Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 371
Filter
1.
Hum Genomics ; 18(1): 97, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39256880

ABSTRACT

BACKGROUND: Bi-allelic variants in DNAH11 have been identified as causative factors in Primary Ciliary Dyskinesia, leading to abnormal respiratory cilia. Nonetheless, the specific impact of these variants on human sperm flagellar and their involvement in male infertility remain largely unknown. METHODS: A collaborative effort involving two Chinese reproductive centers conducted a study with 975 unrelated infertile men. Whole-exome sequencing was employed for variant screening, and Sanger sequencing confirmed the identified variants. Morphological and ultrastructural analyses of sperm were conducted using Scanning Electron Microscopy and Transmission Electron Microscopy. Western Blot Analysis and Immunofluorescence Analysis were utilized to assess protein levels and localization. ICSI was performed to evaluate its efficacy in achieving favorable pregnancy outcomes for individuals with DNAH11 variants. RESULTS: In this study, we identified seven novel variants in the DNAH11 gene in four asthenoteratozoospermia subjects. These variants led the absence of DNAH11 proteins and ultrastructure defects in sperm flagella, particularly affecting the outer dynein arms (ODAs) and adjacent structures. The levels of ODA protein DNAI2 and axoneme related proteins were down regulated, instead of inner dynein arms (IDA) proteins DNAH1 and DNAH6. Two out of four individuals with DNAH11 variants achieved clinical pregnancies through ICSI. The findings confirm the association between male infertility and bi-allelic deleterious variants in DNAH11, resulting in the aberrant assembly of sperm flagella and contributing to asthenoteratozoospermia. Importantly, ICSI emerges as an effective intervention for overcoming reproductive challenges caused by DNAH11 gene variants.


Subject(s)
Asthenozoospermia , Axonemal Dyneins , Exome Sequencing , Infertility, Male , Humans , Male , Asthenozoospermia/genetics , Asthenozoospermia/pathology , Axonemal Dyneins/genetics , Female , Infertility, Male/genetics , Infertility, Male/pathology , Adult , Sperm Tail/pathology , Sperm Tail/ultrastructure , Sperm Tail/metabolism , Sperm Injections, Intracytoplasmic , Pregnancy , Spermatozoa/ultrastructure , Spermatozoa/pathology , Dyneins/genetics
2.
Clin Genet ; 106(4): 437-447, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39221575

ABSTRACT

Male infertility due to asthenozoospermia is quite frequent, but its etiology is poorly understood. We recruited two infertile brothers, born to first-cousin parents from Pakistan, displaying idiopathic asthenozoospermia with mild stuttering disorder but no ciliary-related symptoms. Whole-exome sequencing identified a splicing variant (c.916+1G>A) in ARMC3, recessively co-segregating with asthenozoospermia in the family. The ARMC3 protein is evolutionarily highly conserved and is mostly expressed in the brain and testicular tissue of human. The ARMC3 splicing mutation leads to the exclusion of exon 8, resulting in a predicted truncated protein (p.Glu245_Asp305delfs*16). Quantitative real-time PCR revealed a significant decrease at mRNA level for ARMC3 and Western blot analysis did not detect ARMC3 protein in the patient's sperm. Individuals homozygous for the ARMC3 splicing variant displayed reduced sperm motility with frequent morphological abnormalities of sperm flagella. Transmission electron microscopy of the affected individual IV: 2 revealed vacuolation in sperm mitochondria at the midpiece and disrupted flagellar ultrastructure in the principal and end piece. Altogether, our results indicate that this novel homozygous ARMC3 splicing mutation destabilizes sperm flagella and leads to asthenozoospermia in our patients, providing a novel marker for genetic counseling and diagnosis of male infertility.


Subject(s)
Asthenozoospermia , Consanguinity , Homozygote , Pedigree , RNA Splicing , Sperm Tail , Adult , Humans , Male , Asthenozoospermia/genetics , Asthenozoospermia/pathology , Exome Sequencing , Infertility, Male/genetics , Infertility, Male/pathology , Mutation , RNA Splicing/genetics , Sperm Motility/genetics , Sperm Tail/pathology , Sperm Tail/ultrastructure , Sperm Tail/metabolism , Spermatozoa/ultrastructure , Spermatozoa/pathology
3.
Zool Res ; 45(5): 1073-1087, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39245651

ABSTRACT

Infertility represents a significant health concern, with sperm quantity and quality being crucial determinants of male fertility. Oligoasthenoteratozoospermia (OAT) is characterized by reduced sperm motility, lower sperm concentration, and morphological abnormalities in sperm heads and flagella. Although variants in several genes have been implicated in OAT, its genetic etiologies and pathogenetic mechanisms remain inadequately understood. In this study, we identified a homozygous nonsense mutation (c.916C>T, p.Arg306*) in the coiled-coil domain containing 146 ( CCDC146) gene in an infertile male patient with OAT. This mutation resulted in the production of a truncated CCDC146 protein (amino acids 1-305), retaining only two out of five coiled-coil domains. To validate the pathogenicity of the CCDC146 mutation, we generated a mouse model ( Ccdc146 mut/mut ) with a similar mutation to that of the patient. Consistently, the Ccdc146 mut/mut mice exhibited infertility, characterized by significantly reduced sperm counts, diminished motility, and multiple defects in sperm heads and flagella. Furthermore, the levels of axonemal proteins, including DNAH17, DNAH1, and SPAG6, were significantly reduced in the sperm of Ccdc146 mut/mut mice. Additionally, both human and mouse CCDC146 interacted with intraflagellar transport protein 20 (IFT20), but this interaction was lost in the mutated versions, leading to the degradation of IFT20. This study identified a novel deleterious homozygous nonsense mutation in CCDC146 that causes male infertility, potentially by disrupting axonemal protein transportation. These findings offer valuable insights for genetic counseling and understanding the mechanisms underlying CCDC146 mutant-associated infertility in human males.


Subject(s)
Asthenozoospermia , Microtubule-Associated Proteins , Animals , Humans , Male , Mice , Asthenozoospermia/genetics , Codon, Nonsense , Homozygote , Infertility, Male/genetics , Mutation , Oligospermia/genetics , Sperm Motility/genetics , Spermatozoa , Microtubule-Associated Proteins/genetics
4.
Nat Commun ; 15(1): 8465, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39349455

ABSTRACT

Spermiogenesis, the complex transformation of haploid spermatids into mature spermatozoa, relies on precise spatiotemporal regulation of gene expression at the post-transcriptional level. The mechanisms underlying this critical process remain incompletely understood. Here, we identify centrosomal protein 112 (CEP112) as an essential regulator of mRNA translation during this critical developmental process. Mutations in CEP112 are discovered in oligoasthenoteratospermic patients, and Cep112-deficient male mice recapitulate key phenotypes of human asthenoteratozoospermia. CEP112 localizes to the neck and atypical centrioles of mature sperm and forms RNA granules during spermiogenesis, enriching target mRNAs such as Fsip2, Cfap61, and Cfap74. Through multi-omics analyses and the TRICK reporter assay, we demonstrate that CEP112 orchestrates the translation of target mRNAs. Co-immunoprecipitation and mass spectrometry identify CEP112's interactions with translation-related proteins, including hnRNPA2B1, EEF1A1, and EIF4A1. In vitro, CEP112 undergoes liquid-liquid phase separation, forming condensates that recruit essential proteins and mRNAs. Moreover, variants in patient-derived CEP112 disrupt phase separation and impair translation efficiency. Our results suggest that CEP112 mediates the assembly of RNA granules through liquid-liquid phase separation to control the post-transcriptional expression of fertility-related genes. This study not only clarifies CEP112's role in spermatogenesis but also highlights the role of phase separation in translational regulation, providing insights into male infertility and suggesting potential therapeutic targets.


Subject(s)
Protein Biosynthesis , Spermatogenesis , Male , Animals , Spermatogenesis/genetics , Humans , Mice , RNA, Messenger/metabolism , RNA, Messenger/genetics , Fertility/genetics , Mice, Knockout , Asthenozoospermia/genetics , Asthenozoospermia/metabolism , Mutation , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Spermatozoa/metabolism , Spermatids/metabolism , Oligospermia/genetics , Oligospermia/metabolism , Gene Expression Regulation , Phase Separation
5.
J Cell Mol Med ; 28(18): e18580, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39331689

ABSTRACT

Oligo-astheno-teratozoospermia (OAT) is a common cause of male infertility, but the genetic basis of most OAT cases is still unknown. Here, one homozygous loss-of-function (LOF) variant in TDRD6, c.G1825T/p.Gly609X, was identified in an infertile patient with severe OAT by whole-exome sequencing (WES) and Sanger confirmation. Furthermore, Tdrd6-mutant mice (p.Gly615X; equivalent to p.Gly609X in human TDRD6) were generated. Remarkably, the Tdrd6-mutated mice mimicked the severe OAT symptoms of the patient. In addition, the architecture of chromatoid bodies (CBs) were disrupted in round spermatids from Tdrd6-mutant mice, leading to blocked spermatogenesis in the round spermatids. The assembly of PIWIL1, TDRD1, TDRD7 and DDX25 in CBs was disturbed in the Tdrd6-mutant mice. Applying immunoprecipitation-mass spectrometry (IP-MS), we identified some TDRD6-interacting partners, including CB proteins TDRD7, MAEL and PCBP1. Moreover, we described the assisted reproductive technology (ART) outcomes of the infertile patient and his partner. Altogether, our findings provide necessary evidences to support the idea that the homozygous LOF variant in TDRD6 induces male infertility with severe OAT, suggesting that TDRD6 could be a useful genetic diagnostic target for male infertility.


Subject(s)
Infertility, Male , Male , Animals , Humans , Mice , Infertility, Male/genetics , Infertility, Male/pathology , Spermatogenesis/genetics , Loss of Function Mutation , Exome Sequencing , Teratozoospermia/genetics , Teratozoospermia/pathology , Oligospermia/genetics , Oligospermia/pathology , Asthenozoospermia/genetics , Asthenozoospermia/pathology , Disease Models, Animal , Homozygote , Adult
6.
J Cell Mol Med ; 28(18): e70092, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39289782

ABSTRACT

Primary ciliary dyskinesia (PCD) is an autosomal recessive genetic disorder characterized by ultrastructural defects in the cilia or flagella of cells, causing respiratory abnormalities, sinusitis, visceral transposition, and male infertility. DNAAF3 plays an important role in the assembly and transportation of axonemal dynein complexes in cilia or flagella and has been shown to be associated with PCD. To date, only two cases of PCD with infertility associated with DNAAF3 mutations have been reported, and no mouse models for this gene have been successfully constructed. This study was conducted on an infertile Chinese male patient with a history of bronchitis. Examination of the patient's semen revealed severe asthenozoospermia and teratospermia. Whole exome sequencing revealed a new homozygous loss-of-function DNAAF3 mutation. CRISPR-Cas9 gene-editing technology was used to construct the same mutation in C57/B6 mice, revealing that homozygous C57/B6 mice were characterized by severe hydrocephalus and early death. The results of this study expand the mutation spectrum of DNAAF3 and confirm its correlation with PCD pathogenesis. This study provides new insights on the mechanisms underlying male infertility related to DNAAF3 mutation and PCD.


Subject(s)
Asthenozoospermia , Homozygote , Mutation , Teratozoospermia , Male , Humans , Animals , Asthenozoospermia/genetics , Asthenozoospermia/pathology , Mice , Mutation/genetics , Teratozoospermia/genetics , Exome Sequencing , Infertility, Male/genetics , Mice, Inbred C57BL , Adult , Ciliary Motility Disorders/genetics
7.
Syst Biol Reprod Med ; 70(1): 272-288, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39292564

ABSTRACT

Asthenozoospermia (AZS) is one of the most common types of male infertility. Current evidence revealed that type 2 diabetes mellitus (T2DM) is closely associated with declining semen quality, especially for poor sperm motility. This study aimed to uncover the genetic interrelationships and important biomarkers between AZS and T2DM. Transcriptome data regarding AZS and T2DM were downloaded from the Gene Expression Omnibus (GEO) database. We performed GO and pathway analysis, and protein-protein interaction (PPI) network construction for T2DM-related differentially expressed genes (DMRGs). Moreover, we calculated receiver operator characteristic (ROC) curve and conducted external independent validation. Expression of hub DMRGs was assessed for patients using the qPCR method. MiRNA interaction and immune infiltration were subsequently characterized. A total of 554 overlapping DMRGs were identified between the AZS/T2DM and healthy groups. These overlapping DMRG participated in the DNA damage-, energy metabolism-, and immune-related biological pathways. Module function analysis discovered that the top three PPI modules were tightly correlated with DNA damage-related processes. After external validation in other independent datasets, two hub DMRGs (TBC1D12 and SCG5) were obtained. ROC analysis revealed that TBC1D12 and SCG5 had good diagnostic performance (area under the curve > 0.75). Immune infiltration profile showed that the level of T cell co-stimulation and CD8+_T_cells were negatively related to the hub DMRGs expression. Mirna interaction analysis showed 15 significant hub DMRGs-miRNA interactions. The qPCR results showed that expression of TBC1D12 and SCG5 were significantly different between sperm samples from diabetic patients with AZS and controls. The present study revealed molecular signatures and critical pathways between the AZS and T2DM, and identified two hub DMRGs of TBC1D12 and SCG5. The data would provide novel understandings of shared pathogenic mechanisms in T2DM-associated AZS.


Subject(s)
Asthenozoospermia , Diabetes Mellitus, Type 2 , Humans , Male , Diabetes Mellitus, Type 2/genetics , Asthenozoospermia/genetics , Protein Interaction Maps , Gene Regulatory Networks , MicroRNAs/genetics , Computer Simulation , Transcriptome , Gene Expression Profiling , Databases, Genetic
8.
BMC Urol ; 24(1): 180, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39192248

ABSTRACT

BACKGROUND: Male infertility due to spermatogenesis defects affects millions of men worldwide. However, the genetic etiology of the vast majority remains unclear. The present study was undertaken to assess the association of DNAH6 and ATPase6 genes in asthenozoospermia patients in the northern region of India. METHODS: A total of 60 semen samples were collected for the study, of which 30 were from the case group and 30 were from the control group. The semen samples for the case group (asthenozoospermia) and control groups were collected from IVF and Reproductive Biology Centre, Maulana Azad Medical College, New Delhi. Sperm count and motility were classified as per World Health Organization (WHO 2021) protocol. A total genomic DNA was extracted as per the stranded TRIZOL method with little modification. RESULTS: In-vitro molecular characterizations of DNAH6 and ATPase6 genes in both groups were checked by Polymerase Chain Reaction (PCR). The 675 bp and 375 bp amplicons were amplified using PCR for ATPase6 and DNAH6 genes. Our study results showed a significant (P ≤ 0.05) null deletion of DNAH6 and ATPase6 genes in asthenozoospermia patients as compared to the control. We found the significant null deletion of DNAH6 in case 45.0%, and the control group was 11.7%. However, in the case of APTase6, it was 26.7% and 10.0%, respectively. CONCLUSIONS: Our study concluded that the presence of DHAH6 and ATPase6 genes had a significant impact on male infertility.


Subject(s)
Asthenozoospermia , Humans , Male , Asthenozoospermia/genetics , India , Adult , Mitochondrial Proton-Translocating ATPases/genetics , DNA, Mitochondrial/genetics
9.
Zhonghua Nan Ke Xue ; 30(3): 199-208, 2024 Mar.
Article in Chinese | MEDLINE | ID: mdl-39177385

ABSTRACT

OBJECTIVE: To explore the potential impact of lipid metabolism-related single nucleotide polymorphisms (SNP) on semen quality in men. METHODS: We selected 284 semen samples from Xingtai Infertility Hospital and Hebei Human Sperm Bank collected between February and October 2023, 33 from oligozoospermia (OS), 97 from asthenozoospermia (AS) and 54 from oligoasthenozoospermia (OAS) patients and the other 100 from normal men. We performed computer-assisted semen analysis (CASA) of the samples, extracted blood DNA and, using the MassARRAY System, genotyped the target genes, determined the genotypes of 13 SNPs and compared their distribution, their correlation with BMI and semen quality in different groups. RESULTS: The mutant homozygous (TT) genotype of the FADS2 rs2727270 gene seemed to be a risk factor for AS (OR = 4.420, P= 0.047), while the APOA2 rs5082-A allele and MC4R rs17782313 heterozygous (TC) genotype important protective factors for OS (OR = 0.422 and 0.389; P= 0.045 and 0.043, respectively). A significantly higher sperm concentration was found associated with the MC4R rs17782313 heterozygous (TC) genotype than with the homozygous (CC) genotype. Stratification analysis showed that the protective effect of the TC genotype was decreased with increased BMI and remained with the interaction of the rs5082 and rs17782313 genotypes. CONCLUSION: FADS2 rs2727270, APOA2 rs5082 and MC4R rs17782313 were significantly correlated with the risk of abnormal semen parameters.


Subject(s)
Genotype , Lipid Metabolism , Polymorphism, Single Nucleotide , Semen Analysis , Humans , Male , Lipid Metabolism/genetics , Asthenozoospermia/genetics , Fatty Acid Desaturases/genetics , Oligospermia/genetics , Infertility, Male/genetics , Alleles , Adult , Sperm Count , Risk Factors , Spermatozoa/metabolism
10.
Mol Genet Genomics ; 299(1): 69, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38992144

ABSTRACT

TTC12 is a cytoplasmic and centromere-localized protein that plays a role in the proper assembly of dynein arm complexes in motile cilia in both respiratory cells and sperm flagella. This finding underscores its significance in cellular motility and function. However, the wide role of TTC12 in human spermatogenesis-associated primary ciliary dyskinesia (PCD) still needs to be elucidated. Whole-exome sequencing (WES) and Sanger sequencing were performed to identify potentially pathogenic variants causing PCD and multiple morphological abnormalities of sperm flagella (MMAF) in an infertile Pakistani man. Diagnostic imaging techniques were used for PCD screening in the patient. Real-time polymerase chain reaction (RT‒PCR) was performed to detect the effect of mutations on the mRNA abundance of the affected genes. Papanicolaou staining and scanning electron microscopy (SEM) were carried out to examine sperm morphology. Transmission electron microscopy (TEM) was performed to examine the ultrastructure of the sperm flagella, and the results were confirmed by immunofluorescence staining. Using WES and Sanger sequencing, a novel homozygous missense variant (c.C1069T; p.Arg357Trp) in TTC12 was identified in a patient from a consanguineous family. A computed tomography scan of the paranasal sinuses confirmed the symptoms of the PCD. RT-PCR showed a decrease in TTC12 mRNA in the patient's sperm sample. Papanicolaou staining, SEM, and TEM analysis revealed a significant change in shape and a disorganized axonemal structure in the sperm flagella of the patient. Immunostaining assays revealed that TTC12 is distributed throughout the flagella and is predominantly concentrated in the midpiece in normal spermatozoa. In contrast, spermatozoa from patient deficient in TTC12 showed minimal staining intensity for TTC12 or DNAH17 (outer dynein arms components). This could lead to MMAF and result in male infertility. This novel TTC12 variant not only illuminates the underlying genetic causes of male infertility but also paves the way for potential treatments targeting these genetic factors. This study represents a significant advancement in understanding the genetic basis of PCD-related infertility.


Subject(s)
Homozygote , Infertility, Male , Mutation, Missense , Sperm Tail , Humans , Male , Mutation, Missense/genetics , Pakistan , Infertility, Male/genetics , Infertility, Male/pathology , Sperm Tail/pathology , Sperm Tail/ultrastructure , Sperm Tail/metabolism , Adult , Pedigree , Asthenozoospermia/genetics , Asthenozoospermia/pathology , Ciliary Motility Disorders/genetics , Ciliary Motility Disorders/pathology , Exome Sequencing , Oligospermia/genetics , Oligospermia/pathology , Kartagener Syndrome/genetics , Kartagener Syndrome/pathology
11.
Zhonghua Nan Ke Xue ; 30(1): 18-25, 2024 Jan.
Article in Chinese | MEDLINE | ID: mdl-39046409

ABSTRACT

OBJECTIVE: To explore the expressions of zinc homeostasis-related proteins, G protein-coupled receptor 39 (GPR39) and ANO1 mRNA in the sperm of patients with asthenozoospermia (AS), and analyze their correlation with sperm motility. METHODS: We collected semen samples from 82 male subjects with PR+NP < 40%, PR < 32% and sperm concentration > 15×106/ml (the AS group, n = 40) or PR+NP ≥ 40%, PR ≥ 32% and sperm concentration > 15×106/ml (the normal control group, n = 42). We analyzed the routine semen parameters and measured the zinc content in the seminal plasma using the computer-assisted sperm analysis system, detected the expressions of zinc transporters (ZIP13, ZIP8 and ZNT10), metallothioneins (MT1G, MT1 and MTF), GPR39, and calcium-dependent chloride channel protein (ANO1) in the sperm by real-time quantitative PCR (RT qPCR), examined free zinc distribution in the sperm by laser confocal microscopy, and determined the expressions of GPR39 and MT1 proteins in the sperm by immunofluorescence staining, followed by Spearman rank correlation analysis of their correlation with semen parameters. RESULTS: There was no statistically significant difference in the zinc concentration in the seminal plasma between the AS and normal control groups (P>0.05). Compared with the controls, the AS patients showed a significantly reduced free zinc level (P<0.05), relative expressions of MT1G, MTF, ZIP13, GPR39 and ANO1 mRNA (P<0.05), and that of the GPR39 protein in the AS group (P<0.05). No statistically significant differences were observed in the relative expression levels of ZIP8, ZNT10 and MT1 mRNA between the two groups (P>0.05). The relative expression levels of GPR39, ANO1, MT1G and MTF mRNA were positively correlated with sperm motility and the percentage of progressively motile sperm (P<0.05). CONCLUSION: The expressions of zinc homeostasis proteins (MT1G, MTF and ZIP13), GPR39 and ANO1 mRNA are downregulated in the sperm of asthenozoospermia patients, and positively correlated with sperm motility.


Subject(s)
Anoctamin-1 , Asthenozoospermia , Cation Transport Proteins , RNA, Messenger , Receptors, G-Protein-Coupled , Sperm Motility , Spermatozoa , Zinc , Humans , Male , Asthenozoospermia/metabolism , Asthenozoospermia/genetics , Anoctamin-1/metabolism , Anoctamin-1/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Zinc/metabolism , Spermatozoa/metabolism , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Metallothionein/metabolism , Metallothionein/genetics , Homeostasis , Adult , Semen Analysis , Clinical Relevance , Neoplasm Proteins
12.
Cell Mol Life Sci ; 81(1): 317, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39066891

ABSTRACT

Inner dynein arms (IDAs) are formed from a protein complex that is essential for appropriate flagellar bending and beating. IDA defects have previously been linked to the incidence of asthenozoospermia (AZS) and male infertility. The testes-enriched ZMYND12 protein is homologous with an IDA component identified in Chlamydomonas. ZMYND12 deficiency has previously been tied to infertility in males, yet the underlying mechanism remains uncertain. Here, a CRISPR/Cas9 approach was employed to generate Zmynd12 knockout (Zmynd12-/-) mice. These Zmynd12-/- mice exhibited significant male subfertility, reduced sperm motile velocity, and impaired capacitation. Through a combination of co-immunoprecipitation and mass spectrometry, ZMYND12 was found to interact with TTC29 and PRKACA. Decreases in the levels of PRKACA were evident in the sperm of these Zmynd12-/- mice, suggesting that this change may account for the observed drop in male fertility. Moreover, in a cohort of patients with AZS, one patient carrying a ZMYND12 variant was identified, expanding the known AZS-related variant spectrum. Together, these findings demonstrate that ZMYND12 is essential for flagellar beating, capacitation, and male fertility.


Subject(s)
Infertility, Male , Mice, Knockout , Sperm Motility , Animals , Humans , Male , Mice , Asthenozoospermia/genetics , Asthenozoospermia/metabolism , Asthenozoospermia/pathology , CRISPR-Cas Systems , Dyneins/metabolism , Dyneins/genetics , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Mice, Inbred C57BL , Sperm Capacitation/genetics , Sperm Motility/genetics , Spermatozoa/metabolism , Contactin 2/genetics , Contactin 2/metabolism
13.
Asian J Androl ; 26(5): 484-489, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39054792

ABSTRACT

ABSTRACT: The cause of asthenozoospermia (AZS) is not well understood because of its complexity and heterogeneity. Although some gene mutations have been identified as contributing factors, they are only responsible for a small number of cases. Radial spokes (RSs) are critical for adenosine triphosphate-driven flagellar beating and axoneme stability, which is essential for flagellum motility. In this study, we found novel compound heterozygous mutations in leucine-rich repeat-containing protein 23 ( LRRC23 ; c.1018C>T: p.Q340X and c.881_897 Del: p.R295Gfs*32) in a proband from a nonconsanguineous family with AZS and male infertility. Diff-Quik staining and scanning electron microscopy revealed no abnormal sperm morphology. Western blotting and immunofluorescence staining showed that these mutations suppressed LRRC23 expression in sperm flagella. Additionally, transmission electron microscopy showed the absence of RS3 in sperm flagella, which disrupts stability of the radial spoke complex and impairs motility. Following in vitro fertilization and embryo transfer, the proband's spouse achieved successful pregnancy and delivered a healthy baby. In conclusion, our study indicates that two novel mutations in LRRC23 are associated with AZS, but successful fertility outcomes can be achieved by in vitro fertilization-embryo transfer techniques.


Subject(s)
Asthenozoospermia , Mutation , Adult , Female , Humans , Male , Pregnancy , Asthenozoospermia/genetics , Pedigree , Sperm Motility/genetics , Sperm Tail/pathology , Sperm Tail/ultrastructure , Sperm Tail/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism
14.
Genet Test Mol Biomarkers ; 28(8): 337-341, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38837151

ABSTRACT

Aims: Asthenozoospermia is the most common factor of male infertility, mainly caused by multiple morphological abnormalities of the sperm flagella (MMAF) and primary ciliary dyskinesia (PCD). Previous studies have shown that genetic factors may contribute to MMAF and PCD. The study aimed to identify novel potentially pathogenic gene mutations in a Chinese infertile man with MMAF and PCD-like phenotypes. Methods: A Chinese infertile man with MMAF and PCD was enrolled in this study. Whole exome sequencing and Sanger sequencing were performed to identify potential causative genes and mutations. Results: A novel homozygous missense mutation (c.1450G>A; p.E484K) of CCDC40 was finally identified and Sanger sequencing confirmed that the patient carried the homozygous mutation, which was inherited from his parents. We reported the first homozygous missense CCDC40 mutation in infertile men with MMAF but had other milder PCD symptoms. Conclusion: Our findings not only broaden the disease-causing mutation spectrum of CCDC40 but also provide new insight into the correlation between CCDC40 mutations and MMAF.


Subject(s)
Asian People , Homozygote , Infertility, Male , Mutation, Missense , Phenotype , Sperm Tail , Humans , Male , Infertility, Male/genetics , Mutation, Missense/genetics , Adult , China , Asian People/genetics , Sperm Tail/metabolism , Sperm Tail/pathology , Ciliary Motility Disorders/genetics , Exome Sequencing/methods , Pedigree , Mutation , Asthenozoospermia/genetics , East Asian People
15.
J Genet Genomics ; 51(10): 1007-1019, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38909778

ABSTRACT

Oligoasthenoteratozoospermia is an important factor affecting male fertility and has been found to be associated with genetic factors. However, there are still a proportion of oligoasthenoteratozoospermia cases that cannot be explained by known pathogenic genetic variants. Here, we perform genetic analyses and identify bi-allelic loss-of-function variants of MFSD6L from an oligoasthenoteratozoospermia-affected family. Mfsd6l knock-out male mice also present male subfertility with reduced sperm concentration, motility, and deformed acrosomes. Further mechanistic analyses reveal that MFSD6L, as an acrosome membrane protein, plays an important role in the formation of acrosome by interacting with the inner acrosomal membrane protein SPACA1. Moreover, poor embryonic development is consistently observed after intracytoplasmic sperm injection treatment using spermatozoa from the MFSD6L-deficient man and male mice. Collectively, our findings reveal that MFSD6L is required for the anchoring of sperm acrosome and head shaping. The deficiency of MFSD6L affects male fertility and causes oligoasthenoteratozoospermia in humans and mice.


Subject(s)
Acrosome , Membrane Proteins , Mice, Knockout , Male , Animals , Mice , Acrosome/pathology , Acrosome/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Asthenozoospermia/genetics , Asthenozoospermia/pathology , Infertility, Male/genetics , Infertility, Male/pathology , Spermatozoa/metabolism , Spermatozoa/pathology , Sperm Motility/genetics , Oligospermia/genetics , Oligospermia/pathology
16.
BMC Genomics ; 25(1): 602, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886667

ABSTRACT

BACKGROUND: Spermatogenesis is a highly regulated and complex process in which DNA methylation plays a crucial role. This study aimed to explore the differential methylation profiles in sperm DNA between patients with asthenospermia (AS) and healthy controls (HCs), those with oligoasthenospermia (OAS) and HCs, and patients with AS and those with OAS. RESULTS: Semen samples and clinical data were collected from five patients with AS, five patients with OAS, and six age-matched HCs. Reduced representation bisulfite sequencing (RRBS) was performed to identify differentially methylated regions (DMRs) in sperm cells among the different types of patients and HCs. A total of 6520, 28,019, and 16,432 DMRs were detected between AS and HC, OAS and HC, and AS and OAS groups, respectively. These DMRs were predominantly located within gene bodies and mapped to 2868, 9296, and 9090 genes in the respective groups. Of note, 12, 9, and 8 DMRs in each group were closely associated with spermatogenesis and male infertility. Furthermore, BDNF, SMARCB1, PIK3CA, and DDX27; RBMX and SPATA17; ASZ1, CDH1, and CHDH were identified as strong differentially methylated candidate genes in each group, respectively. Meanwhile, the GO analysis of DMR-associated genes in the AS vs. HC groups revealed that protein binding, cytoplasm, and transcription (DNA-templated) were the most enriched terms in the biological process (BP), cellular component (CC), and molecular function (MF), respectively. Likewise, in both the OAS vs. HC and AS vs. OAS groups, GO analysis revealed protein binding, nucleus, and transcription (DNA-templated) as the most enriched terms in BP, CC, and MF, respectively. Finally, the KEGG analysis of DMR-annotated genes and these genes at promoters suggested that metabolic pathways were the most significantly associated across all three groups. CONCLUSIONS: The current study results revealed distinctive sperm DNA methylation patterns in the AS vs. HC and OAS vs. HC groups, particularly between patients with AS and those with OAS. The identification of key genes associated with spermatogenesis and male infertility in addition to the differentially enriched metabolic pathways may contribute to uncovering the potential pathogenesis in different types of abnormal sperm parameters.


Subject(s)
Asthenozoospermia , DNA Methylation , Oligospermia , Humans , Male , Asthenozoospermia/genetics , Adult , Oligospermia/genetics , Spermatozoa/metabolism , Spermatogenesis/genetics , Case-Control Studies , Epigenesis, Genetic
17.
Front Endocrinol (Lausanne) ; 15: 1373774, 2024.
Article in English | MEDLINE | ID: mdl-38863929

ABSTRACT

Background: Asthenozoospermia, a type of male infertility, is primarily caused by dysfunctional sperm mitochondria. Despite previous bioinformatics analysis identifying potential key lncRNAs, miRNAs, hub genes, and pathways associated with asthenospermia, there is still a need to explore additional molecular mechanisms and potential biomarkers for this condition. Methods: We integrated data from Gene Expression Omnibus (GEO) (GSE22331, GSE34514, and GSE160749) and performed bioinformatics analysis to identify differentially expressed genes (DEGs) between normozoospermia and asthenozoospermia. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were conducted to gain insights into biological processes and signaling pathways. Weighted Gene Co-expression Network Analysis (WGCNA) identified gene modules associated with asthenozoospermia. Expression levels of key genes were assessed using datasets and experimental data. Gene Set Enrichment Analysis (GSEA) and correlation analysis identified pathways associated with the hub gene and explore the relationship between the ZNF764 and COQ9 and mitochondrial autophagy-related genes. Competitive endogenous RNA (ceRNA) networks were constructed, and in vitro experiments using exosome samples were conducted to validate this finding. Results: COQ9 was identified as a marker gene in asthenozoospermia, involved in autophagy, ATP-dependent chromatin remodeling, endocytosis, and cell cycle, etc. The ceRNA regulatory network (LINC00893/miR-125a-5p/COQ9) was constructed, and PCR demonstrated that LINC00893 and COQ9 were downregulated in asthenozoospermia, while miR-125a-5p and m6A methylation level of LINC00893 were upregulated in asthenozoospermia compared to normozoospermic individuals. Conclusion: The ceRNA regulatory network (LINC00893/miR-125a-5p/COQ9) likely plays a crucial role in the mechanism of asthenozoospermia. However, further functional experiments are needed to fully understand its significance.


Subject(s)
Asthenozoospermia , Biomarkers , Computational Biology , Gene Regulatory Networks , Humans , Male , Asthenozoospermia/genetics , Asthenozoospermia/metabolism , Computational Biology/methods , Biomarkers/metabolism , Gene Expression Profiling , MicroRNAs/genetics , MicroRNAs/metabolism , Gene Ontology , Signal Transduction/genetics , Spermatozoa/metabolism
18.
Genes (Basel) ; 15(5)2024 05 08.
Article in English | MEDLINE | ID: mdl-38790229

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.


Subject(s)
Sperm Motility , Spermatozoa , Humans , Male , Spermatozoa/metabolism , Spermatozoa/pathology , Sperm Motility/genetics , Asthenozoospermia/genetics , Asthenozoospermia/pathology , Infertility, Male/genetics , Infertility, Male/pathology , Teratozoospermia/genetics , Teratozoospermia/pathology , DNA, Mitochondrial/genetics , Genetic Testing
19.
Front Endocrinol (Lausanne) ; 15: 1377780, 2024.
Article in English | MEDLINE | ID: mdl-38745955

ABSTRACT

Objective: Multiple morphological abnormalities of the sperm flagella (MMAF) is characterized by abnormal flagellar phenotypes, which is a particular kind of asthenoteratozoospermia. Previous studies have reported a comparable intracytoplasmic sperm injection (ICSI) outcome in terms of fertilization rate and clinical pregnancy rate in patients with MMAF compared with those with no MMAF; however, others have conflicting opinions. Assisted reproductive technology (ART) outcomes in individuals with MMAF are still controversial and open to debate. Methods: A total of 38 patients with MMAF treated at an academic reproductive center between January 2014 and July 2022 were evaluated in the current retrospective cohort study and followed up until January 2023. Propensity score matching was used to adjust for the baseline clinical characteristics of the patients and to create a comparable control group. The genetic pathogenesis of MMAF was confirmed by whole exome sequencing. The main outcomes were the embryo developmental potential, the cumulative pregnancy rate (CLPR), and the cumulative live birth rate (CLBR). Results: Pathogenic variants in known genes of DNAH1, DNAH11, CFAP43, FSIP2, and SPEF2 were identified in patients with MMAF. Laboratory outcomes, including the fertilization rate, 2PN cleavage rate, blastocyst formation rate, and available blastocyst rate, followed a trend of decline in the MMAF group (p < 0.05). Moreover, according to the embryo transfer times and complete cycles, the CLPR in the cohort of MMAF was lower compared with the oligoasthenospermia pool (p = 0.033 and p = 0.020, respectively), while no statistical differences were observed in the neonatal outcomes. Conclusion: The current study presented decreased embryo developmental potential and compromised clinical outcomes in the MMAF cohort. These findings may provide clinicians with evidence to support genetic counseling and clinical guidance in specific patients with MMAF.


Subject(s)
Embryonic Development , Pregnancy Rate , Sperm Injections, Intracytoplasmic , Sperm Tail , Humans , Male , Female , Pregnancy , Adult , Retrospective Studies , Sperm Tail/pathology , Embryonic Development/physiology , Asthenozoospermia/genetics , Asthenozoospermia/pathology , Infertility, Male/genetics , Infertility, Male/pathology , Spermatozoa/pathology
20.
J Hum Genet ; 69(8): 401-409, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38769386

ABSTRACT

Human infertility affects 10-15% of couples. Asthenozoospermia accounts for 18% of men with infertility and is a common male infertility phenotype. The nexin-dynein regulatory complex (N-DRC) is a large protein complex in the sperm flagellum that connects adjacent doublets of microtubules. Defects in the N-DRC can disrupt cilia/flagellum movement, resulting in primary ciliary dyskinesia and male infertility. Using whole-exome sequencing, we identified a pathological homozygous variant of the dynein regulatory complex subunit 3 (DRC3) gene, which expresses leucine-rich repeat-containing protein 48, a component of the N-DRC, in a patient with asthenozoospermia. The variant ENST00000313838.12: c.644dup (p. Glu216GlyfsTer36) causes premature translational arrest of DRC3, resulting in a dysfunctional DRC3 protein. The patient's semen count, color, and pH were normal according to the reference values of the World Health Organization guidelines; however, sperm motility and progressive motility were reduced. DRC3 protein was not detected in the patient's sperm and the ultrastructure of the patient's sperm flagella was destroyed. More importantly, the DRC3 variant reduced its interaction with other components of the N-DRC, including dynein regulatory complex subunits 1, 2, 4, 5, 7, and 8. Our data not only revealed the essential biological functions of DRC3 in sperm flagellum movement and structure but also provided a new basis for the clinical genetic diagnosis of male infertility.


Subject(s)
Asthenozoospermia , Homozygote , Infertility, Male , Humans , Male , Asthenozoospermia/genetics , Asthenozoospermia/pathology , Infertility, Male/genetics , Infertility, Male/pathology , Sperm Motility/genetics , Adult , Spermatozoa/metabolism , Spermatozoa/pathology , Exome Sequencing , Sperm Tail/metabolism , Sperm Tail/pathology , Dyneins/genetics , Dyneins/metabolism , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL