Sertoli Cell-Only Syndrome

仅支持细胞综合征
  • 文章类型: Case Reports
    无精子症是男性不育的一种形式,其特征是射精中完全缺乏精子。仅支持细胞综合征(SCOS)是最严重的无精子症,在小管中没有发现生殖细胞。最近,据报道,FANCM基因变异是生精失败的新遗传原因。同时,已知FANCM变体与癌症易感性相关。我们对一名被诊断患有SCOS和健康父亲的男性患者进行了全外显子组测序。在患者中发现了两个FANCM基因的复合杂合错义突变,都是从父母那里继承的。不孕症评估后,患者被诊断为弥漫性星形细胞瘤。患者睾丸和肿瘤组织的免疫组织化学分析和适当的对照显示,第一次,不仅在星形细胞瘤中存在FANCM的细胞质而非核模式,而且在非有丝分裂神经元中也存在。在SCOS患者的睾丸组织中,细胞质抗FANCM染色强度似乎低于对照。我们的病例报告提出了一种新的可能性,即FANCM基因错义变异的不育携带者也容易发生癌症。
    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.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    精原干细胞(SSC)对于持续的精子发生和男性生育能力至关重要。小鼠SSC中选择性剪接(AS)的潜在机制仍不清楚。我们证明了SRSF1对于小鼠SSC中的基因表达和剪接至关重要。交联免疫沉淀和测序数据显示,精原细胞相关基因(例如Plzf,Id4,Setdb1,Stra8,Tial1/Tiar,Bcas2,Ddx5,Srsf10,Uhrf1和Bud31)在小鼠睾丸中被SRSF1结合。小鼠生殖细胞中Srsf1的特异性缺失会损害前体SSC的归巢,导致男性不育。整装染色数据显示成年条件敲除(cKO)小鼠睾丸中不存在生殖细胞,这表明cKO小鼠中的仅支持细胞综合征。精原细胞相关基因的表达(例如Gfra1,Pou5f1,Plzf,在cKO小鼠的睾丸中Dnd1,Stra8和Taf4b)显着降低。此外,多组学分析表明,SRSF1可能通过AS直接结合和调节Tial1/Tiar的表达来影响精原细胞的存活。此外,免疫沉淀质谱和共免疫沉淀数据显示SRSF1与RNA剪接相关蛋白(例如SART1、RBM15和SRSF10)相互作用。总的来说,我们的数据揭示了SRSF1在精原细胞存活中的关键作用,这可能提供一个框架来阐明前体SSC归巢的转录后网络的分子机制。
    Spermatogonial stem cells (SSCs) are essential for continuous spermatogenesis and male fertility. The underlying mechanisms of alternative splicing (AS) in mouse SSCs are still largely unclear. We demonstrated that SRSF1 is essential for gene expression and splicing in mouse SSCs. Crosslinking immunoprecipitation and sequencing data revealed that spermatogonia-related genes (e.g. Plzf, Id4, Setdb1, Stra8, Tial1/Tiar, Bcas2, Ddx5, Srsf10, Uhrf1, and Bud31) were bound by SRSF1 in the mouse testes. Specific deletion of Srsf1 in mouse germ cells impairs homing of precursor SSCs leading to male infertility. Whole-mount staining data showed the absence of germ cells in the testes of adult conditional knockout (cKO) mice, which indicates Sertoli cell-only syndrome in cKO mice. The expression of spermatogonia-related genes (e.g. Gfra1, Pou5f1, Plzf, Dnd1, Stra8, and Taf4b) was significantly reduced in the testes of cKO mice. Moreover, multiomics analysis suggests that SRSF1 may affect survival of spermatogonia by directly binding and regulating Tial1/Tiar expression through AS. In addition, immunoprecipitation mass spectrometry and co-immunoprecipitation data showed that SRSF1 interacts with RNA splicing-related proteins (e.g. SART1, RBM15, and SRSF10). Collectively, our data reveal the critical role of SRSF1 in spermatogonia survival, which may provide a framework to elucidate the molecular mechanisms of the posttranscriptional network underlying homing of precursor SSCs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    高通量单细胞RNA测序(scRNA-seq)广泛用于精子发生。然而,它只显示了生殖细胞和体细胞中的短读数,限制了新转录本和基因的发现。
    目的:这项研究显示了梗阻性无精子症(OA)和仅支持细胞(SCO)患者精子发生的长阅读转录图。
    方法:从OA和NOA患者的睾丸活检组织中分离单细胞。通过比较Pacbio长读数单细胞测序(OAn=3,NOAn=3)与短读数scRNA-seq(OAn=6,NOAn=6)来鉴定细胞培养。根据已知标记对10种生殖细胞类型和8种体细胞类型进行分类。
    方法:Pacbiolong读取单细胞测序,短读scRNA-seq,聚合酶链反应。
    结果:总共130,426份长读转录本(100,517份新转录本和29,909份已知转录本)和49,508份长读转录本(26,002份新转录本,和23,506已知转录物)已在OA和NOA患者中检测到,分别。此外,在OA和NOA患者中鉴定出36,373和1642个新基因,分别。重要的是,在正常精子发生过程中,在生殖细胞和气孔细胞中检测到长阅读转录本的特异性表达。
    结论:我们已经确定了OA和NOA的全长转录本,发现了新的基因.此外,检测到特异性表达的全长转录本,转录本的基因组结构被定位在不同的细胞类型中。这些发现可能为人类精子发生和男性不育的治疗提供有价值的信息。
    BACKGROUND: High-throughput single-cell RNA sequencing (scRNA-seq) is widely used in spermatogenesis. However, it only reveals short reads in germ and somatic cells, limiting the discovery of novel transcripts and genes.
    OBJECTIVE: This study shows the long-read transcriptional landscape of spermatogenesis in obstructive azoospermia (OA) and Sertoli cell-only patients.
    METHODS: Single cells were isolated from testicular biopsies of OA and non-obstructive azoospermia (NOA) patients. Cell culture was identified by comparing PacBio long-read single-cell sequencing (OA n = 3, NOA n = 3) with short-read scRNA-seq (OA n = 6, NOA n = 6). Ten germ cell types and eight somatic cell types were classified based on known markers.
    METHODS: PacBio long-read single-cell sequencing, short-read scRNA-seq, polymerase chain reaction.
    RESULTS: A total of 130 426 long-read transcripts (100 517 novel transcripts and 29 909 known transcripts) and 49 508 long-read transcripts (26 002 novel transcripts and 23 506 known transcripts) have been detected in OA and NOA patients, respectively. Moreover, 36 373 and 1642 new genes are identified in OA and NOA patients, respectively. Importantly, specific expressions of long-read transcripts were detected in germ and stomatic cells during normal spermatogenesis.
    CONCLUSIONS: We have identified total full-length transcripts in OA and NOA, and new genes were found. Furthermore, specific expressed full-length transcripts were detected, and the genomic structure of transcripts was mapped in different cell types. These findings may provide valuable information on human spermatogenesis and the treatment of male infertility.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Meta-Analysis
    目的:关于AZFc微缺失对精子回收和ART结局的影响的现有证据有限且本质上相互矛盾。目的:调查AZFc微缺失是否会影响辅助生殖技术(ART)结局数据来源:搜索电子数据库,以调查从开始到2023年4月AZFc微缺失与ART结局的相关性研究。研究选择和综合:分析了病例对照研究。研究人群包括有和没有AZFc微缺失的不育男性。使用纽卡斯尔-渥太华量表评估研究质量。计算这两类男性的总效应大小(比值比[OR]和95%置信区间[CI])。
    结果:主要结局是成功取出精子,次要结局是ART结局。
    结果:(S):没有病例对照研究报告AZFa和AZFb缺失男性的精子获取率和ART结果。根据3807名男性的数据,AZFc微缺失男性的精子回收率高于未缺失男性[OR=1.82,95%CI0.97,3.41],但差异无统计学意义(P=0.06).受精率显著降低(OR=0.61,95%CI0.50,0.74),与无缺失男性相比,有AZFc缺失男性的临床妊娠率(OR=0.61,95%CI0.42,0.89)和活产率(OR=0.54,95%CI0.40,0.72).胚胎卵裂率无统计学差异,囊胚形成,优质胚胎,两组之间的植入和流产。关于纠正女性因素,受精率(OR=0.76,95%CI0.71,0.82),卵裂率(OR=0.54,95%CI0.41,0.72),AZFc缺失男性的临床妊娠率(OR=0.39,95%CI0.30,0.52)和活产率(OR=0.48,95%CI0.35,0.65)显著低于对照组.
    结论:(S)和相关性:AZFc微缺失与ART的不良结局显著相关。需要进一步的研究来准确阐明AZFc微缺失对ART结果的影响。
    To investigate whether Azoospermia Factor c (AZFc) microdeletions affect Assisted Reproductive Technology (ART) outcomes.
    Systematic review and meta-analysis.
    Not applicable.
    Infertile men with and without AZFc microdeletions.
    Electronic databases were searched for case-control studies reporting sperm retrieval rates and outcomes of ART in infertile men with and without AZFc microdeletions from inception to April 2023. Study quality was assessed using the Newcastle-Ottawa Scale. Summary effect sizes (odds ratio [OR] with 95% confidence interval [CI]) were calculated for both categories of infertile men.
    The primary outcome was successful sperm retrieval and the secondary outcomes were outcomes of ART.
    Case-control studies reporting sperm retrieval rates and ART outcomes in men with AZFa and AZFb deletions were unavailable. On the basis of the data from 3,807 men, sperm retrieval rates were found to be higher in men with AZFc microdeletions compared to their non-deleted counterparts [OR = 1.82, 95% CI 0.97, 3.41], but the difference was not statistically significant. A significantly lower fertilization rate (OR = 0.61, 95% CI [0.50, 0.74]), clinical pregnancy rate (OR = 0.61, 95% CI [0.42, 0.89]), and live birth rate (OR = 0.54, 95% CI [0.40, 0.72]) were observed in men with AZFc deletions compared with men without deletions. There was no statistically significant difference in rates of embryo cleavage, blastocyst formation, good-quality embryos, implantation, and miscarriage between the two groups. On correcting for female factors, the fertilization rate (OR = 0.76, 95% CI [0.71, 0.82]), cleavage rate (OR = 0.54, 95% CI [0.41, 0.72]), clinical pregnancy rate (OR = 0.39, 95% CI [0.30, 0.52]), and live birth rate (OR = 0.48, 95% CI [0.35, 0.65]) were significantly lower in men with AZFc deletions compared with controls.
    Presence of AZFc microdeletions adversely affects outcomes of ART in infertile men. Further in-depth studies delineating the role of the AZF genes in embryonic development are necessary to understand the full-impact of this finding.
    CRD42022311738.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Case Reports
    睾丸间质细胞瘤(LCT)在成人中非常罕见。它仅占总睾丸肿瘤的1%。LCT可以产生类固醇激素,如雌激素,黄体酮,和睾丸激素。在生精小管中发现了支持细胞,它们是血睾丸屏障的一部分.仅支持细胞综合征(SCOS)也称为生殖细胞发育不全,其特征是无精子症,其中睾丸活检的生精小管仅衬有支持细胞。在正常T和LH的情况下,SCOS中预期的激素谱是FSH增加。LCT中的预期激素谱是增加的/正常的FSH和LH,具有增加的T或E2。一位患者出现在我们的诊所,他的右睾丸中有一个界限分明的肿块,并接受了根治性睾丸切除术。肿瘤标志物阴性。在精子图中检测到无精子症。T和E2正常,FSH,LH高。行根治性睾丸切除术。病理结果报告了LCT和SCOS的组合。在LCTs中经常遇到继发于高雄激素水平的无精子症病例。正如我们所介绍的那样,两种不同的睾丸病理可能同时出现,并产生意想不到的荷尔蒙图片。这种情况可能导致实验室掩盖临床事实。
    Leydig Cell Tumor (LCT) is very rare in adults. It constitutes only 1% of total testicular tumors. LCTs can produce steroid hormones such as estrogen, progesterone, and testosterone. Sertoli cells are found in seminiferous tubules, they are part of the blood-testis barrier. Sertoli Cells Only Syndrome (SCOS) also known as germ cell aplasia is characterized by azoospermia in which the seminiferous tubules of testicular biopsy are lined only with Sertoli cells. The expected hormone profile in SCOS is increased FSH with normal T and LH. The expected hormone profile in LCT is increased/normal FSH and LH with increased T or E2. A patient presented to our clinic with a well-circumscribed mass in his right testicle and underwent radical orchiectomy. Tumor markers were negative. Azoospermia was detected in the spermiogram. T and E2 were normal, FSH, and LH were high. Right radical orchiectomy was performed. A combination of LCT and SCOS were reported in pathology results. Azoospermia cases secondary to high androgen levels are frequently encountered in LCTs. As in the case we have presented, two different testicular pathologies may present at the same time and create an unexpected hormonal picture. Such situations can cause the laboratory to mask the clinical truth.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    测试Y染色体的无精子症因子(AZF)缺失是无精子症和严重少精子症男性诊断检查的关键组成部分。2013年欧洲男性科学院(EAA)和EMQNCIC(以前称为欧洲分子遗传学质量网络)实验室指南的修订版总结了最近的临床相关进展,并提供了两个组织联合提供的外部质量评估计划的最新结果。基本的多重PCR反应以及缺失延伸分析仍然是检测和正确解释AZF缺失的金标准方法。最近的数据导致了sY84引物序列的更新,以及对先前被认为是AZFa和AZFb缺失断点的可互换边界标记的改进。更具体地说,sY83和sY143不再推荐用于删除扩展分析,分别留下sY1064和sY1192,作为首选标记。尽管转型,目前在几个国家进行,基于认证试剂盒的诊断,应该指出的是,由于测试标记的数量过多,因此不推荐使用许多这些商业产品,这些目前都没有,据我们所知,根据新的首选标记进行缺失扩展分析。gr/gr部分AZFc缺失仍然是精子产生受损的群体特异性风险因素和睾丸生殖细胞肿瘤的诱发因素。此删除类型的测试是,和以前一样,由诊断实验室和转诊临床医生自行决定。强烈鼓励每年参与外部质量控制计划,EMQN/EAA计划22年的经验清楚地表明,诊断错误急剧下降,报告实践有所改善。
    Testing for AZoospermia Factor (AZF) deletions of the Y chromosome is a key component of the diagnostic workup of azoospermic and severely oligozoospermic men. This revision of the 2013 European Academy of Andrology (EAA) and EMQN CIC (previously known as the European Molecular Genetics Quality Network) laboratory guidelines summarizes recent clinically relevant advances and provides an update on the results of the external quality assessment program jointly offered by both organizations. A basic multiplex PCR reaction followed by a deletion extension analysis remains the gold-standard methodology to detect and correctly interpret AZF deletions. Recent data have led to an update of the sY84 reverse primer sequence, as well as to a refinement of what were previously considered as interchangeable border markers for AZFa and AZFb deletion breakpoints. More specifically, sY83 and sY143 are no longer recommended for the deletion extension analysis, leaving sY1064 and sY1192, respectively, as first-choice markers. Despite the transition, currently underway in several countries, toward a diagnosis based on certified kits, it should be noted that many of these commercial products are not recommended due to an unnecessarily high number of tested markers, and none of those currently available are, to the best of our knowledge, in accordance with the new first-choice markers for the deletion extension analysis. The gr/gr partial AZFc deletion remains a population-specific risk factor for impaired sperm production and a predisposing factor for testicular germ cell tumors. Testing for this deletion type is, as before, left at the discretion of the diagnostic labs and referring clinicians. Annual participation in an external quality control program is strongly encouraged, as the 22-year experience of the EMQN/EAA scheme clearly demonstrates a steep decline in diagnostic errors and an improvement in reporting practice.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    仅支持细胞综合征(SCOS),严重的睾丸生精失败,其特征在于完全不存在雄性生殖细胞。为了更好地扩展对SCOS潜在分子机制的理解,我们使用来自基因表达Omnibus(GEO)和ArrayExpress数据库的微阵列数据集来确定差异表达基因(DEGs)。此外,进行了功能富集分析,包括基因本体论(GO)和京都基因和基因组百科全书(KEGG)。蛋白质-蛋白质相互作用(PPI)网络,模块,和miRNA-mRNA调控网络的构建和分析,并对hub基因进行验证。总共确定了601个共享DEG,包括416个下调基因和185个上调基因。富集分析的结果表明,共享的DEG大多在有性生殖中富集,生殖过程,雄配子一代,免疫反应,和免疫相关途径。此外,六个hub基因(CCNA2、CCNB2、TOP2A、通过使用cytoHubba和MCODE插件从PPI网络中选择CDC20,BUB1和BUB1B)。如微阵列数据所示,与正常精子发生对照相比,SCOS患者的hub基因表达水平显着降低,单细胞转录组数据,和临床样本水平。此外,通过miRNA-mRNA网络构建预测潜在的miRNA.这些hub基因和miRNA可用作可能与SCOS相关的潜在生物标志物。然而,目前尚未证明这些生物标志物的差异表达是SCOS的分子发病机制。我们的发现表明,这些生物标志物可以作为诊断目标的临床工具,并且可能从睾丸生殖细胞的角度对SCOS的精子发生产生一些影响。
    Sertoli cell-only syndrome (SCOS), a severe testicular spermatogenic failure, is characterized by total absence of male germ cells. To better expand the understanding of the potential molecular mechanisms of SCOS, we used microarray datasets from the Gene Expression Omnibus (GEO) and ArrayExpress databases to determine the differentially expressed genes (DEGs). In addition, functional enrichment analysis including the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) was performed. Protein-protein interaction (PPI) networks, modules, and miRNA-mRNA regulatory networks were constructed and analyzed and the validation of hub genes was performed. A total of 601 shared DEGs were identified, including 416 down-regulated and 185 up-regulated genes. The findings of the enrichment analysis indicated that the shared DEGs were mostly enriched in sexual reproduction, reproductive process, male gamete generation, immune response, and immunity-related pathways. In addition, six hub genes (CCNA2, CCNB2, TOP2A, CDC20, BUB1, and BUB1B) were selected from the PPI network by using the cytoHubba and MCODE plug-ins. The expression levels of the hub genes were significantly decreased in patients with SCOS compared to that in normal spermatogenesis controls as indicated by the microarray data, single-cell transcriptomic data, and clinical sample levels. Furthermore, the potential miRNAs were predicted via the miRNA-mRNA network construction. These hub genes and miRNAs can be used as potential biomarkers that may be related to SCOS. However, it has not been proven that the differential expression of these biomarkers is the molecular pathogenesis mechanisms of SCOS. Our findings suggest that these biomarkers can be serve as clinical tool for diagnosis targets and may have some impact on the spermatogenesis of SCOS from a testicular germ cell perspective.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:仅支持细胞综合征(SCOS)是非梗阻性无精子症最严重的病理类型。最近,已经确定了几个与SCOS相关的基因,包括FANCM,TEX14、NR5A1、NANOS2、PLK4、WNK3和FANCA,但不能完全解释SCOS的发病机制。本研究试图通过睾丸组织RNA测序来解释SCOS中的精子发生功能障碍,为SCOS的诊断和治疗提供新的靶点。
    方法:我们基于RNA测序分析了9例SCOS患者和3例梗阻性无精子症和正常精子发生患者的差异表达基因(DEGs)。我们使用ELISA和免疫组织化学进一步探索了鉴定的基因。
    结果:总计,在SCOS样品中表达了9406个DEGs(Log2|FC|≥1;调整后的P值<0.05),并鉴定了21个hub基因。发现了三个上调的核心基因,包括CASP4、CASP1和PLA2G4A。因此,我们推测CASP1和CASP4介导的睾丸细胞焦亡可能参与了SCOS的发生和发展。ELISA证实,SCOS患者睾丸中的CASP1和CASP4活性明显高于精子发生正常患者。免疫组织化学结果显示,CASP1和CASP4在正常生精组中主要表达于生精细胞核,Sertoli,和间质细胞。由于精原细胞和精母细胞的丢失,SCOS组的CASP1和CASP4主要在支持细胞和间质细胞核中表达。SCOS患者睾丸中CASP1和CASP4的表达水平明显高于正常生精患者。此外,SCOS患者睾丸中的焦亡相关蛋白GSDMD和GSDME也明显高于对照组.ELISA还显示炎症因子(IL-1β,IL-18,LDH,和ROS)在SCOS组中显著增加。
    结论:第一次,我们发现,在SCOS患者的睾丸中,细胞焦亡相关基因和关键标志物显著增加.我们还在SCOS中观察到许多炎症和氧化应激反应。因此,我们认为,CASP1和CASP4介导的睾丸细胞焦亡可能参与了SCOS的发生和发展。
    BACKGROUND: Sertoli cell-only syndrome (SCOS) is the most serious pathological type of non-obstructive azoospermia. Recently, several genes related to SCOS have been identified, including FANCM, TEX14, NR5A1, NANOS2, PLK4, WNK3, and FANCA, but they cannot fully explain the pathogenesis of SCOS. This study attempted to explain spermatogenesis dysfunction in SCOS through testicular tissue RNA sequencing and to provide new targets for SCOS diagnosis and therapy.
    METHODS: We analyzed differentially expressed genes (DEGs) based on RNA sequencing of nine patients with SCOS and three patients with obstructive azoospermia and normal spermatogenesis. We further explored the identified genes using ELISA and immunohistochemistry.
    RESULTS: In total, 9406 DEGs were expressed (Log2|FC|≥ 1; adjusted P value < 0.05) in SCOS samples, and 21 hub genes were identified. Three upregulated core genes were found, including CASP4, CASP1, and PLA2G4A. Thus, we hypothesized that testis cell pyroptosis mediated by CASP1 and CASP4 might be involved in SCOS occurrence and development. ELISA verified that CASP1 and CASP4 activities in the testes of patients with SCOS were significantly higher than those in patients with normal spermatogenesis. Immunohistochemical results showed that CASP1 and CASP4 in the normal spermatogenesis group were mainly expressed in the nuclei of spermatogenic, Sertoli, and interstitial cells. CASP1 and CASP4 in the SCOS group were mainly expressed in the nuclei of Sertoli and interstitial cells because of the loss of spermatogonia and spermatocytes. CASP1 and CASP4 expression levels in the testes of patients with SCOS were significantly higher than those in patients with normal spermatogenisis. Furthermore, the pyroptosis-related proteins GSDMD and GSDME in the testes of patients with SCOS were also significantly higher than those in control patients. ELISA also showed that inflammatory factors (IL-1 β, IL-18, LDH, and ROS) were significantly increased in the SCOS group.
    CONCLUSIONS: For the first time, we found that cell pyroptosis-related genes and key markers were significantly increased in the testes of patients with SCOS. We also observed many inflammatory and oxidative stress reactions in SCOS. Thus, we propose that testis cell pyroptosis mediated by CASP1 and CASP4 could participate in SCOS occurrence and development.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Meta-Analysis
    背景:目前还没有系统评价和荟萃分析来分析和总结挽救性微解剖睾丸精子提取(mTESE)中精子提取成功的预测因素。
    目的:我们旨在研究预测初次mTESE或常规睾丸精子提取(cTESE)失败的NOA患者mTESE挽救结果的因素。
    方法:我们在PubMed,WebofScience,EMBASE,和Cochrane图书馆提供的文献描述了在2022年6月之前发表的初始mTESE或cTESE未通过后接受抢救mTESE的NOA患者的特征。
    结果:本荟萃分析包括4项回顾性研究,对332例初次mTESE失败的NOA患者和3项回顾性研究,对177例cTESE失败的NOA患者。结果如下:在首次手术为mTESE的NOA患者中,年轻患者(SMD:-0.28,95%CI:-0.55至-0.01)和双侧睾丸体积(TV)较小的患者(SMD:-0.55,95%CI:-0.95至-0.15),较低水平的FSH(SMD:-0.86,95%CI:-1.18至-0.54)和LH(SMD:-0.68,95%CI:-1.16至-0.19),睾丸组织学类型为精子发生障碍(HS)(OR:3.52,95%CI:1.30至9.53)更有可能成功回收精子。而仅支持细胞综合征(SCOS)(OR:0.41,95%CI:0.24至0.73)的患者在挽救性mTESE中再次失败的可能性更大。此外,在初次cTESE失败后接受抢救mTESE的患者中,那些睾丸组织学类型的精子发生不足(HS)(OR:30.35,95%CI:8.27至111.34)更有可能成功,而那些成熟停滞(MA)(OR:0.39,95%CI:0.18至0.83)的患者很少受益。
    结论:我们发现年龄,电视,FSH,LH,HS,SCOS和MA是救助mTESE的有价值的预测因子,这将有助于男科医生的临床决策,并最大限度地减少对患者不必要的伤害。本文受版权保护。保留所有权利。
    BACKGROUND: There has been no systematic review and meta-analysis to analyze and summarize the predictive factors of successful sperm extraction in salvage microdissection testicular sperm extraction.
    OBJECTIVE: We aimed to investigate the factors predicting the result of salvage microdissection testicular sperm extraction in patients with non-obstructive azoospermia who failed the initial microdissection testicular sperm extraction or conventional testicular sperm extraction.
    METHODS: We conducted a systematic literature search in PubMed, Web of Science, EMBASE, and the Cochrane Library for literature that described the characteristics of patients with non-obstructive azoospermia who underwent salvage microdissection testicular sperm extraction after failing the initial microdissection testicular sperm extraction or conventional testicular sperm extraction published prior to June 2022.
    RESULTS: This meta-analysis included four retrospective studies with 332 patients with non-obstructive azoospermia who underwent a failed initial microdissection testicular sperm extraction and three retrospective studies with 177 non-obstructive azoospermia patients who underwent a failed conventional testicular sperm extraction. The results were as follows: among non-obstructive azoospermia patients whose first surgery was microdissection testicular sperm extraction, younger patients (standard mean difference: -0.28, 95% confidence interval [CI]: -0.55 to -0.01) and those with smaller bilateral testicular volume (standard mean difference: -0.55, 95% CI: -0.95 to -0.15), lower levels of follicle-stimulating hormone (standard mean difference: -0.86, 95% CI: -1.18 to -0.54) and luteinizing hormone (standard mean difference: -0.68, 95% CI: -1.16 to -0.19), and whose testicular histological type was hypospermatogenesis (odds ratio: 3.52, 95% CI: 1.30-9.53) were more likely to retrieve spermatozoa successfully, while patients with Sertoli-cell-only syndrome (odds ratio: 0.41, 95% CI: 0.24-0.73) were more likely to fail again in salvage microdissection testicular sperm extraction. Additionally, in patients who underwent salvage microdissection testicular sperm extraction after a failed initial conventional testicular sperm extraction, those with testicular histological type of hypospermatogenesis (odds ratio: 30.35, 95% CI: 8.27-111.34) were more likely to be successful, while those with maturation arrest (odds ratio: 0.39, 95% CI: 0.18-0.83) rarely benefited.
    CONCLUSIONS: We found that age, testicular volume, follicle-stimulating hormone, luteinizing hormone, hypospermatogenesis, Sertoli-cell-only syndrome, and maturation arrest were valuable predictors of salvage microdissection testicular sperm extraction, which will assist andrologists in clinical decision-making and minimize unnecessary injury to patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    仅支持细胞综合征(SCOS)是导致男性不育的一种睾丸病理衰竭,没有有效的治疗策略。可用于此条件。此外,其发展的分子机制仍然未知。我们基于从基因表达合成数据库获得的睾丸组织样品的四个数据集,将DExD/H-Box解旋酶58(DDX58)鉴定为SCOS中的关键基因。DDX58在SCOS睾丸支持细胞中显著上调。此外,DDX58的高表达与几种睾丸炎症因子的表达呈正相关,如IL-1β,IL-18和IL-6。有趣的是,在D-半乳糖(D-gal)刺激的TM4细胞损伤模型中可以诱导DDX58。而DDX58的沉默抑制了D-gal介导的p65表达,炎性细胞因子释放,增长停滞。机械上,我们发现DDX58作为RNA结合蛋白,通过促进mRNA稳定性来增强p65表达。此外,p65基因沉默降低了D-gal诱导的细胞中炎症细胞因子的表达和细胞生长的抑制。总之,我们的研究结果表明,DDX58通过稳定p65mRNA促进SCOS支持细胞的炎症反应和生长停滞.因此,DDX58/p65调节轴可能是SCOS的治疗靶点.
    Sertoli cell -only syndrome (SCOS) is a type of testicular pathological failure that causes male infertility and no effective treatment strategy, is available for this condition. Moreover, the molecular mechanism underlying its development remains unknown. We identified DExD/H-Box helicase 58 (DDX58) as a key gene in SCOS based on four datasets of testicular tissue samples obtained from the Gene Expression Synthesis database. DDX58 was significantly upregulated in SCOS testicular Sertoli cells. Moreover, high expression of DDX58 was positively correlated with the expression of several testicular inflammatory factors, such as IL -1β, IL-18, and IL-6. Interestingly, DDX58 could be induced in the D-galactose (D-gal)-stimulated TM4 cell injury model. Whereas silencing of DDX58 inhibited D-gal -mediated p65 expression, inflammatory cytokine release, and growth arrest. Mechanistically, we found that DDX58 acts as an RNA-binding protein, which enhances p65 expression by promoting mRNA stability. Furthermore, p65 gene silencing decreased the expression of inflammatory cytokines and inhibition of cell growth in D-gal-induced cells. In conclusion, our findings demonstrate that DDX58 promotes inflammatory responses and growth arrest in SCOS Sertoli cells by stabilizing p65 mRNA. Accordingly, the DDX58/p65 regulatory axis might be a therapeutic target for SCOS.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号