Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Front Endocrinol (Lausanne) ; 12: 761249, 2021.
Article in English | MEDLINE | ID: mdl-34721307

ABSTRACT

To achieve spermatogenesis in vitro, one of the most challenging processes to mimic is meiosis. Meiotic problems, like incomplete synapsis of the homologous chromosomes, or impaired homologous recombination, can cause failure of crossover formation and subsequent chromosome nondisjunction, eventually leading to aneuploid sperm. These meiotic events are therefore strictly monitored by meiotic checkpoints that initiate apoptosis of aberrant spermatocytes and lead to spermatogenic arrest. However, we recently found that, in vitro derived meiotic cells proceeded to the first meiotic division (MI) stage, despite displaying incomplete chromosome synapsis, no discernible XY-body and lack of crossover formation. We therefore optimized our in vitro culture system of meiosis from male germline stem cells (mGSCs) in order to achieve full chromosome synapsis, XY-body formation and meiotic crossovers. In comparison to previous culture system, the in vitro-generated spermatocytes were transferred after meiotic initiation to a second culture dish. This dish already contained a freshly plated monolayer of proliferatively inactivated immortalized Sertoli cells supporting undifferentiated mGSCs. In this way we aimed to simulate the multiple layers of germ cell types that support spermatogenesis in vivo in the testis. We found that in this optimized culture system, although independent of the undifferentiated mGSCs, meiotic chromosome synapsis was complete and XY body appeared normal. However, meiotic recombination still occurred insufficiently and only few meiotic crossovers were formed, leading to MI-spermatocytes displaying univalent chromosomes (paired sister chromatids). Therefore, considering that meiotic checkpoints are not necessarily fully functional in vitro, meiotic crossover formation should be closely monitored when mimicking gametogenesis in vitro to prevent generation of aneuploid gametes.


Subject(s)
Chromosome Pairing/physiology , Chromosomes/physiology , Meiosis/physiology , Aneuploidy , Animals , Azoospermia/congenital , Azoospermia/physiopathology , Cell Differentiation/physiology , Cell Line , Cell Proliferation/physiology , Male , Mice , Mice, Inbred DBA , Sertoli Cells/physiology , Spermatocytes/physiology , Spermatogenesis/physiology , Spermatozoa/physiology , Testis/physiology
2.
Funct Integr Genomics ; 21(5-6): 665-678, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34626308

ABSTRACT

Cattleyaks are the crossbred offspring between cattle and yaks, exhibiting the prominent adaptability to the harsh environment as yaks and much higher growth performances than yaks around Qinghai-Tibet plateau. Unfortunately, cattleyak cannot be effectively used in yak breeding due to its male infertility resulted from spermatogenic arrest. In this study, we performed RNA sequencing (RNA-seq) and bioinformatics analysis to determine the expression profiles of long noncoding RNA (lncRNA) from cattleyak and yak testis. A total of 604 differentially expressed (DE) lncRNAs (135 upregulated and 469 downregulated) were identified in cattleyak with respect to yak. Through gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, we identified several DE lncRNAs regulating the mitotic cell cycle processes by targeting the genes significantly associated with the mitotic cell cycle checkpoint and DNA damage checkpoint term and also significantly involved in p53 signaling pathway, mismatch repair and homologous recombination pathway (P < 0.05). The reverse transcription PCR (RT-PCR) and quantitative Real-Time PCR (qRT-PCR) analysis of the randomly selected fourteen DE lncRNAs and the seven target genes validated the RNA-seq data and their true expressions during spermatogenesis in vivo. Molecular cloning and sequencing indicated that the testis lncRNAs NONBTAT012170 and NONBTAT010258 presented higher similarity among different cattleyak and yak individuals. The downregulation of these target genes in cattleyak contributed to the abnormal DNA replication and spermatogenic arrest during the S phase of mitotic cell cycle. This study provided a novel insight into lncRNA expression profile changes associated with spermatogenic arrest of cattleyak.


Subject(s)
Azoospermia/congenital , Cattle/genetics , RNA, Long Noncoding/genetics , Testis/metabolism , Transcriptome , Animals , Azoospermia/genetics , Azoospermia/veterinary , Gene Expression Profiling , Male
3.
Gene ; 795: 145793, 2021 Aug 30.
Article in English | MEDLINE | ID: mdl-34175398

ABSTRACT

The current study assessed the cross-link between mitochondria-related apoptosis and cell cycle machinery systems during ischemia and reperfusion in a rat model of testicular torsion and detorsion. The Wistar male rats were divided into control, 1 h, 2 h, 4 h and 8 h testicular torsion-induced, and 1 h, 2 h, 4 h and 8 h testicular detorsion-induced groups. The Johnson's score was analyzed. The mRNA and protein contents of Bcl-2, Bax, Caspase-3, Cyclin D1, Cdk4, P21 and P53 were investigated by sqRT-PCR and immunohistochemical staining, respectively. The apoptosis index was analyzed by TUNEL staining. The mRNA levels of bax, p53, p21 and cyclin D1 were increased, and the mRNA levels of bcl-2 and cdk4 were decreased in torsion and reperfusion-induced groups, time-dependently. The caspase-3 mRNA was increased in torsion-induced and diminished in detorsion-induced groups. A time-dependent reduction in Bcl-2+, Caspase-3+, Cyclin D1+, Cdk4+ and P53+ and increment in P21+ cells distribution per mm2 of tissue were revealed after torsion and detorsion. The apoptosis index was increased after torsion and decreased after detorsion. In conclusion, torsion-induced severe DNA damage stimulates the cyclin D1, p53 and p21 mRNA expression while more than 8 h is needed to reveal them as protein content in testicular tissue. About detorsion, decreased Cyclin D1 and Cdk4 proteins and the P53-induced transcriptional effect on p21 expression, stimulates the p21 bind to cdk4 and consequent failure in Cyclin D1/Cdk4 complex formation. This situation in association with apoptotic genes results in spermatogenesis failure.


Subject(s)
Apoptosis/genetics , Azoospermia/congenital , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/genetics , Mitochondria/genetics , Spermatic Cord Torsion/genetics , Testis/blood supply , Animals , Azoospermia/etiology , Azoospermia/genetics , Disease Models, Animal , Gene Expression , Male , Rats , Rats, Wistar , Reperfusion Injury/genetics , Sperm Count , Spermatic Cord Torsion/complications , Spermatic Cord Torsion/pathology , Testis/metabolism , Testis/pathology
4.
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 human 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
5.
Fertil Steril ; 115(5): 1197-1211, 2021 05.
Article in English | MEDLINE | ID: mdl-33602558

ABSTRACT

OBJECTIVE: To investigate microRNA (miRNA) expression profiles in the seminal plasma of nonobstructive azoospermia (NOA) patients with different histopathologic patterns and evaluate potential noninvasive diagnostic biomarkers of NOA. DESIGN: Sequencing and validation using quantitative reverse transcription polymerase chain reaction (qRT-PCR). SETTING: Reproductive center and research institute. PATIENT(S): Thirteen patients with NOA (7 Sertoli cell-only syndrome [SCOS] and 6 hypospermatogenesis to spermatogenesis arrest [SA]) and 7 normal fertile controls for sequencing, six samples per group for validation; 54 patients with NOA (27 SCOS and 27 SA) and 19 normal fertile controls for large-sample qRT-PCR analysis. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): MicroRNA expression profiles in the seminal plasma of patients with NOA with different histopathologic patterns were assessed using high-throughput sequencing and validated using qRT-PCR. RESULT(S): There were 78 overexpressed and 132 underexpressed miRNAs in patients with SCOS and 32 up-regulated and 90 down-regulated miRNAs in patients with SA compared with fertile men with normozoospermia. Two down-regulated and one up-regulated miRNA were validated using qRT-PCR, which indicated that the qRT-PCR and sequencing results were basically consistent. Hsa-miR-34c-5p expression was significantly lower in the seminal plasma of patients with NOA than normal fertile controls. The area under the receiver operating characteristic curve(AUC) for hsa-miR-34c-5p was 0.979 and 0.987 in the seminal plasma of patients with SA and patients with SCOS, respectively, compared with normal fertile controls. The AUC was 0.799 for hsa-miR-34c-5p in the seminal plasma between patients with SA and patients with SCOS. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis of differentially expressed miRNA target genes revealed that the Notch signaling pathway was one of the most abundant signaling pathways. The expression of Hes5, an effector of the Notch signaling pathway, was significantly higher in the seminal plasma of patients with NOA than normal fertile controls. CONCLUSION(S): MicroRNA expression profiles in seminal plasma were altered in patients with NOA compared with normal fertile controls. The profiles differed in patients with NOA with different pathologic patterns. We speculate that miR-34c-5p in seminal plasma could be a potential noninvasive biomarker to diagnose patients with NOA and distinguish different pathologic types of NOA. The Notch signaling pathway may be involved in the pathogenesis of NOA.


Subject(s)
Azoospermia , MicroRNAs/genetics , Semen/metabolism , Adult , Azoospermia/classification , Azoospermia/congenital , Azoospermia/genetics , Azoospermia/metabolism , Azoospermia/pathology , Case-Control Studies , Histological Techniques , Humans , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Male , MicroRNAs/metabolism , Semen Analysis , Sertoli Cell-Only Syndrome/genetics , Sertoli Cell-Only Syndrome/metabolism , Sertoli Cell-Only Syndrome/pathology , Testis/metabolism , Testis/pathology , Transcriptome
6.
PLoS One ; 15(2): e0229503, 2020.
Article in English | MEDLINE | ID: mdl-32092127

ABSTRACT

BACKGROUND: Cattleyak are the hybrid offspring between cattle and yak and combine yak hardiness with cattle productivity. Much attempt has been made to examine the mechanisms of male sterility caused by spermatogenic arrest, but yet there is no research systematically and precisely elucidated testis gene expression profiling between cattleyak and yak. METHODS: To explore the higher resolution comparative transcriptome map between the testes of yak and cattleyak, and further analyze the mRNA expression dynamics of spermatogenic arrest in cattleyak. We characterized the comparative transcriptome profile from the testes of yak and cattleyak using high-throughput sequencing. Then we used quantitative analysis to validate several differentially expressed genes (DEGs) in testicular tissue and spermatogenic cells. RESULTS: Testis transcriptome profiling identified 6477 DEGs (2919 upregulated and 3558 downregulated) between cattleyak and yak. Further analysis revealed that the marker genes and apoptosis regulatory genes for undifferentiated spermatogonia were upregulated, while the genes for differentiation maintenance were downregulated in cattleyak. A majority of DEGs associated with mitotic checkpoint, and cell cycle progression were downregulated in cattleyak during spermatogonial mitosis. Furthermore, almost all DEGs related to synaptonemal complex assembly, and meiotic progression presented no sign of expression in cattleyak. Even worse, dozens of genes involved in acrosome formation, and flagellar development were dominantly downregulated in cattleyak. CONCLUSION: DEGs indicated that spermatogenic arrest of cattleyak may originate from the differentiation stage of spermatogonial stem cells and be aggravated during spermatogonial mitosis and spermatocyte meiosis, which contributes to the scarcely presented sperms in cattleyak.


Subject(s)
Azoospermia/congenital , Chimera/genetics , Infertility, Male/genetics , Animals , Azoospermia/genetics , Cattle/genetics , Gene Expression Profiling/methods , Gene Expression Profiling/veterinary , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/veterinary , Male , Meiosis/genetics , Spermatocytes/metabolism , Spermatogenesis/genetics , Spermatogonia/metabolism , Spermatozoa/metabolism , Testis/metabolism , Transcriptome/genetics
7.
Biol Reprod ; 102(4): 975-983, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31916570

ABSTRACT

In mammals, more than 2000 genes are specifically or abundantly expressed in testis, but gene knockout studies revealed several are not individually essential for male fertility. Tesmin (Metallothionein-like 5; Mtl5) was originally reported as a testis-specific transcript that encodes a member of the cysteine-rich motif containing metallothionein family. Later studies showed that Tesmin has two splicing variants and both are specifically expressed in male and female germ cells. Herein, we clarified that the long (Tesmin-L) and short (Tesmin-S) transcript forms start expressing from spermatogonia and the spermatocyte stage, respectively, in testis. Furthermore, while Tesmin-deficient female mice are fertile, male mice are infertile due to arrested spermatogenesis at the pachytene stage. We were able to rescue the infertility with a Tesmin-L transgene, where we concluded that TESMIN-L is critical for meiotic completion in spermatogenesis and indispensable for male fertility.


Subject(s)
Fertility/genetics , Metallothionein/metabolism , Spermatogenesis/genetics , Spermatozoa/metabolism , Testis/metabolism , Animals , Azoospermia/congenital , Azoospermia/genetics , Azoospermia/metabolism , COS Cells , Chlorocebus aethiops , Male , Meiosis/genetics , Metallothionein/genetics , Mice , Mice, Knockout , Spermatocytes/metabolism , Spermatogonia/metabolism
8.
Fertil Steril ; 112(6): 1059-1070.e3, 2019 12.
Article in English | MEDLINE | ID: mdl-31767154

ABSTRACT

OBJECTIVE: To establish which meiotic checkpoints are activated in males with severe spermatogenic impairment to improve phenotypic characterization of meiotic defects. DESIGN: Retrospective observational study. SETTING: University medical center research laboratory and andrology clinic. PATIENT(S): Forty-eight patients with confirmed spermatogenic impairment (Johnsen scores 3-6) and 15 controls (Johnsen score 10). INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Quantitative assessment of immunofluorescent analyses of specific markers to determine meiotic entry, chromosome pairing, progression of DNA double-strand break repair, crossover formation, formation of meiotic metaphases, metaphase arrest, and spermatid formation, resulting in a novel classification of human meiotic arrest types. RESULT(S): Complete metaphase arrest was observed most frequently (27%), and the patients with the highest frequency of apoptotic metaphases also displayed a reduction in crossover number. Incomplete metaphase arrest was observed in 17% of the patients. Only four patients (8%) displayed a failure to complete meiotic chromosome pairing leading to pachytene arrest. Two new types of meiotic arrest were defined: premetaphase and postmetaphase arrest (15% and 13%, respectively). CONCLUSION(S): Meiotic arrest in men occurs most frequently at meiotic metaphase. This arrest can be incomplete, resulting in low numbers of spermatids, and often occurs in association with reduced crossover frequency. The phenotyping approach described here provides mechanistic insights to help identify candidate infertility genes and to assess genotype-phenotype correlations in individual cases.


Subject(s)
Azoospermia/congenital , Metaphase , Spermatogenesis , Spermatozoa/pathology , Testis/pathology , Apoptosis , Azoospermia/pathology , Azoospermia/physiopathology , Chromosome Pairing , DNA Breaks, Double-Stranded , Humans , Male , Pachytene Stage , Retrospective Studies , Testis/physiopathology
9.
Nan Fang Yi Ke Da Xue Xue Bao ; 39(9): 1059-1064, 2019 Sep 30.
Article in Chinese | MEDLINE | ID: mdl-31640955

ABSTRACT

OBJECTIVE: To explore the effect of small interfering RNA (siRNA)-mediated CEP55 gene silencing on the proliferation of mouse spermatogonia. METHODS: Six patients with azoospermia diagnosed to have maturation arrest (3 cases) or normal spermatogenesis (3 cases) based on testicular biopsy between January 1 and December 31, 2017 in our center were examined for differential proteins in the testicular tissue using isobaric tags for relative and absolute quantitation (iTRAQ), and CEP55 was found to differentially expressed between the two groups of patients. We constructed a CEP55 siRNA for transfection in mouse spermatogonia and examined the inhibitory effects on CEP55 expressions using Western blotting and qPCR. The effect of CEP55 gene silencing on the proliferation of mouse spermatogonia was evaluated with CCK8 assay. RESULTS: In the testicular tissues from the 6 patients with azoospermia, iTRAQ combined with LC/MS/MS analysis identified over two hundred differentially expressed proteins, among which CEP55 showed the most significant differential expression between the patients with maturation arrest and those with normal spermatogenesis. The cell transfection experiment showed that compared with the cells transfected with the vehicle or the negative control sequence, the mouse spermatogonia transfected with CEP55 siRNA showed significantly lowered expressions of CEP55 mRNA and protein (P < 0.05) and significantly decreased proliferation rate as shown by CCK8 assay (P < 0.05). CONCLUSIONS: CEP55 may play a key role in spermatogenesis and may serve as a potential therapeutic target for non-obstructive azoospermia with maturation arrest.


Subject(s)
Azoospermia/congenital , Cell Cycle Proteins/genetics , Nuclear Proteins/genetics , Animals , Azoospermia/genetics , Gene Silencing , Humans , Male , Mice , Spermatogenesis , Spermatogonia , Tandem Mass Spectrometry , Transfection
10.
Andrologia ; 51(11): e13436, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31589772

ABSTRACT

Zinner syndrome (ZS) could represent an uncommon cause of male infertility, as result of the ejaculatory duct block, which typically leads to low seminal volume and azoospermia. A 27-year-old Caucasian man reported persistent events of scrotal-perineal pain and dysuria during the past 6 months. The andrological examination showed testicular volume of 10 ml bilaterally. Follicle-stimulating hormone was 32.0 IU/L, luteinising hormone was 16.3 IU/L, total testosterone was 9.0 nmol/L, and 17-beta-oestradiol was 0.12 nmol/L. The semen analysis revealed absolute azoospermia, semen volume of 0.6 ml and semen pH of 7.6. The abdominal contrast-enhanced computed tomography showed (a) left kidney agenesis; (b) an ovaliform hypodense mass of 65 × 46 millimetres with fluid content, which was shaping the bladder and the left paramedian prostatic region, compatible with a left seminal vesicle pseudocyst; and (c) an enlargement of the right seminal vesicle. The patient was diagnosed with ZS, and he was scheduled for robot-assisted laparoscopic left vesiculectomy. Subsequently, testis biopsy was characterised by complete germ cell aplasia. The onset symptomatology is often blurred and difficult to detect. It is important to diagnose and manage early this condition, because a long-lasting seminal tract obstruction could determine an irreversible secretory testicular injury.


Subject(s)
Azoospermia/congenital , Urogenital Abnormalities/complications , Adult , Azoospermia/diagnostic imaging , Humans , Male , Urogenital Abnormalities/diagnostic imaging
11.
Andrologia ; 51(10): e13376, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31373714

ABSTRACT

Androgens play a key role in spermatogenesis, and their functions are mediated by the androgen receptor (AR). Some mutations in the AR gene have the potential to alter the primary structure and function of the protein. The aim of this study was to investigate the AR gene mutations in a cohort of males with idiopathic azoospermia referred to Royan Institute. Fifty-one biopsy samples were obtained for routine clinical purposes from 15 men with hypospermatogenesis (HS), 17 patients with maturation arrest (MA) and 19 patients with Sertoli cell-only syndrome (SCOS). The AR cDNAs were prepared from tissue mRNAs and were sequenced. One synonymous variant and three nonsynonymous protein coding single nucleotide polymorphisms (nsSNPs) were detected. Protein structure prediction demonstrated that the S815I and M746T nonsynonymous variants would affect protein structure and its normal function. Our study suggests that mutations in the AR gene would change or disturb the receptor's normal activity. Although these variations may influence spermatogenesis, it is difficult to say that they lead to a lack of spermatogenesis.


Subject(s)
Azoospermia/congenital , Oligospermia/genetics , Receptors, Androgen/genetics , Sertoli Cell-Only Syndrome/genetics , Spermatogenesis/genetics , Adult , Azoospermia/genetics , Humans , Male , Middle Aged , Mutation , Polymorphism, Single Nucleotide , Protein Domains/genetics
12.
Sci Rep ; 9(1): 9553, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31267012

ABSTRACT

Peroxisomes are involved in the degradation of very long-chain fatty acids (VLCFAs) by ß-oxidation. Besides neurological defects, peroxisomal dysfunction can also lead to testicular abnormalities. However, underlying alterations in the testes due to a peroxisomal defect are not well characterized yet. To maintain all metabolic functions, peroxisomes require an import machinery for the transport of matrix proteins. One component of this translocation machinery is PEX13. Its inactivation leads to a peroxisomal biogenesis defect. We have established a germ cell-specific KO of Pex13 to study the function of peroxisomes during spermatogenesis in mice. Exon 2 of floxed Pex13 was specifically excised in germ cells prior to meiosis by using a transgenic mouse strain carrying a STRA8 inducible Cre recombinase. Germ cell differentiation was interrupted at the round spermatid stage in Pex13 KO mice with formation of multinucleated giant cells (MNCs) and loss of mature spermatids. Due to a different cellular content in the germinal epithelium of Pex13 KO testes compared to control, whole testes biopsies were used for the analyses. Thus, differences in lipid composition and gene expression are only shown for whole testicular tissue but cannot be limited to single cells. Gas chromatography revealed an increase of shorter fatty acids and a decrease of n-6 docosapentaenoic acid (C22:5n-6) and n-3 docosahexaenoic acid (C22:6n-3), the main components of sperm plasma membranes. Representative genes of the metabolite transport and peroxisomal ß-oxidation were strongly down-regulated. In addition, structural components of the blood-testis barrier (BTB) were altered. To conclude, defects in the peroxisomal compartment interfere with normal spermatogenesis.


Subject(s)
Azoospermia/congenital , Germ Cells/metabolism , Peroxisomes/metabolism , Spermatids/metabolism , Spermatogenesis , Animals , Azoospermia/genetics , Azoospermia/metabolism , Fluorescent Antibody Technique , Immunohistochemistry , Infertility , Lipids/chemistry , Male , Mice , Mice, Knockout , Mice, Transgenic , Peroxisomes/ultrastructure , Testis/metabolism , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism
13.
Andrologia ; 51(8): e13301, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31037746

ABSTRACT

Male infertility affects approximately half of couples who have difficulty becoming pregnant, and its prevalence is continuously rising. Many studies have been performed using animal testes to reveal the mechanisms of male infertility, but few studies have investigated human testes due to various limitations. The aim of this study was to investigate the gene expression profile of impaired human testes through a meta-analysis of microarray data sets, which was accomplished by using 178 human testis samples and 7 microarray data sets. Impaired testes were categorised into four pathological phenotypes or the normal phenotype based on their Johnsen score. Then, a meta-analysis was performed to screen out the differentially expressed genes (DEGs) in each phenotype. The DEGs were used in a subsequent bioinformatics analysis. Our results identified several novel hub genes and pathways and suggested that G1 mitotic cell cycle arrest was a remarkable feature in pre-meiotic arrest. Furthermore, fifteen p53-interacting proteins, such as ABL1 and HDAC2, whose roles in spermatogenesis have not been well characterised, were selected from the DEGs through a strict screening procedure.


Subject(s)
Azoospermia/congenital , G1 Phase Cell Cycle Checkpoints/genetics , Spermatogenesis/genetics , Azoospermia/genetics , Azoospermia/pathology , Computational Biology , Databases, Genetic , Datasets as Topic , Gene Expression Profiling/methods , Gene Regulatory Networks/genetics , Histone Deacetylase 2/genetics , Humans , Male , Oligonucleotide Array Sequence Analysis , Protein Interaction Maps/genetics , Proto-Oncogene Proteins c-abl/genetics , Testis/pathology , Tissue Array Analysis
14.
Hum Reprod ; 34(6): 978-988, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31125047

ABSTRACT

STUDY QUESTION: What is the diagnostic potential of next generation sequencing (NGS) based on a 'mouse azoospermia' gene panel in human non-obstructive azoospermia (NOA)? SUMMARY ANSWER: The diagnostic performance of sequencing a gene panel based on genes associated with mouse azoospermia was relatively successful in idiopathic NOA patients and allowed the discovery of two novel genes involved in NOA due to meiotic arrest. WHAT IS KNOWN ALREADY: NOA is a largely heterogeneous clinical entity, which includes different histological pictures. In a large proportion of NOA, the aetiology remains unknown (idiopathic NOA) and yet, unknown genetic factors are likely to play be involved. The mouse is the most broadly used mammalian model for studying human disease because of its usefulness for genetic manipulation and its genetic and physiological similarities to man. Mouse azoospermia models are available in the Mouse Genome Informatics database (MGI: http://www.informatics.jax.org/). STUDY DESIGN, SIZE, DURATION: The first step was to design of a 'mouse azoospermia' gene panel through the consultation of MGI. The second step was NGS analysis of 175 genes in a group of highly selected NOA patients (n = 33). The third step was characterization of the discovered gene defects in human testis tissue, through meiotic studies using surplus testicular biopsy material from the carriers of the RNF212 and STAG3 pathogenic variants. The final step was RNF212 and STAG3 expression analysis in a collection of testis biopsies. PARTICIPANTS/MATERIALS, SETTING, METHODS: From a total of 1300 infertile patients, 33 idiopathic NOA patients were analysed in this study, including 31 unrelated men and 2 brothers from a consanguineous family. The testis histology of the 31 unrelated NOA patients was as follows: 20 Sertoli cell-only syndrome (SCOS), 11 spermatogenic arrest (6 spermatogonial arrest and 5 spermatocytic arrest). The two brothers were affected by spermatocytic arrest. DNA extracted from blood was used for NGS on Illumina NextSeq500 platform. Generated sequence data was filtered for rare and potentially pathogenic variants. Functional studies in surplus testicular tissue from the carriers included the investigation of meiotic entry, XY body formation and metaphases by performing fluorescent immunohistochemical staining and immunocytochemistry. mRNA expression analysis through RT-qPCR of RNF212 and STAG3 was carried out in a collection of testis biopsies with different histology. MAIN RESULTS AND THE ROLE OF CHANCE: Our approach was relatively successful, leading to the genetic diagnosis of one sporadic NOA patient and two NOA brothers. This relatively high diagnostic performance is likely to be related to the stringent patient selection criteria i.e. all known causes of azoospermia were excluded and to the relatively high number of patients with rare testis histology (spermatocytic arrest). All three mutation carriers presented meiotic arrest, leading to the genetic diagnosis of three out of seven cases with this specific testicular phenotype. For the first time, we report biallelic variants in STAG3, in one sporadic patient, and a homozygous RNF212 variant, in the two brothers, as the genetic cause of NOA. Meiotic studies allowed the detection of the functional consequences of the mutations and provided information on the role of STAG3 and RNF212 in human male meiosis. LIMITATIONS, REASONS FOR CAUTION: All genes, with the exception of 5 out of 175, included in the panel cause azoospermia in mice only in the homozygous or hemizygous state. Consequently, apart from the five known dominant genes, heterozygous variants (except compound heterozygosity) in the remaining genes were not taken into consideration as causes of NOA. We identified the genetic cause in approximately half of the patients with spermatocytic arrest. The low number of analysed patients can be considered as a limitation, but it is a very rare testis phenotype. Due to the low frequency of this specific phenotype among infertile men, our finding may be considered of low clinical impact. However, at an individual level, it does have relevance for prognostic purposes prior testicular sperm extraction. WIDER IMPLICATIONS OF THE FINDINGS: Our study represents an additional step towards elucidating the genetic bases of early spermatogenic failure, since we discovered two new genes involved in human male meiotic arrest. We propose the inclusion of RNF212 and STAG3 in a future male infertility diagnostic gene panel. Based on the associated testis phenotype, the identification of pathogenic mutations in these genes also confers a negative predictive value for testicular sperm retrieval. Our meiotic studies provide novel insights into the role of these proteins in human male meiosis. Mutations in STAG3 were first described as a cause of female infertility and ovarian cancer, and Rnf212 knock out in mice leads to male and female infertility. Hence, our results stimulate further research on shared genetic factors causing infertility in both sexes and indicate that genetic counselling should involve not only male but also female relatives of NOA patients. STUDY FUNDING/COMPETING INTEREST(S): This work was funded by the Spanish Ministry of Health Instituto Carlos III-FIS (grant number: FIS/FEDER-PI14/01250; PI17/01822) awarded to CK and AR-E, and by the European Commission, Reproductive Biology Early Research Training (REPROTRAIN, EU-FP7-PEOPLE-2011-ITN289880), awarded to CK, WB, and AE-M. The authors have no conflict of interest.


Subject(s)
Azoospermia/congenital , Cell Cycle Proteins/genetics , Genetic Testing/methods , Ligases/genetics , Meiosis/genetics , Alleles , Animals , Azoospermia/diagnosis , Azoospermia/genetics , Azoospermia/pathology , DNA Mutational Analysis/methods , Databases, Genetic , Datasets as Topic , Disease Models, Animal , Feasibility Studies , High-Throughput Nucleotide Sequencing , Homozygote , Humans , Male , Mice , Mutation , Testis/cytology , Testis/pathology
15.
Tissue Cell ; 58: 24-32, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31133243

ABSTRACT

The proper assessment of male fertility is essential for diagnosing and treating male infertility. Currently, spermiogram and Johnsen testicular biopsy score counts are used to assess male fertility. However, spermiogram is not a suitable option for non-obstructive azoospermia patients, and Johnsen testicular biopsy scores only represent localized and not the overall spermatogenesis. Whole-mount staining was a novel method for evaluating protein expression in the tissue. Thus, we explored its application in human seminiferous tubules. Testicular biopsies from 57 azoospermia patients were categorized as obstructive azoospermia (OA), maturation arrest (MA) and Sertoli-cells only syndrome (SCOS). We performed whole-mount staining of their seminiferous tubules and evaluated the spermatogonial stem cells (SSCs), differentiated spermatogonia (SG), spermatocytes (SPC) and spermatids (SD) with their respective markers (GFRA1, CD117, SYCP3, and PNA) to assess fertility. GFRA1, CD117, SYCP3, and PNA were not expressed in SCOS patients, whereas all of them were detected in OA patients. In MA patients with arrested spermatogenesis at the SPC stage, GFRA1, CD117, and SYCP3, but not PNA were expressed in the seminiferous tubules. In MA patients with arrested spermatogenesis at the spermatogonia stage, only GFRA1 was expressed in the seminiferous tubules. These results were consistent with the Johnsen testicular biopsy score counts except for one patient, where although only Sertoli cells were indicated by the score, SSCs were also detected in the whole-mounts. Collectively, whole-mount staining could be used to analyze the inherent spermatogenesis of seminiferous tubules through staining of germ cells at different stages. It offers a more accurate and promising faster method for assessing male fertility compared with traditional biopsy screening. And it could have potential value for the clinical purpose for male fertility management.


Subject(s)
Azoospermia/congenital , Fertility , Molecular Imaging , Seminiferous Tubules , Spermatogenesis , Spermatogonia , Adult , Azoospermia/diagnostic imaging , Azoospermia/metabolism , Humans , Male , Seminiferous Tubules/diagnostic imaging , Seminiferous Tubules/metabolism , Spermatogonia/metabolism , Spermatogonia/pathology
16.
Sci Rep ; 9(1): 4036, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30858478

ABSTRACT

Testis tissue xenografting complemented with cryopreservation is a feasible technique for fertility preservation in children with malignancy receiving gonadotoxic therapy and for endangered species with high neonatal mortality rate. However, xenografted testis of human and most endangered species are known to undergo spermatogenic arrest. In this study, we xenografted immature rat testis onto immunodeficient male mice to investigate the plausible underlying causes of spermatogenic arrest. Histological analysis of xenografted testes collected 8-wk post-grafting showed incomplete spermatogenesis with pachytene-stage spermatocytes as the most advanced germ cells. Although the levels of serum luteinizing hormone and testosterone were normal in recipient mice, those of follicle stimulating hormone (FSH) were significantly high, and specific receptors of FSH were absent in the xenografts. The xenografts demonstrated dysregulated expression of Sertoli cell-transcriptional regulators (WT1 and SOX9) and secretory proteins (SCF and GDNF). In conclusion, results from our study suggested that an altered hormonal milieu in recipients and dysregulated protein expression in xenografts could be a potential cause of spermatogenic arrest in xenografted immature rat testis. Further stereological analysis of xenografts can demonstrate precise cellular composition of xenografts to decipher interactions between germ and somatic cells to better understand spermatogenic arrest in xenografted testis.


Subject(s)
Azoospermia/congenital , Heterografts/transplantation , Spermatocytes/metabolism , Spermatogenesis/physiology , Testis/transplantation , Animals , Follicle Stimulating Hormone/blood , Luteinizing Hormone/blood , Male , Mice , Mice, Nude , Rats , Rats, Wistar , Receptors, FSH/blood , SOX9 Transcription Factor/metabolism , Spermatocytes/pathology , Testosterone/blood , WT1 Proteins/metabolism
17.
Reprod Biol ; 18(4): 456-466, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30243528

ABSTRACT

For the reason that adult Sertoli cell specific connexin 43 knockout (SCCx43KO) mice show arrested spermatogenesis at spermatogonial level or Sertoli cell only tubules and significantly reduced germ cell (GC) numbers, the aims of the present study were (1) to characterize the remaining GC population and (2) to elucidate possible mechanisms of their fading. Apoptosis was analyzed in both, KO and wild type (WT) male littermates during postnatal development and in adulthood using TUNEL. Although GC numbers were significantly reduced in KO at 2 and 8 days postpartum (dpp) when compared to WT, no differences were found concerning apoptotic incidence between genotypes. From 10 dpp, the substantial GC deficiency became more obvious. However, significantly higher apoptotic GC numbers were seen in WT during this period, possibly related to the first wave of spermatogenesis, a known phenomenon in normal pubertal testes associated with increased apoptosis. Characterization of residual spermatogonia in postnatal to adult KO and WT mice was performed by immunohistochemical reaction against VASA (marker of GCs in general), Lin28 and Fox01 (markers for undifferentiated spermatogonia) and Stra8 (marker for differentiating spermatogonia and early spermatocytes). During puberty, the GC component in SCCx43KO mice consisted likely of undifferentiated spermatogonia, few differentiating spermatogonia and very few early spermatocytes, which seemed to be rapidly cleared by apoptosis. In adult KOs, spermatogenesis was arrested at the level of undifferentiated spermatogonia. Overall, our data indicate that Cx43 gap junctions in SCs influence male GC development and differentiation rather than their survival.


Subject(s)
Connexin 43/metabolism , Sertoli Cells/metabolism , Spermatogenesis/genetics , Spermatogonia/metabolism , Animals , Apoptosis/genetics , Azoospermia/congenital , Azoospermia/genetics , Azoospermia/metabolism , Cell Count , Connexin 43/genetics , Gap Junctions/metabolism , Male , Mice , Mice, Knockout , Testis/metabolism
18.
Andrologia ; 50(10): e13136, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30159913

ABSTRACT

The reported sperm retrieval rate (SRR) in patients with nonobstructive azoospermia (NOA) due to spermatogenic arrest (SA) is highly variable in the literature. This discrepancy could be explained by the heterogeneity of testicular tissues. Surprisingly, even though inhibin B levels reflect directly Sertoli cell function; no studies have evaluated this parameter in SA. We aimed to clarify the morphological and biological profile in 158 men with SA. From the total population, patients whose seminiferous tubules diameter was below 165 µm have higher SRR (46.9% vs. 27.4%, p < 0.05), lower inhibin levels and a higher frequency of nonuniform SA (71.9% vs. 38.7%, p < 0.001). On multivariate analysis, patients with late SA and a history of cryptorchidism were positively associated with successful sperm extraction. Patients with successful SRR and uniform SA exhibited inhibin levels twofold lower than those with failed TESE (45 pg/ml vs. 95 pg/ml, p < 0.05), whereas FSH levels were similar in the two groups. In this study, we showed for the first time that the mean diameter of the seminiferous tubules may be of value in the diagnosis of SA. Our results suggest that inhibin levels could be useful in the management of NOA with SA, along with FSH levels and testicular volume.


Subject(s)
Azoospermia/congenital , Sperm Retrieval , Testis/pathology , Adult , Azoospermia/blood , Azoospermia/complications , Azoospermia/etiology , Azoospermia/pathology , Azoospermia/therapy , Biopsy , Follicle Stimulating Hormone/blood , Humans , Inhibins/blood , Male , Retrospective Studies
19.
Hum Reprod ; 33(6): 1034-1037, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29659827

ABSTRACT

Although meiotic arrest in males is observed in about 25% of azoospermic patients, pure homogeneous arrest in all seminiferous tubules is less frequent, and may be due to mutation of a single gene. However, given the large number of genes involved in meiosis, this gives rises to extensive genetic heterogeneity. Only two genetic abnormalities have been reported on a regular basis: the X-linked exonic TEX11 deletion, and the AZFb microdeletion on the Y chromosome. Other single gene defects were private and found in consanguineous families. Here, we report on a homozygous missense mutation in the gene coding for meiotic double-stranded break formation protein 1 (MEI1; c.C3307T:p.R1103W) observed in two brothers (from a consanguineous Tunisian family) with non-obstructive azoospermia and meiotic arrest. A fertile brother was heterozygous for the mutation. All the queried databases predicted that this mutation is damaging, and it has previously been reported that Mei1 knock-out is associated with meiotic arrest in a murine model. Hence, meiotic arrest in the two brothers was probably caused by an alteration in a gene known to be fundamental for chromosome synapsis.


Subject(s)
Azoospermia/congenital , Consanguinity , Meiosis/genetics , Mutation, Missense/genetics , Proteins/genetics , Azoospermia/genetics , Cell Cycle Proteins , Humans , Male , Pedigree , Siblings , Tunisia , Exome Sequencing
20.
Sci Rep ; 8(1): 592, 2018 01 12.
Article in English | MEDLINE | ID: mdl-29330490

ABSTRACT

Cattleyak are interspecific hybrids between cattle and yak, exhibiting the same prominent adaptability as yak and much higher performances than yak. However, male infertility of cattleyak resulted from spermatogenic arrest has greatly restricted their effective utilization in yak breeding. In past decades, much work has been done to investigate the mechanisms of spermatogenic arrest, but little is known about the differences of the post-transcriptional regulators between cattleyak and yak, which may contribute to the impaired spermatogenesis. MiRNAs, a class of endogenous non-coding small RNA, were revealed to play crucial roles in regulating gene expression at post-transcriptional level. In the present study, we identified 50 differentially expressed (DE) known miRNAs and 11 novel miRNAs by using Illumina HISeq and bioinformatic analysis. A total of 50 putative target sites for the 13 DE known miRNAs and 30 for the 6 DE novel miRNAs were identified, respectively. GO and KEGG analyses were performed to reveal the functions of target genes for DE miRNAs. In addition, RT-qPCR was performed to validate the expression of the DE miRNAs and its targets. The identification of these miRNAs may provide valuable information for a better understanding of spermatogenic arrest in cattleyak.


Subject(s)
Azoospermia/congenital , Cattle Diseases/genetics , Gene Expression Profiling/methods , MicroRNAs/genetics , Testis/chemistry , Animals , Azoospermia/genetics , Azoospermia/veterinary , Cattle , Computational Biology/methods , Gene Expression Profiling/veterinary , Gene Expression Regulation , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/veterinary , Humans , Male , Sequence Analysis, RNA/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...