Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 519
Filtrar
1.
Arch Ital Urol Androl ; 96(3): 12682, 2024 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-39356016

RESUMO

OBJECTIVES: To determine if histological evaluation of the vasa is useful when post-vasectomy semen analysis (PVSA) compliance is low and to determine whether compliance could be predicted. METHODS: A retrospective evaluation of patients undergoing vasectomy between 2018 and 2022 was undertaken. A comparison of the PVSA between three vasa histological categorisations was made: complete divisions, incomplete division(s), absent vas(a). A multivariate model was constructed to predict PVSA compliance. RESULTS: From 388 patients, 191 (49.2%) undertook PVSA. Four patients had a revision of vasectomy. On 3 occasions this was due to the histology findings and once from semen analysis with normal histology. There was no significant difference in the number of azoospermic samples (95.4% vs 91.2%, ns), of samples with presence of Rare Non-Motile Sperm (RNMS) (2.6% vs 8.8%, ns) and those with sperm present (2.0 vs 0%, ns), between patients with complete division of the vasa on both sides and those with incomplete division on one side respectively. There was no difference in patient characteristics between those who complied with PVSA and those who did not. CONCLUSIONS: This paper suggests that there is a role for histological evaluation of the vasa when PVSA compliance is poor. Incompletely divided vasa on histology are not associated with an adverse PVSA.


Assuntos
Análise do Sêmen , Vasectomia , Humanos , Masculino , Estudos Retrospectivos , Adulto , Análise do Sêmen/métodos , Pessoa de Meia-Idade , Ducto Deferente/patologia , Azoospermia/patologia
2.
Mol Genet Genomics ; 299(1): 84, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39223386

RESUMO

Male infertility is a complex multifactorial reproductive disorder with highly heterogeneous phenotypic presentations. Azoospermia is a medically non-manageable cause of male infertility affecting ∼1% of men. Precise etiology of azoospermia is not known in approximately three-fourth of the cases. To explore the genetic basis of azoospermia, we performed whole exome sequencing in two non-obstructive azoospermia affected siblings from a consanguineous Pakistani family. Bioinformatic filtering and segregation analysis of whole exome sequencing data resulted in the identification of a rare homozygous missense variant (c.962G>C, p. Arg321Thr) in YTHDC2, segregating with disease in the family. Structural analysis of the missense variant identified in our study and two previously reported functionally characterized missense changes (p. Glu332Gln and p. His327Arg) in mice showed that all these three variants may affect Mg2+ binding ability and helicase activity of YTHDC2. Collectively, our genetic analyses and experimental observations revealed that missense variant of YTHDC2 can induce azoospermia in humans. These findings indicate the important role of YTHDC2 deficiency for azoospermia and will provide important guidance for genetic counseling of male infertility.


Assuntos
Azoospermia , Sequenciamento do Exoma , Homozigoto , Mutação de Sentido Incorreto , Linhagem , Irmãos , Adulto , Animais , Humanos , Masculino , Camundongos , Azoospermia/genética , Azoospermia/patologia , Consanguinidade , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Paquistão , RNA Helicases/genética
3.
J Genet ; 1032024.
Artigo em Inglês | MEDLINE | ID: mdl-39223739

RESUMO

The most severe type of male infertility is nonobstructive azoospermia (NOA), where there is no sperm in the ejaculate due to failure of spermatogenesis. The predictable frequency of NOA in the general population is one in 100 men. Genetic studies have recognized dozens of NOA genes. Most NOA aetiologies remain idiopathic. Monogenic mutations can be a reason for a part of idiopathic NOA cases. To address this, we studied the pedigree of a consanguineous family with three NOAs by a family-based exome sequencing. Our goal was to pinpoint the genetic variants responsible for idiopathic NOA to aid future clinical genetic diagnostics and treatment strategies. Bioinformatics analysis followed by Sanger sequencing revealed that NOA patients were homozygous for a rare novel missense variant in PNLDC1(NM_173516:exon9:c.710G>A;p.Gly237Asp). In silico, single-cell RNA sequencing data analysis and protein modelling demonstrated that PNLDC1, Gly237Asp resided in the conserved region of the CAF1 domain which could lead to local instability in the structure and alteration of protein phosphorylation site. We conclude that the novel missense PNLDC1 variant may affect meiosis and spermatogenesis, leading to NOA and the genetic cause of this idiopathic NOA family. Our result helps genetic counselling for idiopathic NOA cases and provides the occasion for more efficient diagnosis in the clinical setting.


Assuntos
Azoospermia , Mutação de Sentido Incorreto , Linhagem , Humanos , Masculino , Azoospermia/genética , Azoospermia/patologia , Sequenciamento do Exoma , Adulto , Espermatogênese/genética
4.
PLoS One ; 19(9): e0309974, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39231187

RESUMO

Azoospermia (the complete absence of spermatozoa in the semen) is a common cause of male infertility. The etiology of azoospermia is poorly understood. Whole-genome analysis of azoospermic men has identified a number of candidate genes, such as the X-linked testis-expressed 11 (TEX11) gene. Using a comparative genomic hybridization array, an exonic deletion (exons 10-12) of TEX11 had previously been identified in two non-apparent azoospermic patients. However, the putative impact of this genetic alteration on spermatogenesis and the azoospermia phenotype had not been validated functionally. We therefore used a CRISPR/Cas9 system to generate a mouse model (Tex11Ex9-11del/Y) with a partial TEX11 deletion that mimicked the human mutation. Surprisingly, the mutant male Tex11Ex9-11del/Y mice were fertile. The sperm concentration, motility, and morphology were normal. Similarly, the mutant mouse line's testis transcriptome was normal, and the expression of spermatogenesis genes was not altered. These results suggest that the mouse equivalent of the partial deletion observed in two infertile male with azoospermia has no impact on spermatogenesis or fertility in mice, at least of a FVB/N genetic background and until 10 months of age. Mimicking a human mutation does not necessarily lead to the same human phenotype in mice, highlighting significant differences species.


Assuntos
Azoospermia , Meiose , Espermatogênese , Animais , Masculino , Camundongos , Espermatogênese/genética , Meiose/genética , Azoospermia/genética , Azoospermia/patologia , Infertilidade Masculina/genética , Deleção de Sequência , Humanos , Testículo/metabolismo , Testículo/patologia , Sistemas CRISPR-Cas
5.
Int J Mol Sci ; 25(15)2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39125928

RESUMO

Azoospermia, the absence of sperm cells in semen, affects around 15% of infertile males. Sertoli cell-only syndrome (SCOS) is the most common pathological lesion in the background of non-obstructive azoospermia and is characterised by the complete absence of germinal epithelium, with Sertoli cells exclusively present in the seminiferous tubules. Studies have shown a correlation between successful spermatogenesis and male fertility with lipid composition of spermatozoa, semen, seminal plasma or testis. The aim of this research was to discover the correlation between the Johnsen scoring system and phospholipid expressions in testicular cryosections of SCOS patients. MALDI imaging mass spectrometry is used to determine spatial distributions of molecular species, such as phospholipids. Phosphatidylcholines (PCs), phosphatidylethanolamines (PEs) and sphingomyelins (SMs) are the most abundant phospholipids in mammalian cells and testis. SMs, the structural components of plasma membranes, are crucial for spermatogenesis and sperm function. Plasmalogens, are unique PCs in testis with strong antioxidative properties. This study, using imaging mass spectrometry, demonstrates the local distribution of phospholipids, particularly SMs, PCs, plasmalogens and PEs in human testicular samples with SCOS for the first time. This study found a strong relationship between the Johnsen scoring system and phospholipid expression levels in human testicular tissues. Future findings could enable routine diagnostic techniques during microTESE procedures for successful sperm extraction.


Assuntos
Síndrome de Células de Sertoli , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Testículo , Masculino , Humanos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Testículo/metabolismo , Testículo/patologia , Síndrome de Células de Sertoli/metabolismo , Síndrome de Células de Sertoli/patologia , Fosfolipídeos/metabolismo , Espermatogênese , Azoospermia/metabolismo , Azoospermia/patologia , Esfingomielinas/metabolismo , Lipídeos/análise , Adulto , Espermatozoides/metabolismo , Espermatozoides/patologia
6.
Reprod Biol Endocrinol ; 22(1): 113, 2024 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-39210375

RESUMO

BACKGROUND: Non-obstructive azoospermia (NOA) is the most severe form of male infertility and affects approximately 1% of men worldwide. Fanconi anemia (FA) genes were known for their essential role in DNA repair and growing evidence showed the crucial role of FA pathway in NOA. However, the underlying mechanisms for Fance deficiency lead to a serious deficit and delayed maturation of male germ cells remain unclear. METHODS: We used Fance deficiency mouse model for experiments, and collected testes or epididymides from mice at 8 weeks (8W), 17.5 days post coitum (dpc), and postnatal 11 (P11) to P23. The mice referred to three genotypes: wildtype (Fance +/+), heterozygous (Fance +/-), and homozygous (Fance -/-). Hematoxylin and eosin staining, immunofluorescence staining, and surface spread of spermatocytes were performed to explore the mechanisms for NOA of Fance -/- mice. Each experiment was conducted with a minimum of three biological replicates and Kruskal-Wallis with Dunn's correction was used for statistical analysis. RESULTS: In the present study, we found that the adult male Fance -/- mice exhibited massive germ cell loss in seminiferous tubules and dramatically decreased sperms in epididymides. During the embryonic period, the number of Fance -/- prospermatogonia decreased significantly, without impacts on the proliferation (Ki-67, PCNA) and apoptosis (cleaved PARP, cleaved Caspase 3) status. The DNA double-strand breaks (γH2AX) increased at the cellular level of Fance -/- prospermatogonia, potentially associated with the increased nonhomologous end joining (53BP1) and decreased homologous recombination (RAD51) activity. Besides, Fance deficiency impeded the progression of meiotic prophase I of spermatocytes. The mechanisms entailed the reduced recruitment of the DNA end resection protein RPA2 at leptotene and recombinases RAD51 and DMC1 at zygotene. It also involved impaired removal of RPA2 at zygotene and FANCD2 foci at pachytene. And the accelerated initial formation of crossover at early pachytene, which is indicated by MLH1. CONCLUSIONS: Fance deficiency caused massive male germ cell loss involved in the imbalance of DNA damage repair in prospermatogonia and altered dynamics of proteins in homologous recombination, DNA end resection, and crossover, providing new insights into the etiology and molecular basis of NOA.


Assuntos
Azoospermia , Dano ao DNA , Reparo do DNA , Camundongos Knockout , Espermatócitos , Espermatogênese , Masculino , Animais , Espermatócitos/metabolismo , Reparo do DNA/genética , Camundongos , Azoospermia/genética , Azoospermia/patologia , Azoospermia/metabolismo , Dano ao DNA/genética , Espermatogênese/genética , Testículo/metabolismo , Testículo/patologia , Camundongos Endogâmicos C57BL
7.
Clin Genet ; 106(5): 625-631, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38956960

RESUMO

Non-obstructive azoospermia (NOA) resulting from primary spermatogenic failure represents one of the most severe forms of male infertility, largely because therapeutic options are very limited. Beyond their diagnostic value, genetic tests for NOA also hold prognostic potential. Specifically, genetic diagnosis enables the establishment of genotype-testicular phenotype correlations, which, in some cases, provide a negative predictive value for testicular sperm extraction (TESE), thereby preventing unnecessary surgical procedures. In this study, we employed whole-genome sequencing (WGS) to investigate two generations of an Iranian family with NOA and identified a homozygous splicing variant in TDRKH (NM_001083965.2: c.562-2A>T). TDRKH encodes a conserved mitochondrial membrane-anchored factor essential for piRNA biogenesis in germ cells. In Tdrkh knockout mice, de-repression of retrotransposons in germ cells leads to spermatogenic arrest and male infertility. Previously, our team reported TDRKH involvement in human NOA cases through the investigation of a North African cohort. This current study marks the second report of TDRKH's role in NOA and human male infertility, underscoring the significance of the piRNA pathway in spermatogenesis. Furthermore, across both studies, we demonstrated that men carrying TDRKH variants, similar to knockout mice, exhibit complete spermatogenic arrest, correlating with failed testicular sperm retrieval.


Assuntos
Azoospermia , Homozigoto , Linhagem , Sequenciamento Completo do Genoma , Masculino , Humanos , Azoospermia/genética , Azoospermia/patologia , Irã (Geográfico) , Espermatogênese/genética , Adulto , Splicing de RNA/genética , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Testículo/patologia , Testículo/metabolismo , RNA Interferente Pequeno/genética
8.
Reproduction ; 168(3)2024 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-38995815

RESUMO

In brief: Since available therapeutic approaches for chemotherapy-induced non-obstructive azoospermia (NOA) patients are not enough efficient, an urgent need for treatment alternatives is felt. This study shows that adipose tissue-derived mesenchymal stem cells-derived exosome (AD-Exo) treatment is more effective in ameliorating busulfan-induced NOA rat models compared to platelet-rich plasma (PRP). Abstract: Patients with non-obstructive azoospermia (NOA) are unable to have their children. Therefore, there is an urgent need for additional treatment alternatives for these patients. Recently, novel treatments based on the exosomes derived from mesenchymal stem cells (MSCs) as the agents responsible for exerting the paracrine effects and consequently biological functions of MSCs are proposed. Besides, platelet-rich plasma (PRP) as a significant blood byproduct has been therapeutically applied in several male infertility studies. In this study, we compared the effects of PRP and exosome treatment on spermatogenesis restoration in NOA rat models. Exosomes and PRP were isolated from the adipose tissue-derived MSCs (AD-MSCs) collected from conditioned medium and peripheral blood of human volunteers, respectively. Non-obstructive azoospermia (NOA) induction was done through two doses of busulfan at a 21-day interval. Thirty-five days after NOA induction, intratesticular injection of AD-MSCs-derived exosome (AD-Exo), PRP, and PBS was performed. The control group did not receive any treatment. Two months later, the rats were euthanized for further analysis. Our results revealed that both AD-Exo and PRP treatments improved the size and weight of testis, modulated the expression level of Dazl, Ddx4, Stra8, Pwil1, and Ccna1, and ameliorated the serum level of LDH, SOD, and GR enzymes in NOA rats. Moreover, the AD-Exo group showed improved testosterone, GPx, MAD, and CAT serum levels, sperm motility, and protein levels of DAZL and DDX4. This investigation verified the more efficient effects of AD-Exo treatment in comparison to PRP in ameliorating busulfan-induced NOA rat models.


Assuntos
Azoospermia , Bussulfano , Modelos Animais de Doenças , Exossomos , Células-Tronco Mesenquimais , Plasma Rico em Plaquetas , Espermatogênese , Masculino , Animais , Exossomos/metabolismo , Exossomos/transplante , Azoospermia/terapia , Azoospermia/patologia , Azoospermia/induzido quimicamente , Espermatogênese/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Ratos , Bussulfano/farmacologia , Plasma Rico em Plaquetas/metabolismo , Humanos , Testículo/metabolismo , Testículo/patologia , Ratos Sprague-Dawley
9.
Fertil Steril ; 122(4): 636-647, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38909671

RESUMO

OBJECTIVE: To explore factors influencing microdissection testicular sperm extraction (micro-TESE) success in hypogonadal men with nonobstructive azoospermia (NOA). DESIGN: A cohort study. SETTING: University-affiliated male reproductive health center. PATIENT(S): A total of 616 consecutive patients with NOA and hypogonadism (total testosterone [T] levels <350 ng/dL) underwent micro-TESE between 2014 and 2021. All patients had no prior sperm retrieval (SR) history. INTERVENTION(S): Patients aged 23-55 years underwent comprehensive clinical, laboratory, and histopathological diagnostic evaluation for NOA and were further categorized into two cohorts on the basis of pre-SR hormonal stimulation. MAIN OUTCOME MEASURE(S): A multivariable logistic regression analysis explored the associations between patient variables and micro-TESE success, defined as the presence of viable spermatozoa in extracted specimens. Adjusted odds ratios (aORs) and 95% confidence intervals (CIs) were computed to assess the relationship between SR success and relevant predictors. Sperm retrieval rates were compared between patients receiving or not hormonal stimulation, and logistic regression analysis evaluated the effect of baseline follicle-stimulating hormone levels (i.e., normogonadotropic vs. hypergonadotropic classes) on SR success. RESULT(S): The overall micro-TESE success rate was 56.6%. Baseline follicle-stimulating hormone levels (aOR, 0.97; 95% CI, 0.94-0.99), pre-SR hormonal stimulation (aOR, 2.54; 95% CI, 1.64-3.93), presence of clinical varicocele (aOR, 0.05; 95% CI, 0.01-0.51), history of previous varicocelectomy (aOR, 2.55; 95% CI, 1.26-5.16), and testicular histopathology were independent predictors of SR success. Among hormone-pretreated patients, pre-micro-TESE T levels and delta T (an absolute increase in T levels from baseline) were associated with SR success. A pre-micro-TESE T level of 418.5 ng/dL (area under the curve value: 0.78) and a delta T of 258 ng/dL (area under the value: 0.76) distinguished patients with positive and negative SR outcomes. Subgroup analysis showed that pre-SR hormonal stimulation yielded a greater benefit for normogonadotropic patients than for those who were hypergonadotropic. CONCLUSION(S): This study underscores the association between clinical factors and micro-TESE success in hypogonadal men with NOA. Although causality is not established, our findings suggest that these patients may benefit from pre-SR interventions, particularly hormonal stimulation and varicocele repair. CLINICAL TRIAL REGISTRATION NUMBER: NCT05110391.


Assuntos
Azoospermia , Hipogonadismo , Microdissecção , Recuperação Espermática , Adulto , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem , Azoospermia/diagnóstico , Azoospermia/patologia , Azoospermia/cirurgia , Azoospermia/sangue , Azoospermia/terapia , Hipogonadismo/diagnóstico , Hipogonadismo/patologia , Hipogonadismo/sangue , Hipogonadismo/cirurgia , Microdissecção/métodos , Estudos Retrospectivos , Testículo/patologia , Testículo/cirurgia , Testosterona/sangue , Resultado do Tratamento
10.
J Assist Reprod Genet ; 41(8): 2079-2098, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38839698

RESUMO

PURPOSE: This study examined SSC proliferation on an epididymosome-enriched decellularized testicular matrix (DTM) hydrogel and spermatogenesis induction in azoospermic mice. METHODS: Epididymosomes were extracted and characterized using SEM and western blotting. After cryopreservation, thawed SSCs were cultured in a hydrogel-based three-dimensional (3D) culture containing 10 ng/mL GDNF or 20 µg/mL epididymosomes. SSCs were assessed using the MTT assay, flow cytometry, and qRT-PCR after two weeks of culture. The isolated SSCs were microinjected into the efferent ducts of busulfan-treated mice. DiI-labeled SSCs were followed, and cell homing was assessed after two weeks. After 8 weeks, the testes were evaluated using morphometric studies and immunohistochemistry. RESULTS: The expression of PLZF, TGF-ß, and miR-10b did not increase statistically significantly in the 3D + GDNF and 3D + epididymosome groups compared to the 3D group. Among the groups, the GDNF-treated group exhibited the highest expression of miR-21 (*P < 0.05). Caspase-3 expression was lower in the epididymosome-treated group than in the other groups (***P < 0.001). Compared to the 3D and negative control groups, the 3D + epididymosomes and 3D + GDNF groups showed an increase in spermatogenic cells. Immunohistochemical results confirmed the growth and differentiation of spermatogonial cells into spermatids in the treatment groups. CONCLUSION: The DTM hydrogel containing 20 µg/mL epididymosomes or 10 ng/mL GDNF is a novel and safe culture system that can support SSC proliferation in vitro to obtain adequate SSCs for transplantation success. It could be a novel therapeutic agent that could recover deregulated SSCs in azoospermic patients.


Assuntos
Azoospermia , Criopreservação , Epididimo , Espermatogênese , Testículo , Animais , Masculino , Camundongos , Azoospermia/patologia , Testículo/crescimento & desenvolvimento , Criopreservação/métodos , Epididimo/metabolismo , Espermatogônias/citologia , Alicerces Teciduais/química , Células-Tronco Germinativas Adultas/metabolismo , Proliferação de Células
11.
J Assist Reprod Genet ; 41(8): 2099-2105, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38878197

RESUMO

PURPOSE: To investigate cryopreserved testicular spermatozoa among patients with azoospermia. METHODS: In this retrospective study spanning from October 1993 to December 2021, we examined men diagnosed with azoospermia who underwent testicular spermatozoa cryopreservation. Data from medical records included utilization and disposal of sperm samples, age at initial cryopreservation. We analyzed the data over 20 years using Kaplan-Meier curves, compared age with the log-rank test, and assessed hazard ratios (HR) with 95% confidence intervals (CI) using Cox regression analysis. RESULTS: A total of 356 patients with a mean age of 32.1 ± 6 were included. Of these, 225 patients utilized thawed testicular sperm for fertility treatments, with 118 patients using all their frozen straws and 107 patients partially using their stored straws. Additionally, 29 patients opted for disposal (six patients partially used their testicular spermatozoa before disposal), resulting in 108 patients who neither used nor disposed of their straws. From a laboratory standpoint, nearly 90% of patients contributed a single testicular sample, which was subsequently divided and cryopreserved as straws, with a median of 4 straws per sample. Notably, in the older age group (> 35 years old), there were a significantly lower usage rate and a higher disposal rate compared to the younger age groups (p < 0.05 for both), corroborated by univariable Cox analysis. CONCLUSIONS: This extensive study unveils unique patterns in the preservation and disposal of testicular spermatozoa among azoospermic patients. Most patients utilize a significant portion of their stored samples, while older patients tend to use their testicular spermatozoa less frequently.


Assuntos
Azoospermia , Criopreservação , Preservação do Sêmen , Espermatozoides , Testículo , Humanos , Azoospermia/patologia , Masculino , Adulto , Espermatozoides/patologia , Testículo/patologia , Estudos Retrospectivos , Preservação da Fertilidade/métodos
12.
J Assist Reprod Genet ; 41(8): 2145-2161, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38902567

RESUMO

PURPOSE: The objective of this study was to discern ferroptosis-related genes (FRGs) linked to non-obstructive azoospermia and investigate the associated molecular mechanisms. METHOD: A dataset related to azoospermia was retrieved from the Gene Expression Omnibus database, and FRGs were sourced from GeneCards. Ferroptosis-related differentially expressed genes (FRDEGs) were discerned. Subsequently, these genes underwent analyses encompassing Gene Ontology and Kyoto Encyclopedia of Genes and Genomes, as well as protein-protein interaction (PPI) networks and assessments of functional similarity. Following the identification of hub genes, an exploration of immune infiltration, single-cell expression, diagnostic utility, and interactions involving hub genes, RNA-binding proteins (RBPs), transcription factors (TFs), microRNAs (miRNAs), and drugs was conducted. RESULTS: A total of 35 differentially expressed FRGs were discerned. These genes demonstrated enrichment in functions and pathways associated with ferroptosis. From the PPI network, eight hub genes were selected. Functional similarity analysis highlighted the potential pivotal roles of HMOX1 and GPX4 in azoospermia. Analysis of immune cell infiltration indicated a significant decrease in activated dendritic cells in the azoospermia group, with notable correlations between hub genes, particularly SAT1 and HMGCR, and immune cell infiltration. Unique expression patterns of hub genes across various cell types in the human testis were observed, with GPX4 prominently enriched in spermatid/sperm. Eight hub genes exhibited robust diagnostic value (AUC > 0.75). Lastly, a comprehensive hub gene-miRNA-TF-RBP-drug network was constructed. CONCLUSION: In summary, our investigation unveiled eight FRDEGs associated with azoospermia, which hold potential as biomarkers for the diagnosis and treatment of azoospermia.


Assuntos
Azoospermia , Biologia Computacional , Ferroptose , Redes Reguladoras de Genes , Mapas de Interação de Proteínas , Humanos , Azoospermia/genética , Azoospermia/patologia , Masculino , Ferroptose/genética , Biologia Computacional/métodos , Mapas de Interação de Proteínas/genética , Redes Reguladoras de Genes/genética , MicroRNAs/genética , Perfilação da Expressão Gênica , Ontologia Genética , Fatores de Transcrição/genética , Regulação da Expressão Gênica/genética
13.
J Assist Reprod Genet ; 41(8): 2163-2171, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38941005

RESUMO

PURPOSE: To investigate the occurrence of idiopathic secondary azoospermia (ISA) in men with oligospermia over time and identify risk factors for ISA in this population. METHODS: This was a retrospective cohort study conducted in a university-affiliated male infertility clinic. A total of 1056 oligospermic men (concentration < 15 million/ml (M/ml) and no azoospermia) with at least two SA done between 2000 and 2019 were included. The primary outcome was the occurrence of ISA by oligospermia severity. RESULTS: In the entire cohort, 31 patients (2.9%) eventually became azoospermic with time. The ≤ 1 M/ml extremely severe oligospermia (ESO) group (283 patients) had significantly higher rates of ISA in each time period compared to the 1-5 M/ml severe oligospermia (SO) (310 patients) and 5-15 M/ml mild oligospermia (MO) (463 patients) groups (p < 0.05 for all comparisons), with rates of 21.1% in the ESO, 4.8% in the SO, and 0% in the MO group (p = 0.02) after 3-5 years, reaching 32% after 5 years in the ESO group compared to no cases in the other two groups (p = 0.006). Parameters shown to predict ISA were initial concentration < 1 M/ml (OR 22.12, p < 0.001) and time interval of > 3 and 5 years (OR 4.83 and 6.84, p = 0.009 and < 0.001, respectively), whereas testosterone levels were negatively associated with ISA (OR 0.88, p = 0.03). CONCLUSIONS: Men with ≤ 1 M/ml, especially those with low testosterone levels, have a dramatically increased chance of becoming azoospermic with time. Therefore, sperm banking should be recommended in these cases. Men with a sperm concentration above 1 M/ml have low chances of becoming azoospermic, even after 3 or more years.


Assuntos
Azoospermia , Oligospermia , Humanos , Masculino , Oligospermia/patologia , Oligospermia/epidemiologia , Azoospermia/patologia , Azoospermia/epidemiologia , Adulto , Estudos Retrospectivos , Contagem de Espermatozoides , Infertilidade Masculina/patologia , Infertilidade Masculina/epidemiologia , Fatores de Risco , Análise do Sêmen , Testosterona/sangue
14.
Front Endocrinol (Lausanne) ; 15: 1393111, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38846492

RESUMO

Non-obstructive azoospermia (NOA) is a disease characterized by spermatogenesis failure and comprises phenotypes such as hypospermatogenesis, mature arrest, and Sertoli cell-only syndrome. Studies have shown that FA cross-linked anemia (FA) pathway is closely related to the occurrence of NOA. There are FA gene mutations in male NOA patients, which cause significant damage to male germ cells. The FA pathway is activated in the presence of DNA interstrand cross-links; the key step in activating this pathway is the mono-ubiquitination of the FANCD2-FANCI complex, and the activation of the FA pathway can repair DNA damage such as DNA double-strand breaks. Therefore, we believe that the FA pathway affects germ cells during DNA damage repair, resulting in minimal or even disappearance of mature sperm in males. This review summarizes the regulatory mechanisms of FA-related genes in male azoospermia, with the aim of providing a theoretical reference for clinical research and exploration of related genes.


Assuntos
Azoospermia , Proteínas de Grupos de Complementação da Anemia de Fanconi , Transdução de Sinais , Animais , Humanos , Masculino , Azoospermia/genética , Azoospermia/metabolismo , Azoospermia/patologia , Dano ao DNA , Reparo do DNA , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Espermatogênese
15.
Genes (Basel) ; 15(6)2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38927643

RESUMO

Azoospermia is a form of male infertility characterized by a complete lack of spermatozoa in the ejaculate. Sertoli cell-only syndrome (SCOS) is the most severe form of azoospermia, where no germ cells are found in the tubules. Recently, FANCM gene variants were reported as novel genetic causes of spermatogenic failure. At the same time, FANCM variants are known to be associated with cancer predisposition. We performed whole-exome sequencing on a male patient diagnosed with SCOS and a healthy father. Two compound heterozygous missense mutations in the FANCM gene were found in the patient, both being inherited from his parents. After the infertility assessment, the patient was diagnosed with diffuse astrocytoma. Immunohistochemical analyses in the testicular and tumor tissues of the patient and adequate controls showed, for the first time, not only the existence of a cytoplasmic and not nuclear pattern of FANCM in astrocytoma but also in non-mitotic neurons. In the testicular tissue of the SCOS patient, cytoplasmic anti-FANCM staining intensity appeared lower than in the control. Our case report raises a novel possibility that the infertile carriers of FANCM gene missense variants could also be prone to cancer development.


Assuntos
Astrocitoma , Mutação de Sentido Incorreto , Síndrome de Células de Sertoli , Humanos , Masculino , Astrocitoma/genética , Astrocitoma/patologia , Astrocitoma/diagnóstico , Síndrome de Células de Sertoli/genética , Síndrome de Células de Sertoli/patologia , Adulto , Sequenciamento do Exoma , DNA Helicases/genética , Azoospermia/genética , Azoospermia/patologia , Azoospermia/diagnóstico
16.
Cells ; 13(10)2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38786072

RESUMO

Spermatogenesis is a highly regulated process dependent on androgen receptor (AR) signaling in Sertoli cells. However, the pathogenic mechanisms of spermatogenic failure, by which loss of AR impairs downstream target genes to affect Sertoli cell function, remain incompletely understood. By using microarray analysis, we identified several AR-regulated genes involved in the maturation of spermatogenesis, including chromodomain Y-like protein (CDYL) and transition proteins 1 (TNP-1), that were significantly decreased in ARKO mouse testes. AR and CDYL were found to co-localize and interact in Sertoli cells. The AR-CDYL complex bound to the promoter regions of TNP1 and modulated their transcriptional activity. CDYL acts as a co-regulator of AR transactivation, and its expression is decreased in the Sertoli cells of human testes from patients with azoospermia. The androgen receptor-chromodomain Y-like protein axis plays a crucial role in regulating a network of genes essential for spermatogenesis in Sertoli cells. Disruption of this AR-CDYL regulatory axis may contribute to spermatogenic failure. These findings provide insights into novel molecular mechanisms targeting the AR-CDYL signaling pathway, which may have implications for developing new therapeutic strategies for male infertility.


Assuntos
Receptores Androgênicos , Células de Sertoli , Transdução de Sinais , Espermatogênese , Animais , Humanos , Masculino , Camundongos , Azoospermia/metabolismo , Azoospermia/genética , Azoospermia/patologia , Proteínas de Homeodomínio , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Androgênicos/metabolismo , Receptores Androgênicos/genética , Células de Sertoli/metabolismo , Espermatogênese/genética , Fatores de Transcrição
17.
Cell Mol Life Sci ; 81(1): 211, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38722330

RESUMO

Spermatogonial stem cells (SSCs) are capable of transmitting genetic information to the next generations and they are the initial cells for spermatogenesis. Nevertheless, it remains largely unknown about key genes and signaling pathways that regulate fate determinations of human SSCs and male infertility. In this study, we explored the expression, function, and mechanism of USP11 in controlling the proliferation and apoptosis of human SSCs as well as the association between its abnormality and azoospermia. We found that USP11 was predominantly expressed in human SSCs as shown by database analysis and immunohistochemistry. USP11 silencing led to decreases in proliferation and DNA synthesis and an enhancement in apoptosis of human SSCs. RNA-sequencing identified HOXC5 as a target of USP11 in human SSCs. Double immunofluorescence, Co-immunoprecipitation (Co-IP), and molecular docking demonstrated an interaction between USP11 and HOXC5 in human SSCs. HOXC5 knockdown suppressed the growth of human SSCs and increased apoptosis via the classical WNT/ß-catenin pathway. In contrast, HOXC5 overexpression reversed the effect of proliferation and apoptosis induced by USP11 silencing. Significantly, lower levels of USP11 expression were observed in the testicular tissues of patients with spermatogenic disorders. Collectively, these results implicate that USP11 regulates the fate decisions of human SSCs through the HOXC5/WNT/ß-catenin pathway. This study thus provides novel insights into understanding molecular mechanisms underlying human spermatogenesis and the etiology of azoospermia and it offers new targets for gene therapy of male infertility.


Assuntos
Apoptose , Proliferação de Células , Espermatogênese , Tioléster Hidrolases , Via de Sinalização Wnt , Humanos , Masculino , Células-Tronco Germinativas Adultas/metabolismo , Apoptose/genética , Azoospermia/metabolismo , Azoospermia/genética , Azoospermia/patologia , beta Catenina/metabolismo , beta Catenina/genética , Proliferação de Células/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Espermatogênese/genética , Espermatogônias/metabolismo , Espermatogônias/citologia , Testículo/metabolismo , Testículo/citologia , Tioléster Hidrolases/genética , Tioléster Hidrolases/metabolismo , Via de Sinalização Wnt/genética
18.
Reprod Biol ; 24(2): 100890, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38723297

RESUMO

Recently we reported expressional alterations in 219 genes and their transcripts in Leydig cell tumors but nowadays there is still a lack of full basic biochemical characteristics of these tumors. The discovery of potential biochemical markers for tumor management from early detection, treatments, and control of therapy results may markedly supplement genetic data. Leydig cell micronodules were obtained from patients with azoospermia who were qualified for testicular biopsy. The biochemistry of Leydig cell tumors was analyzed using histological staining and spectrophotometric measurements of total proteins, carbohydrates, lipids, and nucleic acids. In addition, the levels of calcium (Ca2 +), copper (Cu2 +), zinc (Zn2 +), and selenium (Se2 +) ions were measured. When compared to healthy testis we revealed, for the first time, that in the interstitial tissue with Leydig cell tumors, great amounts of proteins, carbohydrates, lipids, and acids were dislocated from the seminiferous tubules. Measurements of organic compounds showed a decrease (P < 0.05) only in the Cu2 + content in Leydig cell tumors which may be related to their altered biochemical structure. This specific result may be promising for designing further approaches to manage this tumor based on combining morphological and molecular data.


Assuntos
Tumor de Células de Leydig , Neoplasias Testiculares , Humanos , Masculino , Tumor de Células de Leydig/patologia , Tumor de Células de Leydig/metabolismo , Neoplasias Testiculares/patologia , Neoplasias Testiculares/metabolismo , Adulto , Cobre/metabolismo , Testículo/patologia , Testículo/metabolismo , Zinco/metabolismo , Selênio , Cálcio/metabolismo , Azoospermia/metabolismo , Azoospermia/patologia , Células Intersticiais do Testículo/metabolismo , Células Intersticiais do Testículo/patologia
19.
J Med Ultrason (2001) ; 51(3): 465-475, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38710922

RESUMO

PURPOSE: To determine the feasibility of high-frequency ultrasound (HFUS) for assessing seminiferous tubules and to understand high-resolution B-mode images of the testes in cases of azoospermia. METHODS: We verified how the histopathological images of testicular biopsy specimens can be observed using HFUS images and measurement analysis of seminiferous tubules was performed to 28 testes of 14 cases with azoospermia who underwent preoperative ultrasound and microdissection testicular sperm extraction (micro-TESE). The population consisted of obstructive azoospermia (OA) and non-obstructive azoospermia (NOA), including Sertoli cell-only syndrome (SCOS), and the other pathologies. Statistical verification of differences in seminiferous tubule diameters among preoperative ultrasound examination, ultrasound examination of pathological specimens, and histopathological specimens. We also examined the imagingpathology correlation via a case series presentation, aiming to identify imaging markers of testicular pathology and determine the possibility of predicting each condition. RESULTS: A comparison between HFUS images and histopathology from the same biopsy specimens suggested that ultrasonography could be seen as stereoscopic images due to its significantly greater slice thickness. The diameters of tubules were generally larger in pathological tissues as compared to ultrasonographic findings in OA and SCOS, but not in the other conditions. Comparisons provided insights into the predictability of SCOS and revealed imaging findings such as gaps between tubules and decreased diameter reflective of testicular damage. CONCLUSION: Seminiferous tubules can be observed however the diameter of seminiferous tubules varies in imaging and histopathology depending on the pathology. Imaging findings that reflect testicular damage and the predictability of SCOS were revealed in this study, but further verification is required.


Assuntos
Azoospermia , Estudos de Viabilidade , Túbulos Seminíferos , Testículo , Ultrassonografia , Humanos , Masculino , Azoospermia/diagnóstico por imagem , Azoospermia/patologia , Ultrassonografia/métodos , Adulto , Túbulos Seminíferos/diagnóstico por imagem , Túbulos Seminíferos/patologia , Testículo/diagnóstico por imagem , Testículo/patologia , Pessoa de Meia-Idade , Biópsia
20.
Cells ; 13(10)2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38786055

RESUMO

Infertility is an important personal and society disease, of which the male factor represents half of all causes. One of the aspects less studied in male infertility is the immunological testicular microenvironment. Mast cells (MCs), having high potential for regulating spermatogenesis due to fine-tuning the state of the integrative buffer metabolic environment, are one of the most crucial cellular subpopulations of the testicular interstitium. One important component of the MC secretome is proteases that can act as proinflammatory agents and in extracellular matrix (ECM) remodeling. In the testis, MCs are an important cell component of the testicular interstitial tissue (TIT). However, there are still no studies addressing the analysis of a specific MC protease-carboxypeptidase A3 (CPA3)-in cases with altered spermatogenesis. The cytological and histotopographic features of testicular CPA3+ MCs were examined in a study involving 34 men with azoospermia. As revealed, in cases with non-obstructive azoospermia, a higher content of CPA3+ MCs in the TIT and migration to the microvasculature and peritubular tissue of seminiferous tubules were observed when compared with cases with obstructive azoospermia. Additionally, a high frequency of CPA3+ MCs colocalization with fibroblasts, Leydig cells, and elastic fibers was detected in cases with NOA. Thus, CPA3 seems to be of crucial pathogenetic significance in the formation of a profibrogenic background of the tissue microenvironment, which may have direct and indirect effects on spermatogenesis.


Assuntos
Azoospermia , Mastócitos , Testículo , Adulto , Humanos , Masculino , Azoospermia/patologia , Azoospermia/metabolismo , Carboxipeptidases A/metabolismo , Mastócitos/metabolismo , Mastócitos/patologia , Espermatogênese , Testículo/metabolismo , Testículo/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...