Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Genet ; 140(8): 1169-1182, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33963445

ABSTRACT

Male infertility impacts millions of couples yet, the etiology of primary infertility remains largely unknown. A critical element of successful spermatogenesis is maintenance of genome integrity. Here, we present a genomic study of spermatogenic failure (SPGF). Our initial analysis (n = 176) did not reveal known gene-candidates but identified a potentially significant single-nucleotide variant (SNV) in X-linked germ-cell nuclear antigen (GCNA). Together with a larger follow-up study (n = 2049), 7 likely clinically relevant GCNA variants were identified. GCNA is critical for genome integrity in male meiosis and knockout models exhibit impaired spermatogenesis and infertility. Single-cell RNA-seq and immunohistochemistry confirm humanĀ GCNA expression from spermatogonia to elongated spermatids. Five identified SNVs were located in key functional regions, including N-terminal SUMO-interacting motif and C-terminal Spartan-like protease domain. Notably, variant p.Ala115ProfsTer7 results in an early frameshift, while Spartan-like domain missense variants p.Ser659Trp and p.Arg664Cys change conserved residues, likely affecting 3D structure. For variants within GCNA's intrinsically disordered region, we performed computational modeling for consensus motifs. Two SNVs were predicted to impact the structure of these consensus motifs. All identified variants have an extremely low minor allele frequency in the general population and 6 of 7 were not detected in > 5000 biological fathers. Considering evidence from animal models, germ-cell-specific expression, 3D modeling, and computational predictions for SNVs, we propose that identified GCNA variants disrupt structure and function of the respective protein domains, ultimately arresting germ-cell division. To our knowledge, this is the first study implicating GCNA, a key genome integrity factor, in human male infertility.


Subject(s)
Azoospermia/congenital , Genes, X-Linked , Infertility, Male/genetics , Mutation , Nuclear Proteins/genetics , Spermatozoa/metabolism , Adult , Animals , Azoospermia/diagnosis , Azoospermia/genetics , Azoospermia/metabolism , Azoospermia/pathology , Base Sequence , Cohort Studies , Follicle Stimulating Hormone/blood , Gene Expression , Genome, Human , Genomic Instability , Humans , Infertility, Male/diagnosis , Infertility, Male/metabolism , Infertility, Male/pathology , Luteinizing Hormone/blood , Male , Meiosis , Models, Molecular , Nuclear Proteins/deficiency , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Spermatogenesis/genetics , Spermatozoa/pathology , Testis/metabolism , Testis/pathology , Testosterone/blood , Exome Sequencing
2.
Gynecol Endocrinol ; 35(4): 294-297, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30449224

ABSTRACT

Androgen insensitivity syndrome (AIS) is a congenital disorder in which a defect in the androgen receptor (AR) gene leads to cellular resistance to androgens. Defects in the AR gene, located on the X chromosome, result in the development of a feminine phenotype in chromosomally male (46, XY) individuals. In this case report, we present a 44 years old patient with complete androgen insensitivity syndrome (CAIS) initially presenting with primary amenorrhea. The patient underwent a full clinical evaluation, revealing hypoplastic vagina and a lack of uterus and ovaries. Hormonal evaluation revealed markedly elevated testosterone, FSH, and LH serum concentrations. Diagnostic imaging, including pelvic MRI, confirmed the presence of two solid masses in the inguinal canals (right 26 Ɨ 13 mm, left 25 Ɨ 15 mm). The patient underwent genetic testing, revealing a 46 XY karyotype and an as of yet unprecedented androgen receptor mutation. The type of the mutation was a single-base exchange - the substitution from cytosine to thymine in chromosome X:66942710 position (referred to human reference genome GRCh37), which has resulted in an amino acid changes from leucine (CTT) to phenyloalanine (TTT) in ligand-binding domain.


Subject(s)
Androgen-Insensitivity Syndrome/genetics , Receptors, Androgen/genetics , Adult , Female , Humans , Male , Mutation, Missense
3.
Int J Mol Sci ; 20(21)2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31671693

ABSTRACT

We analyzed three cases of Complete Androgen Insensitivity Syndrome (CAIS) and report three hitherto undisclosed causes of the disease. RNA-Seq, Real-timePCR, Western immunoblotting, and immunohistochemistry were performed with the aim of characterizing the disease-causing variants. In case No.1, we have identified a novel androgen receptor (AR) mutation (c.840delT) within the first exon in the N-terminal transactivation domain. This thymine deletion resulted in a frameshift and thus introduced a premature stop codon at amino acid 282. In case No.2, we observed a nonsynonymous mutation in the ligand-binding domain (c.2491C>T). Case No.3 did not reveal AR mutation; however, we have found a heterozygous mutation in CYP11A1 gene, which has a role in steroid hormone biosynthesis. Comparative RNA-Seq analysis of CAIS and control revealed 4293 significantly deregulated genes. In patients with CAIS, we observed a significant increase in the expression levels of PLCXD3, TM4SF18, CFI, GPX8, and SFRP4, and a significant decrease in the expression of SPATA16, TSACC, TCP10L, and DPY19L2 genes (more than 10-fold, p < 0.05). Our findings will be helpful in molecular diagnostics of patients with CAIS, as well as the identified genes could be also potential biomarkers for the germ cells differentiation process.


Subject(s)
Androgen-Insensitivity Syndrome/genetics , Cholesterol Side-Chain Cleavage Enzyme/genetics , Mutation , Receptors, Androgen/genetics , Sequence Analysis, DNA/methods , Adolescent , Adult , Androgen-Insensitivity Syndrome/metabolism , Case-Control Studies , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Exons , Female , Frameshift Mutation , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Protein Domains , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Young Adult
4.
Med Sci Monit ; 21: 2494-504, 2015 Aug 24.
Article in English | MEDLINE | ID: mdl-26299772

ABSTRACT

BACKGROUND: The population of healthy Polish men has not been frequently and systematically investigated for fertility status. The aim of this study was to assess the quality of semen in a randomly recruited population of young males. The most important task was to find a relationship between semen parameters, sex hormones, and AR gene polymorphism. MATERIAL AND METHODS: Semen and blood samples from young men from the Poznan (n=113) and Lublin regions (n=89) were collected for semen analysis, assessment of hormonal concentrations, and calculation of the CAG and GGN repeats of the AR gene. RESULTS: Statistical comparisons of the hormones and circulating proteins and the seminological parameters revealed significant differences between the regional groups of males studied. Among the correlations found, we emphasize the positive relationship between inhibin B levels and both the number of spermatozoa per ml (R=0.37; p=0.0001) and the total sperm concentration (R=0.40; p=0.00003). Positive correlations between IGF1 and sperm morphology was also found (R=0.40; p=0.000004). The mean number of CAG repeats in our tested groups was 21.93Ā±2.79, in a range from 16 to 31. The mean number of GGN repeats was 23.2Ā±1.66 and ranged from 16 to 29. Numerous significant correlations were found between CAG or GGN repeats and blood hormones or circulating proteins and semen parameters; however, Spearman's rank correlations revealed rather weak coefficients. CONCLUSIONS: This report attempted to determine the quality of semen samples and sex hormones in a population of Polish young men. The results were found to be similar to data obtained in Scandinavia. The calculated means and range of CAG or GGN repeats of the AR gene in Polish males were similar to West European epidemiological data.


Subject(s)
Gonadal Steroid Hormones/blood , Polymorphism, Genetic , Receptors, Androgen/genetics , Semen Analysis , Adolescent , Adult , Blood Proteins/metabolism , Cohort Studies , Humans , Inhibins/blood , Male , Poland , Sperm Count , Sperm Motility , Trinucleotide Repeats , Young Adult
5.
Reprod Biol ; 24(4): 100949, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39236514

ABSTRACT

The gonadotropin treatment of infertile men may improve spermatogenesis and lead to sperm cell production, however, only a small fraction of treated patients positively responds to such therapy. To identify individual treatment prognostic biomarkers associated with responsiveness to gonadotropins, we compared the gene expression profiles of testicular oligobiopsies from 3 patients with non-obstructive azoospermia (NOA) who positively responded to therapy with a combination of human chorionic gonadotropin and recombinant follicle-stimulating hormone (hCG/rFSH) to those of 3 non-responders. We used Affymetrix Human Gene 1.0 ST microarrays. The results of the microarray evaluation were validated by the qPCR technique while gene variants of the HLA-DQB1 (major histocompatibility complex, class II, DQ beta 1) were subsequently sequenced. In our microarrays, we have identified most significantly 5 transcripts with different expression levels in responders versus non-responders groups. Our interest has been primarily focused on the transcript associated with the HLA-DQB1 gene. Because the expression of this gene was up-regulated in the non-responding patients and only patients with heterozygotic alleles of HLA-DQB1 turned out to be positive to gonadotropin therapy, we suggest that this gene may be a biomarker of potential significance for the gonadotropin treatment of male infertility. We also compared the testicular gene expression profile in one individual before and after gonadotropin treatment. In the re-biopsied sample, we have identified over 600 genes that showed differences in testicular expression; some of these genes are critical for spermiogenesis. Thus, we documented that the applied gonadotropins successfully stimulated the spermatogenetic wave in patients with NOA.

6.
Hum Reprod Open ; 2024(2): hoae020, 2024.
Article in English | MEDLINE | ID: mdl-38650655

ABSTRACT

STUDY QUESTION: Is the Tcte1 mutation causative for male infertility? SUMMARY ANSWER: Our collected data underline the complex and devastating effect of the single-gene mutation on the testicular molecular network, leading to male reproductive failure. WHAT IS KNOWN ALREADY: Recent data have revealed mutations in genes related to axonemal dynein arms as causative for morphology and motility abnormalities in spermatozoa of infertile males, including dysplasia of fibrous sheath (DFS) and multiple morphological abnormalities in the sperm flagella (MMAF). The nexin-dynein regulatory complex (N-DRC) coordinates the dynein arm activity and is built from the DRC1-DRC7 proteins. DRC5 (TCTE1), one of the N-DRC elements, has already been reported as a candidate for abnormal sperm flagella beating; however, only in a restricted manner with no clear explanation of respective observations. STUDY DESIGN SIZE DURATION: Using the CRISPR/Cas9 genome editing technique, a mouse Tcte1 gene knockout line was created on the basis of the C57Bl/6J strain. The mouse reproductive potential, semen characteristics, testicular gene expression levels, sperm ATP, and testis apoptosis level measurements were then assessed, followed by visualization of N-DRC proteins in sperm, and protein modeling in silico. Also, a pilot genomic sequencing study of samples from human infertile males (n = 248) was applied for screening of TCTE1 variants. PARTICIPANTS/MATERIALS SETTING METHODS: To check the reproductive potential of KO mice, adult animals were crossed for delivery of three litters per caged pair, but for no longer than for 6 months, in various combinations of zygosity. All experiments were performed for wild-type (WT, control group), heterozygous Tcte1+/- and homozygous Tcte1-/- male mice. Gross anatomy was performed on testis and epididymis samples, followed by semen analysis. Sequencing of RNA (RNAseq; Illumina) was done for mice testis tissues. STRING interactions were checked for protein-protein interactions, based on changed expression levels of corresponding genes identified in the mouse testis RNAseq experiments. Immunofluorescence in situ staining was performed to detect the N-DRC complex proteins: Tcte1 (Drc5), Drc7, Fbxl13 (Drc6), and Eps8l1 (Drc3) in mouse spermatozoa. To determine the amount of ATP in spermatozoa, the luminescence level was measured. In addition, immunofluorescence in situ staining was performed to check the level of apoptosis via caspase 3 visualization on mouse testis samples. DNA from whole blood samples of infertile males (n = 137 with non-obstructive azoospermia or cryptozoospermia, n = 111 samples with a spectrum of oligoasthenoteratozoospermia, including n = 47 with asthenozoospermia) was extracted to perform genomic sequencing (WGS, WES, or Sanger). Protein prediction modeling of human-identified variants and the exon 3 structure deleted in the mouse knockout was also performed. MAIN RESULTS AND THE ROLE OF CHANCE: No progeny at all was found for the homozygous males which were revealed to have oligoasthenoteratozoospermia, while heterozygous animals were fertile but manifested oligozoospermia, suggesting haploinsufficiency. RNA-sequencing of the testicular tissue showed the influence of Tcte1 mutations on the expression pattern of 21 genes responsible for mitochondrial ATP processing or linked with apoptosis or spermatogenesis. In Tcte1-/- males, the protein was revealed in only residual amounts in the sperm head nucleus and was not transported to the sperm flagella, as were other N-DRC components. Decreased ATP levels (2.4-fold lower) were found in the spermatozoa of homozygous mice, together with disturbed tail:midpiece ratios, leading to abnormal sperm tail beating. Casp3-positive signals (indicating apoptosis) were observed in spermatogonia only, at a similar level in all three mouse genotypes. Mutation screening of human infertile males revealed one novel and five ultra-rare heterogeneous variants (predicted as disease-causing) in 6.05% of the patients studied. Protein prediction modeling of identified variants revealed changes in the protein surface charge potential, leading to disruption in helix flexibility or its dynamics, thus suggesting disrupted interactions of TCTE1 with its binding partners located within the axoneme. LARGE SCALE DATA: All data generated or analyzed during this study are included in this published article and its supplementary information files. RNAseq data are available in the GEO database (https://www.ncbi.nlm.nih.gov/geo/) under the accession number GSE207805. The results described in the publication are based on whole-genome or exome sequencing data which includes sensitive information in the form of patient-specific germline variants. Information regarding such variants must not be shared publicly following European Union legislation, therefore access to raw data that support the findings of this study are available from the corresponding author upon reasonable request. LIMITATIONS REASONS FOR CAUTION: In the study, the in vitro fertilization performance of sperm from homozygous male mice was not checked. WIDER IMPLICATIONS OF THE FINDINGS: This study contains novel and comprehensive data concerning the role of TCTE1 in male infertility. The TCTE1 gene is the next one that should be added to the 'male infertility list' because of its crucial role in spermatogenesis and proper sperm functioning. STUDY FUNDING/COMPETING INTERESTS: This work was supported by National Science Centre in Poland, grants no.: 2015/17/B/NZ2/01157 and 2020/37/B/NZ5/00549 (to M.K.), 2017/26/D/NZ5/00789 (to A.M.), and HD096723, GM127569-03, NIH SAP #4100085736 PA DoH (to A.N.Y.). The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.

7.
Sci Rep ; 13(1): 16563, 2023 10 02.
Article in English | MEDLINE | ID: mdl-37783880

ABSTRACT

Infertility is a problem that affects approximately 15% of couples, and male infertility is responsible for 40-50% of these cases. The cause of male infertility is still poorly diagnosed and treated. One of the prominent causes of male infertility is disturbed spermatogenesis, which can lead to nonobstructive azoospermia (NOA). Whole-genome sequencing (WGS) allows us to identify novel rare variants in potentially NOA-associated genes, among others, in the ESX1 gene. The aim of this study was to activate the ESX1 gene using CRISPRa technology in human germ cells (testicular seminoma cells-TCam-2). Successful activation of the ESX1 gene in TCam-2 cells using the CRISPRa system was achieved, and the expression level of the ESX1 gene was significantly higher in modified TCam-2 cells than in WT cells or the negative control with nontargeted gRNA (p < 0.01). Using RNA-seq, a network of over 50 genes potentially regulated by the ESX1 gene was determined. Finally, 6 genes, NANOG, CXCR4, RPS6KA5, CCND1, PDE1C, and LINC00662, participating in cell proliferation and differentiation were verified in azoospermic patients with and without a mutation in the ESX1 gene as well as in men with normal spermatogenesis, where inverse correlations in the expression levels of the observed genes were noted.


Subject(s)
Azoospermia , Infertility, Male , Humans , Male , Azoospermia/genetics , Azoospermia/metabolism , Infertility, Male/genetics , Spermatogenesis/genetics , Mutation , Testis/metabolism
8.
Front Genet ; 14: 1134849, 2023.
Article in English | MEDLINE | ID: mdl-37234866

ABSTRACT

Introduction: Human spermatogenesis is a highly intricate process that requires the input of thousands of testis-specific genes. Defects in any of them at any stage of the process can have detrimental effects on sperm production and/or viability. In particular, the function of many meiotic proteins encoded by germ cell specific genes is critical for maturation of haploid spermatids and viable spermatozoa, necessary for fertilization, and is also extremely sensitive to even the slightest change in coding DNA. Methods: Here, using whole exome and genome approaches, we identified and reported novel, clinically significant variants in testis-expressed gene 15 (TEX15), in unrelated men with spermatogenic failure (SPGF). Results: TEX15 mediates double strand break repair during meiosis. Recessive loss-of-function (LOF) TEX15 mutations are associated with SPGF in humans and knockout male mice are infertile. We expand earlier reports documenting heterogeneous allelic pathogenic TEX15 variants that cause a range of SPGF phenotypes from oligozoospermia (low sperm) to nonobstructive azoospermia (no sperm) with meiotic arrest and report the prevalence of 0.6% of TEX15 variants in our patient cohort. Among identified possible LOF variants, one homozygous missense substitution c.6835G>A (p.Ala2279Thr) co-segregated with cryptozoospermia in a family with SPGF. Additionally, we observed numerous cases of inferred in trans compound heterozygous variants in TEX15 among unrelated individuals with varying degrees of SPGF. Variants included splice site, insertions/deletions (indels), and missense substitutions, many of which resulted in LOF effects (i.e., frameshift, premature stop, alternative splicing, or potentially altered posttranslational modification sites). Conclusion: In conclusion, we performed an extensive genomic study of familial and sporadic SPGF and identified potentially damaging TEX15 variants in 7 of 1097 individuals of our combined cohorts. We hypothesize that SPGF phenotype severity is dictated by individual TEX15 variant's impact on structure and function. Resultant LOFs likely have deleterious effects on crossover/recombination in meiosis. Our findings support the notion of increased gene variant frequency in SPGF and its genetic and allelic heterogeneity as it relates to complex disease such as male infertility.

9.
Kardiol Pol ; 80(7-8): 813-824, 2022.
Article in English | MEDLINE | ID: mdl-35554929

ABSTRACT

BACKGROUND: The application of a circulating miR-195 inhibitor could be a helping factor in the in vitro model of human skeletal muscle-derived stem/progenitor cells (SkMDS/PCs). Previously, microRNA-195 (miR-195) expression has been reported to be a negative factor for myogenesis. AIMS: The study aimed to obtain anti-apoptotic and anti-aging effects in in vitro cultured myoblasts and to improve their ability to form myotubes by suppressing miR-195 expression. METHODS: Human wild-type (WT) SkMDS/PC cells incubated with control (nonspecific) miRNA inhibitor and miR-195-inhibited SkMDS/PCs were studied. Functional assays (myotube formation and cell aging), antioxidant, and myogenic gene expression analyses were performed at two time points, at the seventh and eleventh cell passages. RESULTS: Myotube formation was found to be almost 2-fold higher in the miR-195-inhibited SkMDS/PCs population (P < 0.05) compared to WT cells. miR-195 inhibition did not appear to affect cell aging or rejuvenate human SkMDS/PCs. Antioxidant (SOD3 and FOXO) gene expression was augmented in the miR-195-inhibited SkMDS/PCs population, but no positive effect on the remaining antioxidant genes (SOD1, SOD2, and catalase) was observed. A significant increase in MyoD gene expression with a concomitant decrease in MyoG (P < 0.05) was further documented in miR-195- -inhibited SkMDS/PCs compared to WT cells (the eleventh cell passage). CONCLUSIONS: The performed studies may lead to the preconditioning of myogenic stem cells to extend their potential for pro-regenerative activity. The miR-195 inhibitor may serve as a conditioning factor augmenting selective antioxidant gene expression and proliferative potential of SkMDS/PCs, but it does not have an impact on cell aging and/ or apoptosis.


Subject(s)
Antioxidants , MicroRNAs , Cell Differentiation/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal , Myoblasts/metabolism , Stem Cells/metabolism
10.
Reprod Biol ; 22(1): 100597, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34959194

ABSTRACT

In mammals, testicular Heat shock-related 70 kDa protein 2 (HSPA2) is a chaperon strictly linked to spermatogenesis status, whereas its presence in spermatozoa ensures successful oocyte fertilization. However, there is little information on this protein in seminal plasma in infertile males. Based on our previous two independent studies, we have selected HSPA2 to evaluate this seminal plasma protein is a potential biomarker of correct spermatogenesis. Using immunoblotting and mass spectrometry (MS) we have screened human seminal plasma samples for the presence of HSPA2. Samples were obtained from individuals with normozoospermia, cryptozoospermia, non-obstructive and obstructive azoospermia. Our results showed a lack of HSPA2 in seminal plasma in all azoospermic males however, in cryptozoospermia the results were extremely diversified. Additionally, the application of 2-dimensional gel electrophoresis (2-DE) indicated the presence of additional protein isoforms suggesting possible mechanisms underlying the male infertility. Our findings suggest seminal plasma HSPA2 protein as a possible biomarker not only of spermatogenesis status, especially in cryptozoospermic males, but also as a biomarker predicting the success of reproductive treatment including assisted reproductive techniques (ART).


Subject(s)
Semen , Spermatogenesis , Animals , Biomarkers/metabolism , Blood Proteins , HSP70 Heat-Shock Proteins/metabolism , Humans , Male , Semen/metabolism , Spermatozoa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL