61 affects sperm flagellum development and induces male infertility in mice. Further investigations of the CFAP61 gene in humans alongside clinical evidence showing MMAF phenotype in humans should contribute to our understanding of developmental processes underlying sperm flagellum formation and the pathogenic mechanisms that cause male infertility.
Collapse
|