Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
1.
J Urol ; 211(5): 678-686, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38375822

RESUMO

PURPOSE: We evaluate microscopic (micro) testicular sperm extraction (TESE) timing relative to oocyte retrieval on intracytoplasmic sperm injection outcome. MATERIALS AND METHODS: Couples with nonobstructive azoospermia who underwent intracytoplasmic sperm injection with freshly retrieved spermatozoa were analyzed based on whether micro-TESE was performed at least 1 day prior to oocyte retrieval (TESE-day-before group) or on the day of oocyte retrieval (TESE-day-of group). Embryology and clinical outcomes were compared. RESULTS: The percentage of patients who underwent a successful testicular sperm retrieval was significantly lower in the TESE-day-before cohort (62%) than in the TESE-day-of cohort (69%; odds ratio [OR] 1.4, 95% CI [1.1, 1.7], P < .001). The fertilization rate was also found to be significantly lower in the TESE-day-before group (45%) than in the TESE-day-of group (53%; OR 1.4, 95% CI [1.2, 1.7], P = .01). Although the association between the cleavage rate and TESE timing was not statistically significant, the implantation rate was found to be significantly higher in the day-before cohort (28%) than in the day-of cohort (22%; OR 0.7, 95% CI [0.6, 0.9], P = .01). Nevertheless, it was found that the clinical pregnancy and delivery rates were not statistically significantly associated with the TESE timing. CONCLUSIONS: Although sperm retrieval and fertilization rates were lower in the TESE-day-before cohort, the 2 cohorts showed comparable embryologic and clinical outcomes. Micro-TESE can be performed before oocyte harvesting to provide physicians ample time to decide between cancelling oocyte retrieval or retrieving oocytes for cryopreservation.


Assuntos
Azoospermia , Injeções de Esperma Intracitoplásmicas , Gravidez , Feminino , Humanos , Masculino , Recuperação de Oócitos , Testículo/patologia , Sêmen , Azoospermia/terapia , Azoospermia/patologia , Espermatozoides/patologia , Recuperação Espermática , Biópsia , Estudos Retrospectivos
2.
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
3.
Cell Mol Life Sci ; 80(3): 67, 2023 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-36814036

RESUMO

Male infertility can be caused by quantitative and/or qualitative abnormalities in spermatogenesis, which affects men's physical and mental health. Sertoli cell-only syndrome (SCOS) is the most severe histological phenotype of male infertility characterized by the depletion of germ cells with only Sertoli cells remaining in the seminiferous tubules. Most SCOS cases cannot be explained by the already known genetic causes including karyotype abnormalities and microdeletions of the Y chromosome. With the development of sequencing technology, studies on screening new genetic causes for SCOS are growing in recent years. Directly sequencing of target genes in sporadic cases and whole-exome sequencing applied in familial cases have identified several genes associated with SCOS. Analyses of the testicular transcriptome, proteome, and epigenetics in SCOS patients provide explanations regarding the molecular mechanisms of SCOS. In this review, we discuss the possible relationship between defective germline development and SCOS based on mouse models with SCO phenotype. We also summarize the advances and challenges in the exploration of genetic causes and mechanisms of SCOS. Knowing the genetic factors of SCOS offers a better understanding of SCO and human spermatogenesis, and it also has practical significance for improving diagnosis, making appropriate medical decisions, and genetic counseling. For therapeutic implications, SCOS research, along with the achievements in stem cell technologies and gene therapy, build the foundation to develop novel therapies for SCOS patients to produce functional spermatozoa, giving them hope to father children.


Assuntos
Azoospermia , Infertilidade Masculina , Síndrome de Células de Sertoli , Animais , Camundongos , Criança , Humanos , Masculino , Síndrome de Células de Sertoli/genética , Síndrome de Células de Sertoli/patologia , Azoospermia/genética , Azoospermia/patologia , Testículo/patologia , Túbulos Seminíferos , Espermatogênese/genética , Infertilidade Masculina/patologia
4.
Int J Urol ; 31(1): 17-24, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37737473

RESUMO

Approximately 1% of the general male population has azoospermia, and nonobstructive azoospermia accounts for the majority of cases. The causes vary widely, including chromosomal and genetic abnormalities, varicocele, drug-induced causes, and gonadotropin deficiency; however, the cause is often unknown. In azoospermia caused by hypogonadotropic hypogonadism, gonadotropin replacement therapy can be expected to produce sperm in the ejaculate. In some cases, upfront varicocelectomy for nonobstructive azoospermia with varicocele may result in the appearance of ejaculated spermatozoa; however, the appropriate indication should be selected. Each guideline recommends microdissection testicular sperm extraction for nonobstructive azoospermia in terms of successful sperm retrieval and avoidance of complications. Sperm retrieval rates generally ranged from 20% to 70% but vary depending on the causative disease. Various attempts have been made to predict sperm retrieval and improve sperm retrieval rates; however, the evidence is insufficient. Further evidence accumulation is needed for salvage treatment in cases of failed sperm retrieval. In Japan, there is inadequate provision on the right to know the origin of children born from artificial insemination of donated sperm and the rights of sperm donors, as well as information on unrelated family members, and the development of these systems is challenging. In the future, it is hoped that the pathogenesis of nonobstructive azoospermia with an unknown cause will be elucidated and that technology for omics technologies, human spermatogenesis using pluripotent cells, and organ culture methods will be developed.


Assuntos
Azoospermia , Varicocele , Criança , Humanos , Masculino , Azoospermia/etiologia , Azoospermia/terapia , Varicocele/complicações , Varicocele/cirurgia , Microdissecção/efeitos adversos , Sêmen , Estudos Retrospectivos , Gonadotropinas , Testículo/patologia
5.
Hum Reprod ; 38(2): 306-314, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36524333

RESUMO

STUDY QUESTION: Can the analysis of a large Turkish consanguineous family via whole exome sequencing (WES) identify novel causative genetic variation responsible for nonobstructive azoospermia (NOA) characterized by arrest at primary spermatocyte stage? SUMMARY ANSWER: WES analysis revealed a homozygous nonsense variant in HORMAD1 in three affected brothers of a Turkish family. WHAT IS KNOWN ALREADY: Studying patient cohorts in small or large consanguineous families using high-throughput sequencing allows the identification of genetic causes of different pathologies, including infertility. Over the last two decades, a number of genes involved in human male infertility have been discovered, but only 14 genes have been identified as being at least moderately linked to isolated NOA or oligozoospermia in men. STUDY DESIGN, SIZE, DURATION: The study included a Turkish family comprising three brothers with NOA. Two brothers had a normal karyotype, normal hormonal levels and no Yq microdeletion. The testicular histopathology analysis revealed the complete arrest of spermatogenesis at the primary spermatocyte stage. PARTICIPANTS/MATERIALS, SETTING, METHODS: We recruited a consanguineous Turkish family where parents were first-degree cousins and had seven children; three sons who had NOA, two sons who were fertile and two daughters for whom no information was available. Saliva samples from the index patient, his two affected brothers, parents and two nonaffected brothers (seven samples in total) were collected. Prior to WES, the index patient underwent targeted genetic testing using an infertility panel, which includes 133 infertility genes. No pathogenic variations were identified. WES was then performed on the DNA of the seven family members available. Bioinformatics analysis was performed using an in-house pipeline. Detected variants were scored and ranked, and copy number variants were called and annotated.The consequences of mutation on protein expression and localization were investigated by cell transfection followed by immunofluorescence or immunoblotting. MAIN RESULTS AND THE ROLE OF CHANCE: WES revealed a homozygous nonsense variant chr1:150675797G>A; HORMAD1 (NM_032132.5): c.1021C>T, p.Gln341* in exon 13, which was confirmed in all three affected brothers. HORMAD1 encodes the HORMA domain-containing protein 1. The parents as well as the two fertile brothers were carriers of this variant. This variant may lead to the production of a truncated protein lacking the nuclear localization signal; therefore, human cells were transfected with the wild-type and mutated form, in fusion with green fluorescent protein. Immunoblotting experiments confirmed the production of a truncated HORMAD1 protein, and immunofluorescence microscopy revealed that the mutated protein displayed cytoplasmic localization while the wild type protein located to the nucleus. Altogether, our findings validate HORMAD1 as an essential genetic factor in the meiotic process in human. LIMITATIONS, REASONS FOR CAUTION: According to one scoring system used to evaluate the clinical validity of male infertility genes, this study would classify HORMAD1 as displaying limited clinical evidence of being involved in male infertility. However, such a score is the maximum possible when only one family is analyzed and the addition of one patient showing a pathogenic or likely pathogenic variant would immediately change this classification to 'moderate'. Thus, this report should prompt other researchers to screen patients with NOA for this genetic variant. WIDER IMPLICATIONS OF THE FINDINGS: Identification of new genetic factors involved in the human meiosis process will contribute to an improvement of our knowledge at the basic level, which in turn will allow the management of better care for infertile patients. Since Hormad1-/- knock-out female mice are also infertile, HORMAD1 could also be involved in human female infertility. Our findings have direct implications for the genetic counseling of patients and their family members. STUDY FUNDING/COMPETING INTEREST(S): The study was funded by Fondation Maladies Rares (High Throughput Sequencing and Rare Diseases-2018, 'GenOmics of rare diseases'). The authors declare that they have no conflict of interest. TRIAL REGISTRATION NUMBER: N/A.


Assuntos
Azoospermia , Infertilidade Masculina , Animais , Camundongos , Criança , Humanos , Masculino , Feminino , Azoospermia/genética , Azoospermia/patologia , Consanguinidade , Doenças Raras , Infertilidade Masculina/genética , Proteínas/genética , Proteínas de Ciclo Celular/genética
6.
Reprod Biol Endocrinol ; 20(1): 90, 2022 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-35710416

RESUMO

BACKGROUND: Nonobstructive azoospermia (NOA) is one of the most difficult forms of male infertility to treat, and its pathogenesis is still unclear. miRNAs can regulate autophagy by affecting their target gene expression. Our previous study found that miR-188-3p expression in NOA patients was low. There are potential binding sites between the autophagy gene ATG7 and miR-188-3p. This study aimed to verify the binding site between miR-188-3p and ATG7 and whether miR-188-3p affects autophagy and participates in NOA by regulating ATG7 to influence the autophagy marker genes LC3 and Beclin-1. METHODS: Testicular tissue from 16 NOA patients and 16 patients with normal spermatogenesis and 5 cases in each group of pathological sections were collected. High-throughput sequencing was performed to detect mRNA expression differences. Quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting, immunohistochemical staining and immunofluorescence were used to detect protein localization and expression. Autophagosome changes were detected by electron microscopy. The targeting relationship between miR-188-3p and ATG7 was confirmed by a luciferase assay. RESULTS: ATG7 protein was localized in the cytoplasm of spermatogenic cells at all levels, and the ATG7 gene (p = 0.019) and protein (p = 0.000) were more highly expressed in the NOA group. ATG7 expression after overexpression/inhibition of miR-188-3p was significantly lower (p = 0.029)/higher (p = 0.021) than in the control group. After overexpression of miR-188-3p, the ATG7 3'UTR-WT luciferase activity was impeded (p = 0.004), while the ATG7 3'UTR-MUT luciferase activity showed no significant difference (p = 0.46). LC3 (p = 0.023) and Beclin-1 (p = 0.041) expression in the NOA group was significantly higher. LC3 and Beclin-1 gene expression after miR-188-3p overexpression/inhibition was significantly lower (p = 0.010 and 0.024, respectively) and higher (p = 0.024 and 0.049, respectively). LC3 punctate aggregation in the cytoplasm decreased after overexpression of miR-188-3p, while the LC3 punctate aggregation in the miR-188-3p inhibitor group was higher. The number of autophagosomes in the miR-188-3p mimic group was lower than the number of autophagosomes in the mimic NC group. CONCLUSIONS: LC3 and Beclin-1 were more highly expressed in NOA testes and negatively correlated with the expression of miR-188-3p, suggesting that miR-188-3p may be involved in the process of autophagy in NOA. miR-188-3p may regulate its target gene ATG7 to participate in autophagy anDual luciferase experiment d affect the development of NOA.


Assuntos
Azoospermia , MicroRNAs , Regiões 3' não Traduzidas , Autofagia/genética , Proteína 7 Relacionada à Autofagia/genética , Azoospermia/genética , Proteína Beclina-1/genética , Humanos , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo
7.
Cell Biochem Funct ; 40(8): 865-879, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36121211

RESUMO

DNA repair processes are critical to maintaining genomic integrity. As a result, dysregulation of repair genes is likely to be linked with health implications, such as an increased prevalence of infertility and an accelerated rate of aging. We evaluated all the DNA repair genes (322 genes) by microarray. This study has provided insight into the connection between DNA repair genes, including RAD23B, OBFC2A, PMS1, UBE2V1, ERCC5, SMUG1, RFC4, PMS2L5, MMS19, SHFM1, INO80, PMS2L1, CHEK2, TRIP13, and POLD4. The microarray analysis of six human cases with different nonobstructive azoospermia revealed that RAD23B, OBFC2A, PMS1, UBE2V1, ERCC5, SMUG1, RFC4, PMS2L5, MMS19, SHFM1, and INO80 were upregulated, and expression of PMS2L1, CHEK2, TRIP13, and POLD4 was downregulated versus the normal case. For this purpose, Enrich Shiny GO, STRING, and Cytoscape online evaluation was applied to predict proteins' functional and molecular interactions and then performed to recognize the master pathways. Functional enrichment analysis revealed that the biological process (BP) terms "base-excision repair, AP site formation," "nucleotide-excision repair, DNA gap filling," and "nucleotide-excision repair, preincision complex assembly" was significantly overexpressed in upregulated differentially expressed genes (DEGs). BP analysis of downregulated DEGs highlighted "histone phosphorylation," "DNA damage response, detection DNA response," "mitotic cell cycle checkpoint signaling," and "double-strand break repair." Overrepresented molecular function (MF) terms in upregulated DEGs included "Oxidized base lesion DNA N-glycosylase activity," "uracil DNA N-glycosylase activity," "bubble DNA binding" and "DNA clamp loader activity." Interestingly, MF investigation of downregulated DEGs showed overexpression in "heterotrimeric G-protein complex," "5'-deoxyribose-5-phosphate lyase activity," "minor groove of adenine-thymine-rich DNA binding," and "histone kinase activity." Our findings suggest that these genes and their interacting hub proteins could help determine the pathophysiology of germ cell abnormalities and infertility.


Assuntos
Reparo do DNA , Testículo , Humanos , Masculino , DNA/metabolismo , Análise em Microsséries , Nucleotídeos/metabolismo , Testículo/metabolismo
8.
J Assist Reprod Genet ; 39(5): 1195-1203, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35486194

RESUMO

PURPOSE: To investigate the genetic cause of nonobstructive azoospermia (NOA). METHODS: We performed whole exome sequencing (WES) on the proband who had three relatives suffering from NOA. We used a list of candidate genes which have high expression level in testis and their mutations have been reported in NOA. Sanger sequencing verified the identified variant and its structural and functional consequence was evaluated by protein three-dimensional (3D) structure prediction and protein-ligand docking. RESULTS: WES revealed a novel splice-acceptor mutation (c.1832-2A>T) in helicase for meiosis 1 (HFM1) gene, which co-segregated with the NOA in this family. 3D structural models were generated and verified. Molecular docking indicated that the c.1832-2A>T mutation affects not only the ADP binding residues but also the hydrogen bond interactions. The ADP binding site will be lost in the mutant protein, potentially causing defective crossover and synapsis. CONCLUSION: We report that the c.1832-2A>T mutation is the likely cause of NOA in the family studied. Regarding that many reported NOA genes are involved in the formation of crossovers and synapsis and have critical roles in the production of germ cells, we suggest that such genes should be considered for screening of infertility among large cohorts of infertile individuals.


Assuntos
Azoospermia , Difosfato de Adenosina/metabolismo , Azoospermia/diagnóstico , DNA Helicases/genética , Humanos , Masculino , Simulação de Acoplamento Molecular , Mutação/genética , Testículo/metabolismo
9.
Mol Hum Reprod ; 27(9)2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34515795

RESUMO

Nonobstructive azoospermia (NOA) and diminished ovarian reserve (DOR) are two disorders that can lead to infertility in males and females. Genetic factors have been identified to contribute to NOA and DOR. However, the same genetic factor that can cause both NOA and DOR remains largely unknown. To explore the candidate pathogenic gene that causes both NOA and DOR, we conducted whole-exome sequencing (WES) in a non-consanguineous family with two daughters with DOR and a son with NOA. We detected one pathogenic frameshift variant (NM_007068:c.28delG, p. Glu10Asnfs*31) following a recessive inheritance mode in a meiosis gene DMC1 (DNA meiotic recombinase 1). Clinical analysis showed reduced antral follicle number in both daughters with DOR, but metaphase II oocytes could be retrieved from one of them. For the son with NOA, no spermatozoa were found after microsurgical testicular sperm extraction. A further homozygous Dmc1 knockout mice study demonstrated total failure of follicle development and spermatogenesis. These results revealed a discrepancy of DMC1 action between mice and humans. In humans, DMC1 is required for spermatogenesis but is dispensable for oogenesis, although the loss of function of this gene may lead to DOR. To our knowledge, this is the first report on the homozygous frameshift mutation as causative for both NOA and DOR and demonstrating that DMC1 is dispensable in human oogenesis.


Assuntos
Azoospermia/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Adulto , Animais , Células Cultivadas , China , Análise Mutacional de DNA , Feminino , Mutação da Fase de Leitura , Predisposição Genética para Doença , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linhagem , Insuficiência Ovariana Primária/genética
10.
Andrologia ; 53(5): e14010, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33591612

RESUMO

The aim of this study is to investigate the efficiency of elastic light single-scattering spectroscopy system, a noninvasive method, to acquire spectra during testicular biopsy from normal and damaged seminiferous tubules with various degrees of germ cell loss. Adult control rats and doxorubicin-injected rats to achieve seminiferous germ cell loss (for 10 days [10D], 20 days [D20], 30 days [D30], 40 days [D40], and 50 days [D50]) were used. Spectroscopic measurements were acquired utilising a single-fibre optical probe, and histopathology of the biopsied testicular tissue samples were compared. Time-dependent testicular damage comprising various degrees of seminiferous tubule degeneration after doxorubicin-administration was observed. In D30, D40 and D50 groups, where significant germ cell loss was identified, elastic light single-scattering spectroscopy system signals were well correlated with disturbed spermatogenesis where significant differences in spectral signals were obtained. Our findings indicate that the elastic light single-scattering spectroscopy system has the potential to enable instant imaging of spermatogenesis in rats and could also be useful in humans for clinical applications, such as to increase sperm recovery success during micro-TESE for men with nonobstructive azoospermia.


Assuntos
Azoospermia , Recuperação Espermática , Adulto , Animais , Humanos , Masculino , Ratos , Túbulos Seminíferos , Análise Espectral , Espermatogênese , Espermatozoides , Testículo
11.
Andrologia ; 53(5): e14040, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33682176

RESUMO

Our ability to predict the potential of testicular spermatozoa to support embryonic development is still limited. Although motility of testicular spermatozoa is associated with embryo development, the impact of morphology and the presence of spermatozoa in the testicular sample has not been previously researched. Moreover, while the majority of data indicate no effect of cryopreservation, there are studies reporting impaired clinical outcomes due to testicular cryopreservation. In a retrospective study, 118 ICSI-TESE cycles were analysed to study the impact of (a) total quality of testicular tissue, (b) testicular tissue cryopreservation and (c) presence/motility/morphology of testicular spermatozoa in fertilisation rate, embryonic development, clinical pregnancy (CPR), ongoing pregnancy (OPR) and live birth rate (LBR). Results showed that fertilisation rate was significantly affected by both total quality of testicular tissue (p < .05) and rare presence of spermatozoa (p < .01). Moreover, total tissue quality (p < .01), cryopreservation of low-quality samples (p < .01), absence of motile testicular spermatozoa (p < .01) and poor spermatozoa morphology (p < .05) had a negative impact on the number of good quality day 3 embryos. CPR, OPR or LBR was not affected by any parameters examined. Our data suggest that the quality of testicular tissue influences both fertilisation rate and embryo development. Moreover, cryopreservation of low-quality testicular samples has a negative impact on the number of available embryos for transfer.


Assuntos
Preservação do Sêmen , Criopreservação , Feminino , Humanos , Masculino , Gravidez , Taxa de Gravidez , Estudos Retrospectivos , Injeções de Esperma Intracitoplásmicas , Espermatozoides , Testículo
12.
Andrologia ; 53(11): e14220, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34423455

RESUMO

Limited factors effectively predict sperm retrieval with microdissection testicular sperm extraction in men with nonobstructive azoospermia. We therefore sought to evaluate the role of serum anti-Müllerian hormone as a predictive biomarker for successful sperm retrieval. We included patients with pre-operative anti-Müllerian hormone levels and stratified them based on prior history of prior sperm retrieval procedure. We compared hormone levels between those who did and did not have a successful sperm retrieval and used receiver operating curves to determine an optimal cut-off value. A total of 46 men were included, of whom 18 (39.1%) had no prior sperm retrieval and 11 (61.1%) had sperm successfully retrieved. Pre-operative serum anti-Müllerian hormone levels were predictive of sperm retrieval in patients with no prior attempts at retrieval (p = .03). Receiver operating curve for those without prior retrieval was 0.6753. The optimal anti-Müllerian hormone cut-off for those without prior sperm retrieval was 0.133 ng/ml with a sensitivity of 0.91 and specificity of 0.29. Therefore, serum anti-Müllerian hormone levels have modest predictive value for sperm retrieval in this cohort. The combination of clinical history, examination and laboratory investigations should continue to be used to guide surgeons in counselling patients regarding the chance of sperm retrieval.


Assuntos
Hormônio Antimülleriano , Azoospermia , Azoospermia/cirurgia , Humanos , Masculino , Microdissecção , Estudos Retrospectivos , Recuperação Espermática , Espermatozoides , Testículo/cirurgia
13.
Andrologia ; 53(11): e14208, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34352113

RESUMO

OBJECTIVES: Men with nonobstructive azoospermia (NOA), cryptozoospermia and severe oligozoospermia are candidates for microdissection testicular sperm extraction (micro-TESE) and intracytoplasmic sperm injection (ICSI). We sought to evaluate micro-TESE outcomes and the need for bilateral testicular exploration in the three groups of men. METHODS: We conducted a retrospective study of 233 consecutive micro-TESEs in men with nonobstructive azoospermia (n = 173), cryptozoospermia (n = 43) and severe oligozoospermia (n = 17). The decision to terminate the micro-TESE after a unilateral or bilateral testicular exploration was determined at the time of surgery and was based on the presence or absence of mature spermatozoa in the harvested micro-biopsies. Final assessment of sperm recovery, on the day of ICSI, was reported as successful (available spermatozoon for ICSI) or unsuccessful (no spermatozoon for ICSI). RESULTS: Unilateral testicular exploration resulted in successful sperm retrieval in 43% (75/173), 79% (34/43) and 100% (17/17) of men with NOA, cryptozoospermia and severe oligozoospermia respectively. Therefore, 57%, 21% and none of the men with NOA, cryptozoospermia and severe oligozoospermia, respectively, required a bilateral micro-TESE. Overall, micro-TESE resulted in successful sperm retrieval in 52% (90/173), 91% (39/43) and 100% (17/17) of men with NOA, cryptozoospermia and severe oligozoospermia respectively. CONCLUSION: Our data indicate that men with severe oligozoospermia are unlikely to require a bilateral testicular exploration at micro-TESE. Moreover, most cryptozoospermic men will have a successful sperm retrieval by micro-TESE with the majority of these patients requiring a unilateral exploration. In contrast, over 50% of the men with nonobstructive azoospermia will require a bilateral micro-TESE.


Assuntos
Azoospermia , Oligospermia , Azoospermia/cirurgia , Humanos , Masculino , Microdissecção , Oligospermia/terapia , Estudos Retrospectivos , Recuperação Espermática , Espermatozoides , Testículo/cirurgia
14.
Andrologia ; 53(9): e14170, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34196417

RESUMO

This study evaluates the efficacy of vas ligation in enhancing sperm retrieval in nonobstructive azoospermia cases, by accumulating intratesticular spermatozoa. Fifty-six mature male rats with equally sized testes were included in this study. Forty-six were in the study group, and 10 were in the control group. Bilateral testicular fine needle aspiration was performed for all, to confirm presence of spermatozoa in all testes. Nonobstructive azoospermia was induced in all 56 rats, using Dienogest (40 mg/kg) + Testosterone Undecanoate (25 mg/kg) every month for three months. Monthly aspirations confirmed nonobstructive azoospermia from all rats, within the three months treatment. This was followed by unilateral vas ligation and was performed for 46 rats of the study group, with no ligation performed in the control group. After a further period of 90 days (2 spermatogenic cycles) with the same medical treatment maintained, bilateral testicular sperm extraction was performed. Sperm retrieval was evaluated, comparing the outcome of vas-ligated testicles to the nonligated. Upon evaluation, spermatozoa were found in 14/46 of the vas-ligated testes (30.4%), compared to none of the nonligated (0/66), p = .0005. Ligation of the vas deferens in rats with nonobstructive azoospermia may enhance the results of sperm retrieval via sperm accumulation.


Assuntos
Azoospermia , Oligospermia , Vasectomia , Animais , Azoospermia/tratamento farmacológico , Humanos , Masculino , Ratos , Estudos Retrospectivos , Recuperação Espermática , Espermatozoides , Testículo/cirurgia
15.
Andrologia ; 53(3): e13936, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33427330

RESUMO

Altered protamine 1 (PRM1)/ protamine 2 (PRM2) mRNA ratio in testicular biopsy samples correlates with sperm quality and its fertilising ability. This study is planned to assess PRM1/ PRM2 mRNA ratio in subgroups of azoospermia to suggest a more reliable and accurate marker for assessing sperm quality in nonobstructive azoospermia (NOA). A cross-sectional study was done on testicular biopsy samples, taken from 106 azoospermic patients. Samples were histologically classified into subgroups: 36 obstructive azoospermia (OA), and two groups of NOA: 41 round spermatid maturation arrest (SMA) and 29 Sertoli cell-only syndrome (SCOS). OA samples showed histologically normal spermatogenesis and serve as a positive control. mRNA expression of jumonji domain-containing 1A (JMJD1A), PRM1, PRM2 and transition nuclear proteins (TNP1, TNP2) genes was determined, by RT-qPCR. Significantly lower expression of JMJD1A (p < .001), PRM1 (p = .0265) and PRM2 (p = .0032) has been seen in the SCOS group of NOA. We found significant (p < .001) increase in PRM1/PRM2 mRNA ratio in testicular biopsy samples of SCOS group of NOA patients and significant negative correlation of PRM1/PRM2 mRNA ratio with JMJD1A. Hence, PRM1/PRM2 mRNA ratio may represent a more reliable and accurate marker to assess sperm quality in NOA in addition to standard semen parameters.


Assuntos
Azoospermia , Protaminas/genética , Azoospermia/genética , Estudos Transversais , Humanos , Masculino , RNA Mensageiro/genética , Testículo
16.
Yi Chuan ; 43(5): 473-486, 2021 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-33972217

RESUMO

About 15% couples suffer from infertility, half of which are caused by male factors. Male infertility usually manifests as teratozoospermia, oligospermia and/or asthenospermia, of which the most severe form is azoospermia. In this review, we summarize the recent progress in the study of genetic factors involved in nonobstructive azoospermia and teratozoospermia, Recently, with the rapid development of high-throughput chips and sequencing technologies, many genetic factors of spermatogenesis have been discovered and analyzed. For the nonobstructive azoospermia, genome-wide association studies (GWAS) and high-throughput sequencing revealed many risk loci of nonobstructive azoospermia. For the teratozoospermia, the application of whole-exome sequencing (WES) revealed a series of disease-causing genes, greatly enriching our knowledge of teratozoospermia including multiple morphological abnormalities of the flagella (MMAF). The discovery of lots of disease genes helped the characterization of the pathological mechanisms of male infertility. Therefore, a comprehensive and in-depth understanding of genetic factors in spermatogenesis abnormalities will play important roles in the clinical diagnosis, treatment and genetic counseling of male infertility.


Assuntos
Azoospermia , Infertilidade Masculina , Azoospermia/genética , Estudo de Associação Genômica Ampla , Humanos , Infertilidade Masculina/genética , Masculino , Mutação , Espermatogênese/genética
17.
Hum Reprod ; 35(11): 2413-2427, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32914196

RESUMO

STUDY QUESTION: Whether the testis-specific extracellular vesicle (EV) long noncoding RNAs (lncRNAs) in seminal plasma could be utilized to predict the presence of testicular spermatozoa in nonobstructive azoospermia (NOA) patients? SUMMARY ANSWER: Our findings indicate that the panel based on seminal plasma EV lncRNAs was a sensitive and specific method in predicting the presence of testicular spermatozoa and may improve clinical decision-making of NOA. WHAT IS KNOWN ALREADY: The adoption of sperm retrieval techniques, especially microdissection testicular sperm extraction (mTESE), in combination with ICSI has revolutionized treatment for NOA. However, there are no precise and noninvasive methods for predicting whether there are testicular spermatozoa in NOA patients before mTESE. STUDY DESIGN, SIZE, DURATION: RNA sequencing was performed on seminal plasma EVs from 6 normozoospermic men who underwent IVF due to female factor and 5 idiopathic NOA patients who failed to obtain testicular spermatozoa by mTESE and were diagnosed as having Sertoli cell-only syndrome by postoperative pathology. A biomarker panel of lncRNAs was constructed and verified in 96 NOA patients who underwent mTESE. Decision-making process was established based on the panel in seminal plasma EVs from 45 normozoospermia samples, 43 oligozoospermia samples, 62 cryptozoospermia samples, 96 NOA samples. PARTICIPANTS/MATERIALS, SETTING, METHODS: RNA sequencing was done to examine altered profiles of EV lncRNAs in seminal plasma. Furthermore, a panel consisting of EV lncRNAs was established and evaluated in training set and validation sets. MAIN RESULTS AND THE ROLE OF CHANCE: A panel consisting of nine differentially expressed testis-specific lncRNAs, including LOC100505685, SPATA42, CCDC37-DT, GABRG3-AS1, LOC440934, LOC101929088 (XR_927561.2), LOC101929088 (XR_001745218.1), LINC00343 and LINC00301, was established in the training set and the AUC was 0.986. Furthermore, the AUC in the validation set was 0.960. Importantly, the panel had a unique advantage when compared with models based on serum hormones from the same group of NOA cases (AUC, 0.970 vs 0.723; 0.959 vs 0.687, respectively). According to the panel of lncRNAs, a decision-making process was established, that is when the score of an NOA case exceeds 0.532, sperm retrieval surgery may be recommended. LIMITATIONS, REASONS FOR CAUTION: In the future, the sample size needs to be further expanded. Meanwhile, the regulatory functions and mechanism of lncRNAs in spermatogenesis also need to be elucidated. WIDER IMPLICATIONS OF THE FINDINGS: When the score of our panel is below 0.532, subjecting the NOA patients to ineffective surgical interventions may not be recommended due to poor sperm retrieval rate. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the National Natural Science Foundation of China (81871110, 81971314 and 81971759); the Guangdong Special Support Plan-Science and Technology Innovation Youth Top Talents Project (2016TQ03R444); the Science and Technology Planning Project of Guangdong Province (2016B030230001 and 201707010394); the Key Scientific and Technological Program of Guangzhou City (201604020189); the Pearl River S&T Nova Program of Guangzhou (201806010089); the Transformation of Scientific and Technological Achievements Project of Sun Yat-sen University (80000-18843235) and the Youth Teacher Training Project of Sun Yat-sen University (17ykpy68 and 18ykpy09). There are no competing interests related to this study. TRIAL REGISTRATION NUMBER: N/A.


Assuntos
Azoospermia , Vesículas Extracelulares , RNA Longo não Codificante , Adolescente , Azoospermia/diagnóstico , Azoospermia/genética , Azoospermia/terapia , China , Feminino , Humanos , Masculino , RNA Longo não Codificante/genética , Estudos Retrospectivos , Sêmen , Recuperação Espermática , Espermatozoides , Testículo
18.
Andrologia ; 52(2): e13486, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31825116

RESUMO

The effectiveness of varicocelectomy in nonobstructive azoospermia is controversial. The current study assessed the efficacy of microsurgical subinguinal varicocelectomy in nonobstructive azoospermic men with palpable varicocele and to evaluate predictive parameters of outcome. We reviewed the records of 723 patients who had microsurgical varicocelectomy and diagnostic testicular biopsy between 2012 and 2016 at a tertiary medical centre. Data pertaining to the physical, laboratory (semen analysis and hormonal profile) and histopathology features were examined, exploring the predictors of improvement in semen analysis post-varicocelectomy. In total, 42 patients with mean age 35.71 ± 6.35 years were included. After a mean varicocelectomy follow-up of 6.7 months, motile spermatozoa in the ejaculate could be observed in 11 patients (26.2). Out of all the factors examined, only testicular histopathology significantly predicted post-varicocelectomy outcome, where 8/11 patients exhibited hypospermatogenesis, and 3/11 Sertoli cell-only regained spermatozoa in semen. Microsurgical varicocelectomy in nonobstructive azoospermic men with clinically palpable varicocele can result in sperm appearance in the ejaculate with the highest success expected in hypospermatogenesis.


Assuntos
Azoospermia/cirurgia , Análise do Sêmen , Procedimentos Cirúrgicos Urológicos Masculinos , Varicocele/cirurgia , Adulto , Humanos , Masculino , Estudos Retrospectivos
19.
Andrologia ; 52(11): e13814, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32894622

RESUMO

Nonobstructive azoospermia (NOA), which is considered the most severe form of male infertility, has placed a heavy burden on families and society. As vital regulators of transcriptional and post-transcriptional levels, Noncoding RNAs (ncRNAs) are closely related to all the pathophysiological processes involved in infertility in males, especially spermatogenesis. Our study explored the expression levels of circ_0049356 in both the whole blood and seminal plasma samples of idiopathic NOA patients via quantitative real-time PCR. Furthermore, the relative expression of its host gene (CARM1) was also determined using the same methods. In addition, as circRNAs have been demonstrated to regulate gene expression as miRNAs sponge, we predicted a total of five miRNAs and 101 mRNAs as putative downstream targets and constructed a circRNA-miRNA-mRNA network. Based on the predictions, Gene Ontology and KEGG pathway analyses were performed for further bioinformatics analysis to explore the potential function and investigate the circ_0049356-miRNA-mRNA interactions. Our results show target mRNAs that have been predicted to regulate guanyl-nucleotide exchange factor activity to mediate the GTP/GDP exchange, and downstream targets possibly involved in the regulation of the actin cytoskeleton, which play a significant role in cytoskeleton rearrangement of germ cells during spermatogenesis.


Assuntos
Azoospermia , MicroRNAs , RNA Circular , Azoospermia/genética , Ontologia Genética , Humanos , Masculino , MicroRNAs/genética , RNA Mensageiro/genética
20.
Andrologia ; 52(2): e13475, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31820482

RESUMO

The association of genetic variants and congenital bilateral absence of the vas deferens (CBAVD) has been well acknowledged. By contrast, the link between nonobstructive azoospermia (NOA) or oligospermia and alterations in the cystic fibrosis transmembrane conductive regulator (CFTR) remains inconclusive. To clarify the problem, a meta-analysis was performed out after systematically searching Pubmed, Web of Science, Embase and the Chinese national knowledge infrastructure (CNKI) database. As we know, the ∆F508 and IVS8-5T gene mutations are the most studied genetic variants in CFTR gene. We reviewed the data from male patients who underwent the aforementioned genetic test. Our study revealed that the IVS8-5T mutation may be positively associated with the risk of nonobstructive male infertility (odds ratio (OR) 1.69; 95% CI: 1.12-2.55). This association strengthened when concerning NOA (OR: 2.62; 95% CI: 1.49-4.61). However, the ∆F508 mutation seemed to be a smaller contributing factor to this risk (OR: 1.63; 95% CI: 0.86-3.08). Our study aims to clarify the association between the ∆F508 and IVS8-5T gene mutations and nonobstructive male infertility. Therefore, screening for the IVS8-5T mutation in the CFTR gene may be recommended for men with NOA or severe oligozoospermia seeking assisted reproductive technology (ART).


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Infertilidade Masculina/genética , Humanos , Masculino , Mutação
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa