Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Publication year range
1.
Reprod Biol Endocrinol ; 21(1): 53, 2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37296437

ABSTRACT

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 ß, 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.


Subject(s)
Azoospermia , Sertoli Cell-Only Syndrome , Male , Humans , Testis/metabolism , Sertoli Cell-Only Syndrome/genetics , Sertoli Cell-Only Syndrome/metabolism , Sertoli Cell-Only Syndrome/pathology , Azoospermia/pathology , Pyroptosis/genetics , Spermatogenesis/genetics , Protein Serine-Threonine Kinases/metabolism , DNA Helicases/metabolism , Transcription Factors/metabolism
2.
Acta Endocrinol (Buchar) ; 19(2): 252-255, 2023.
Article in English | MEDLINE | ID: mdl-37908894

ABSTRACT

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.

3.
Andrologia ; 54(1): e14266, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34623703

ABSTRACT

Klinefelter syndrome and monozygotic twins are both rare. The reports of monozygotic twins with Klinefelter syndrome to have undergone fertility treatment are uncommon. This case report describes a case of 30-year-old monozygotic adult twin brothers diagnosed with nonmosaic Klinefelter syndrome following the complaint of infertility. The result of semen analysis showed cryptozoospermia (very low sperm count) and azoospermia (zero sperm count) with physical findings and lifestyles being very similar. They both underwent microtesticular sperm extraction. One had successful sperm retrieval and achieved pregnancy through intracytoplasmic sperm injection, whereas the other did not. Testicular pathological findings showed Sertoli cell-only syndrome. To the best of our knowledge, this is the first report on monozygotic adult twins both of whom underwent microtesticular sperm extraction and resulted in different outcomes.


Subject(s)
Azoospermia , Klinefelter Syndrome , Adult , Azoospermia/etiology , Female , Humans , Klinefelter Syndrome/complications , Klinefelter Syndrome/diagnosis , Klinefelter Syndrome/genetics , Male , Pregnancy , Sperm Injections, Intracytoplasmic , Sperm Retrieval , Spermatozoa , Testis
4.
Genomics ; 113(4): 1845-1854, 2021 07.
Article in English | MEDLINE | ID: mdl-33865957

ABSTRACT

Studies increasingly show the involvement of circular RNAs (circRNAs) in several diseases. This study aims to explore the circRNA expression pattern in the testicular tissues of patients with Sertoli only cell syndrome (SCOS) and their potential functions. High throughput circRNA microarray analysis indicated that 399 circRNAs were upregulated and 1195 were down-regulated (fold change >2, P < 0.05) in SCOS relative to obstructive azoospermia (OA). The hsa_circRNA_101222, hsa_circRNA_001387, hsa_circRNA_001153, hsa_circRNA_101373 and hsa_circRNA_103864 were validated by qRT-PCR. Furthermore, the hosting genes of the differentially expressed circRNAs (DEcircRNAs) were enriched in biological processes related to cell cycle and intercellular communication. Also, the overlapping genes between the hosting genes of SCOS-related DEcircRNAs and those highly expressed in Sertoli cells of non-obstructive azoospermia (NOA) were enriched in immune cell development and cell communication. Taken together, aberrantly expressed circRNAs likely mediate SCOS development by regulating the function of Sertoli cells and the spermatogenic microenvironment.


Subject(s)
Azoospermia , Sertoli Cell-Only Syndrome , Azoospermia/genetics , Humans , Male , Microarray Analysis , RNA, Circular , Sertoli Cell-Only Syndrome/genetics , Spermatogenesis
5.
Am J Hum Genet ; 103(2): 200-212, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30075111

ABSTRACT

Infertility affects around 7% of men worldwide. Idiopathic non-obstructive azoospermia (NOA) is defined as the absence of spermatozoa in the ejaculate due to failed spermatogenesis. There is a high probability that NOA is caused by rare genetic defects. In this study, whole-exome sequencing (WES) was applied to two Estonian brothers diagnosed with NOA and Sertoli cell-only syndrome (SCOS). Compound heterozygous loss-of-function (LoF) variants in FANCM (Fanconi anemia complementation group M) were detected as the most likely cause for their condition. A rare maternally inherited frameshift variant p.Gln498Thrfs∗7 (rs761250416) and a previously undescribed splicing variant (c.4387-10A>G) derived from the father introduce a premature STOP codon leading to a truncated protein. FANCM exhibits enhanced testicular expression. In control subjects, immunohistochemical staining localized FANCM to the Sertoli and spermatogenic cells of seminiferous tubules with increasing intensity through germ cell development. This is consistent with its role in maintaining genomic stability in meiosis and mitosis. In the individual with SCOS carrying bi-allelic FANCM LoF variants, none or only faint expression was detected in the Sertoli cells. As further evidence, we detected two additional NOA-affected case subjects with independent FANCM homozygous nonsense variants, one from Estonia (p.Gln1701∗; rs147021911) and another from Portugal (p.Arg1931∗; rs144567652). The study convincingly demonstrates that bi-allelic recessive LoF variants in FANCM cause azoospermia. FANCM pathogenic variants have also been linked with doubled risk of familial breast and ovarian cancer, providing an example mechanism for the association between infertility and cancer risk, supported by published data on Fancm mutant mouse models.


Subject(s)
Azoospermia/genetics , DNA Helicases/genetics , Loss of Heterozygosity/genetics , Adult , Animals , Breast Neoplasms/genetics , Codon, Nonsense/genetics , Female , Frameshift Mutation/genetics , Gene Silencing/physiology , Genetic Predisposition to Disease/genetics , Homozygote , Humans , Male , Mice , Middle Aged , Ovarian Neoplasms/genetics , Pedigree , Phenotype , Spermatozoa/pathology , Testis/pathology , Exome Sequencing/methods
6.
Andrologia ; 53(3): e13936, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33427330

ABSTRACT

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.


Subject(s)
Azoospermia , Protamines/genetics , Azoospermia/genetics , Cross-Sectional Studies , Humans , Male , RNA, Messenger/genetics , Testis
7.
J Assist Reprod Genet ; 38(3): 559-572, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33428073

ABSTRACT

Almost 50% of infertility cases are due to male factors, and spermatogenesis failure is one of the most severe forms of male infertility. Sertoli cell-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 definitive diagnosis of SCOS is by diagnostic testicular biopsy. Although SCOS may be a result of Klinefelter syndrome, most of the SCOS men have a normal karyotype. Along with genetic aberrations, signaling pathways and endocrine processes might be major factors in the development of SCOS. Sperm retrieval and intracytoplasmic sperm injection (ICSI) are available treatments for SCOS. However, some SCOS patients do not have therapeutic options to help them having a biological child. This review aims to summarize our present knowledge about SCOS and to highlight the importance of future researches in the diagnosis and treatment of this disorder.


Subject(s)
Sertoli Cell-Only Syndrome/etiology , Sertoli Cell-Only Syndrome/prevention & control , Disease Management , Humans , Male , Sertoli Cell-Only Syndrome/pathology
8.
Int J Mol Sci ; 22(15)2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34360857

ABSTRACT

Herein, for the first time, the potential relationships between the cytoskeleton-associated proteins DAAM1 and PREP with different testicular disorders, such as classic seminoma (CS), Leydig cell tumor (LCT), and Sertoli cell-only syndrome (SOS), were evaluated. Six CS, two LCT, and two SOS tissue samples were obtained during inguinal exploration in patients with a suspect testis tumor based on clinical examination and ultrasonography. DAAM1 and PREP protein levels and immunofluorescent localization were analyzed. An increased DAAM1 protein level in CS and SOS as compared to non-pathological (NP) tissue was observed, while LCT showed no significant differences. Conversely, PREP protein level increased in LCT, while it decreased in CS and SOS compared to NP tissue. These results were strongly supported by the immunofluorescence staining, revealing an altered localization and signal intensity of DAAM1 and PREP in the analyzed samples, highlighting a perturbed cytoarchitecture. Interestingly, in LCT spermatogonia, a specific DAAM1 nuclear localization was found, probably due to an enhanced testosterone production, as confirmed by the increased protein levels of steroidogenic enzymes. Finally, although further studies are needed to verify the involvement of other formins and microtubule-associated proteins, this report raised the opportunity to indicate DAAM1 and PREP as new potential markers, supporting the cytoskeleton dynamics changes occurring during normal and/or pathological cell differentiation.


Subject(s)
Microfilament Proteins/metabolism , Mitochondrial Proteins/metabolism , Seminoma/metabolism , Serine Endopeptidases/metabolism , Testicular Neoplasms/metabolism , rho GTP-Binding Proteins/metabolism , Biomarkers, Tumor/metabolism , Cytoskeleton/metabolism , Humans , Male , Spermatogonia/metabolism
9.
J Urol ; 203(4): 809-816, 2020 04.
Article in English | MEDLINE | ID: mdl-31742474

ABSTRACT

PURPOSE: We sought to determine whether gene products from spermatogenic cells could be detected in Sertoli cell-only testes. Transcriptome analysis using next generation sequencing was performed using human testicular samples. MATERIALS AND METHODS: This study enrolled 20 men with nonobstructive azoospermia with a histological diagnosis of Sertoli cell-only and no germ cells on in vitro fertilization analysis and 5 with obstructive azoospermia and histologically normal spermatogenesis. Individual differences in the transcriptome of obstructive azoospermia testicular tissues generated on the Illumina® platform were compared as the number of fragments per kilobase of exon per million mapped sequence reads. We confirmed mRNA expression and targeted gene product localization by quantitative reverse transcriptase-polymerase chain reaction and immunohistochemical analysis, respectively. RESULTS: Using next generation sequencing we analyzed 5,666 of the 23,003 screened genes. As representative markers of immature germ cells, UTF1, CD9, DDX4, EPCAM, GFRA1, KIT, LIN28, DMRT, GPR125, UCHL1 and NANOG transcripts were detected in 13 Sertoli cell-only samples (65%). Reverse transcriptase-polymerase chain reaction showed significant mRNA expression of UTF1, CD9, DDX4 and KIT in Sertoli cell-only samples. Immunostaining revealed the localization of DDX4-positive cells, presumably spermatogonia, in 9 Sertoli cell-only samples (45%). These cells lacked proliferative cell nuclear antigen expression. CONCLUSIONS: A number of Sertoli cell-only testes contain immature germ cells (spermatogonia and spermatogenic stem cells) with various gene expression profiles. Sertoli cell-only is a heterogeneous pathophysiology which may be corrected by medical treatment or regenerative technologies.


Subject(s)
Azoospermia/pathology , Spermatogenesis , Spermatogonia/metabolism , Testis/cytology , Adult , Azoospermia/diagnosis , Azoospermia/therapy , Biomarkers/analysis , Biomarkers/metabolism , Cell Separation/methods , Humans , Male , Middle Aged , RNA, Messenger/metabolism , RNA-Seq , Sertoli Cells/pathology , Testis/pathology
10.
Andrologia ; 51(7): e13288, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30995700

ABSTRACT

The cause of Sertoli cell-only syndrome (SCOS), a condition in which only Sertoli cells line the seminiferous tubules in the testis, is unknown. Three microarray data sets were downloaded from public databases and were used to compare SCOS and control group. A total of 291 genes differentially expressed (Log2 |FC| ≥ 1 and adjusted p value < 0.05) in SCOS patients. Further 238 genes were significantly downregulated, and 53 genes were significantly upregulated. To identify the hub genes in the differentially expressed genes, we constructed a protein-protein interaction network, and CCNB1, CCNA2, AURKA, KIF11, CCNB2, CDC6, PRC1, NCAPG, KIF2C and PLK4 were screened from the network for the downregulated genes. Since the upregulated genes could not form a network, we concentrated on the genes with a higher fold change, and CPA3, NFIB, LONRF2, LYVE1, ATP8B4, IGF1, ITPR1 and PLAT were identified as the top 50% fold change genes in any of the three microarray data sets. Among downregulated hub genes, CDC6, CCNA2, CCNB1 and CCNB2 were involved in APC/C-mediated cell cycle progression. Among key upregulated genes, IGF1 was involved in the PI3K/AKT pathway, while the other genes have not been reported in Sertoli or Leydig cells. In conclusion, SCOS appears to be caused by disordered APC/C-mediated cell cycle progression and PI3K/AKT signalling.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Cell Cycle , Seminiferous Tubules/cytology , Sertoli Cell-Only Syndrome/pathology , Sertoli Cells/pathology , Datasets as Topic , Down-Regulation , Gene Expression Profiling , Humans , Insulin-Like Growth Factor I/metabolism , Male , Phosphatidylinositol 3-Kinases/metabolism , Protein Interaction Maps , Proto-Oncogene Proteins c-akt/metabolism , Seminiferous Tubules/pathology , Signal Transduction , Tissue Array Analysis , Up-Regulation
11.
Int J Urol ; 26(2): 292-298, 2019 02.
Article in English | MEDLINE | ID: mdl-30478911

ABSTRACT

OBJECTIVES: To detect autosomal genetic defects and to determine candidate genes in Sertoli cell-only syndrome infertile men. METHODS: Single-nucleotide polymorphism + comparative genomic hybridization microarray technology was carried out on 39 Sertoli cell-only syndrome infertile patients in the present study. Array comparative genomic hybridization compares the patient's genome against a reference genome, and identifies uncover deletions, amplifications and loss of heterozygosity. RESULTS: A link between defective spermatogenesis genes and infertility was examined, and amplifications and deletions in several genes were detected, including homeobox gene; synaptonemal complex element protein 1; collagen, type I, alpha 1; imprinted maternally expressed transcript; and potassium voltage-gated channel subfamily Q member 1. CONCLUSIONS: The present data suggest that several genes can play an important role in spermatogenesis and progression of Sertoli cell-only syndrome.


Subject(s)
Genome, Human/genetics , Sertoli Cell-Only Syndrome/genetics , Spermatogenesis/genetics , Adult , Cell-Free Nucleic Acids/genetics , Cell-Free Nucleic Acids/isolation & purification , Comparative Genomic Hybridization/methods , Disease Progression , Gene Amplification , Humans , Loss of Heterozygosity , Male , Polymorphism, Single Nucleotide , Retrospective Studies , Seminiferous Tubules/pathology , Sertoli Cell-Only Syndrome/blood , Sertoli Cell-Only Syndrome/pathology
12.
Int J Mol Sci ; 20(3)2019 Jan 22.
Article in English | MEDLINE | ID: mdl-30678285

ABSTRACT

Sertoli cell-only syndrome (SCOS) affects about 26.3⁻57.8% of azoospermic men, with their seminiferous tubules containing only Sertoli cells. Recently, it was reported that testicular biopsies from nonobstructive azoospermic (NOA) patients contained germ cells, and that sperm could be found in the tubules of 20% of SCOS patients using testicular sperm extraction technology. Since the patients without sperm in their testicular biopsies do not have therapy to help them to father a biological child, in vitro maturation of spermatogonial stem cells (SSCs) isolated from their testis is a new approach for possible future infertility treatment. Recently, the induction of human and mice SSCs proliferation and differentiation was demonstrated using different culture systems. Our group reported the induction of spermatogonial cell proliferation and differentiation to meiotic and postmeiotic stages in mice, rhesus monkeys, and prepubertal boys with cancer using 3D agar and methylcellulose (MCS) culture systems. The aim of the study was to identify the type of spermatogenic cells present in biopsies without sperm from SCOS patients, and to examine the possibility of inducing spermatogenesis from isolated spermatogonial cells of these biopsies in vitro using 3D MCS. We used nine biopsies without sperm from SCOS patients, and the presence of spermatogenic markers was evaluated by PCR and specific immunofluorescence staining analyses. Isolated testicular cells were cultured in MCS in the presence of StemPro enriched media with different growth factors and the development of colonies/clusters was examined microscopically. We examined the presence of cells from the different stages of spermatogenesis before and after culture in MCS for 3⁻7 weeks. Our results indicated that these biopsies showed the presence of premeiotic markers (two to seven markers/biopsy), meiotic markers (of nine biopsies, cAMP responsive element modulator-1 (CREM-1) was detected in five, lactate dehydrogenase (LDH) in five, and BOULE in three) and postmeiotic markers (protamine was detected in six biopsies and acrosin in three). In addition, we were able to induce the development of meiotic and/or postmeiotic stages from spermatogonial cells isolated from three biopsies. Thus, our study shows for the first time the presence of meiotic and/or postmeiotic cells in biopsies without the sperm of SCOS patients. Isolated cells from some of these biopsies could be induced to meiotic and/or postmeiotic stages under in vitro culture conditions.


Subject(s)
Sertoli Cell-Only Syndrome/pathology , Spermatozoa/cytology , Spermatozoa/pathology , Testis/cytology , Testis/pathology , Adult , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Humans , Male , Middle Aged , Seminiferous Tubules/cytology , Seminiferous Tubules/pathology , Sertoli Cells/cytology , Sertoli Cells/pathology , Spermatogenesis/physiology , Spermatogonia/cytology , Spermatogonia/pathology , Young Adult
13.
J Obstet Gynaecol ; 39(3): 434-436, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30744435

ABSTRACT

Approximately 15% of couples are infertile, with half of these cases being due to a male factor. Testis-specific cytoplasmic poly(A) polymerase beta (PAPOLB) is known to be critical for spermatogenesis. In mice, the loss of function of the Papolb gene results in the arrest of spermiogenesis and in male infertility. To analyse the role of the PAPOLB gene in human male infertility, this study investigated the relevance of this gene to human Sertoli-cell-only syndrome (SCOS) with azoospermia. Mutation analysis of the PAPOLB coding region was performed on 139 Japanese patients by PCR and direct sequence analysis. No critical mutations directly causing SCOS were detected, but three single-nucleotide polymorphisms (SNPs; SNP1 (c1101C > T), SNP2 (c1347T > C) and SNP3 (c1903C > A)) were found in the coding region. However, there were no significant associations in the allelic and genotypic distributions of these three SNPs between the SCOS and control groups (p>.05). This study suggests a lack of association of PAPOLB with azoospermia due to SCOS in humans.


Subject(s)
Azoospermia/etiology , Sertoli Cell-Only Syndrome/complications , Animals , Asian People , Azoospermia/genetics , Case-Control Studies , DNA Polymerase beta , Humans , Japan , Male , Mice , Polymorphism, Single Nucleotide , Retrospective Studies , Sertoli Cell-Only Syndrome/genetics
14.
Zhonghua Nan Ke Xue ; 24(3): 221-225, 2018 Mar.
Article in Zh | MEDLINE | ID: mdl-30161307

ABSTRACT

OBJECTIVE: To study the expression of CLAUDIN-11 in the testis tissue of non-obstructive azoospermia (NOA) patients with different severities and investigate its clinical significance. METHODS: Sixty-two NOA patients were divided into a hypospermatogenesis (HS) group (n = 30) and a Sertoli cell only syndrome (SCO) group (n =32). The expression of CLAUDIN-11 in the testicular tissue of the patients was detected by immunohistochemistry, that of CLAUDIN-11 mRNA determined by real-time fluorescence quantitative polymerase chain reaction (RT-qPCR), and the levels of serum reproductive hormones measured by chemiluminescent immunoassay. RESULTS: Immunohistochemistry showed that the expression of CLAUDIN-11 was mainly in the cytoplasm of the Sertoli cells around the seminiferous tubule wall in the HS group, but diffusely distributed in the membrane of the Sertoli cells in the SCO group. RT-qPCR revealed a significantly lower expression of CLAUDIN-11 mRNA in the HS than in the SCO group (0.008 ± 0.001 vs 0.013 ± 0.002, t = 10.616, P<0.01). The level of serum luteotropic hormone (LH) was also markedly lower in the HS than in the SCO group (ï¼»3.62 ± 1.34ï¼½ vs ï¼»4.96 ± 3.10ï¼½ IU/L, P<0.05) and so was that of follicle-stimulating hormone (FSH) (ï¼»5.36 ± 2.80ï¼½ vs ï¼»10.65 ± 9.18ï¼½ IU/L, P<0.05). CONCLUSIONS: The up-regulated expression of CLAUDIN-11 in Sertoli cells may play an important role in the development and progression of spermatogenic dysfunction in NOA patients.


Subject(s)
Azoospermia/metabolism , Claudins/metabolism , Oligospermia/metabolism , Sertoli Cell-Only Syndrome/metabolism , Testis/metabolism , Azoospermia/genetics , Follicle Stimulating Hormone/metabolism , Humans , Male , Oligospermia/genetics , RNA, Messenger/metabolism , Seminiferous Tubules/metabolism , Sertoli Cell-Only Syndrome/genetics , Sertoli Cells/metabolism , Spermatogenesis
15.
Biochim Biophys Acta ; 1863(6 Pt A): 1093-105, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26876306

ABSTRACT

The present study showed that RNA helicase Belle (DDX3) was required intrinsically for mitotic progression and survival of germline stem cells (GSCs) and spermatogonial cells in the Drosophila melanogaster testes. We found that deficiency of Belle in the male germline resulted in a strong germ cell loss phenotype. Early germ cells are lost through cell death, whereas somatic hub and cyst cell populations are maintained. The observed phenotype is related to that of the human Sertoli Cell-Only Syndrome caused by the loss of DBY (DDX3) expression in the human testes and results in a complete lack of germ cells with preservation of somatic Sertoli cells. We found the hallmarks of mitotic G2 delay in early germ cells of the larval testes of bel mutants. Both mitotic cyclins, A and B, are markedly reduced in the gonads of bel mutants. Transcription levels of cycB and cycA decrease significantly in the testes of hypomorph bel mutants. Overexpression of Cyclin B in the germline partially rescues germ cell survival, mitotic progression and fertility in the bel-RNAi knockdown testes. Taken together, these results suggest that a role of Belle in GSC maintenance and regulation of early germ cell divisions is associated with the expression control of mitotic cyclins.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Germ Cells/metabolism , RNA Helicases/metabolism , Stem Cells/metabolism , Animals , Animals, Genetically Modified , Apoptosis/genetics , Blotting, Western , Cell Division/genetics , Cyclin A/genetics , Cyclin A/metabolism , Cyclin B/genetics , Cyclin B/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Female , Male , Microscopy, Confocal , Mutation , RNA Helicases/genetics , RNA Interference , Testis/cytology , Testis/metabolism
16.
Reprod Biomed Online ; 33(2): 189-96, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27211570

ABSTRACT

Cell-free seminal mRNA (cfs-mRNA) contains testis-specific transcripts from bilateral testes. This study determined the presence of DEAD box polypeptide 4 (DDX4) in cfs-mRNA to identify and characterize the incidence of Sertoli cell-only (SCO) syndrome in men with non-obstructive azoospermia (NOA). DDX4 cfs-mRNA was determined in 315 men with NOA, and compared with testicular samples obtained by microdissection from 19 NOA patients. Karyotype and azoospermia factor microdeletion analysis were performed, and clinical features were evaluated. Negative DDX4 cfs-mRNA suggestive of SCO was found in 13.7% of NOA patients, with a similar incidence in NOA men with known genetic causes and those without known genetic causes. DDX4 cfs-mRNA was absent in 44% of SCO cases diagnosed by testicular histopathology, but present in all patients presenting with maturation arrest or hypospermatogenesis. Furthermore, 84.2% of NOA men with DDX4 cfs-positive mRNA had a DDX4-positive testicular sample. In NOA men without genetic causes, SCO patients discriminated by negative DDX4 cfs-mRNA showed different clinical features when compared with non-SCO cases. These results suggest that the evaluation of DDX4 cfs-mRNA is more accurate than testicular histopathology in discriminating SCO, and also permits the identification of a specific group of NOA men with distinct clinical features.


Subject(s)
Azoospermia/diagnosis , Azoospermia/therapy , DEAD-box RNA Helicases/genetics , Sertoli Cell-Only Syndrome/diagnosis , Sertoli Cell-Only Syndrome/therapy , Adult , Azoospermia/genetics , Azoospermia/metabolism , Germ Cells/cytology , Humans , Karyotyping , Male , Microdissection , Oligospermia/pathology , Phenotype , RNA, Messenger/metabolism , Retrospective Studies , Semen , Sertoli Cell-Only Syndrome/genetics , Sperm Retrieval , Spermatozoa/pathology , Testis/metabolism , Testis/pathology
17.
Zhonghua Nan Ke Xue ; 22(6): 516-519, 2016 Jun.
Article in Zh | MEDLINE | ID: mdl-28963840

ABSTRACT

OBJECTIVE: To investigate the 5'-flanking regulatory sequence methylation status of the Boule gene in the testis tissue of infertile men with Sertoli cell-only syndrome (SCOS). METHODS: We collected biopsy samples of the testis tissue from 12 men with obstructive azoospermia (the control group) and 15 cases of SCOS, all without varicocele, cryptorchidism, or infectious disease. We extracted genomic DNA from the testis tissue of the SCOS patients, analyzed the characteristics of the 5'-flanking regulatory sequence of the Boule gene using the bioinformatics method, and detected the methylation status of the Boule gene by sodium bisulfite sequencing. RESULTS: A CpG island was observed in the 5'-flanking regulation region of the Boule gene. The methylation level of the Boule gene was remarkably higher in the SCOS group than in the obstructive azoospermia controls (61.4% vs 21.7%, P<0.01), with significant differences in the methylation levels of 14 CpG sites, namely, -58 bp, -50 bp, -48 bp, -38 bp, -28 bp, -24 bp, -20 bp, -15 bp, -1 bp, +5 bp, +8 bp, +15 bp, +29 bp, and +58 bp. CONCLUSIONS: The methylation level of the Boule gene is significantly higher in the SCOS patients than in the obstructive azoospermia males, which suggests that the changes in Boule methylation may be associated with spermatogenic dysfunction.


Subject(s)
DNA Methylation , RNA-Binding Proteins/genetics , Sertoli Cell-Only Syndrome/genetics , Testis/metabolism , Case-Control Studies , Humans , Male , Spermatogenesis
18.
J Urol ; 194(5): 1491-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26004865

ABSTRACT

PURPOSE: Sertoli-cell only syndrome is one of the reasons for male infertility but its pathogenesis remains unclear. PRPS2, a subset of PRS, is reported to be a potential protein associated with Sertoli-cell only syndrome. In this study we further investigated the correlation between PRPS2 and Sertoli-cell only syndrome, and evaluated the effect of PRPS2 expression on apoptosis of TM4 Sertoli cells. MATERIALS AND METHODS: PRPS2 expression was detected in patients with Sertoli-cell only syndrome and normal spermatogenesis, and in Sertoli-cell only syndrome mouse models by immunohistochemistry, quantitative reverse transcription-polymerase chain reaction and Western blot. PRPS2 expression in TM4 Sertoli cells was then down-regulated and up-regulated by lentivirus vectors. The effect of PRPS2 expression on cell apoptosis and cell cycle transition was evaluated by flow cytometry. RESULTS: PRPS2 expression in patients with Sertoli-cell only syndrome was significantly greater than in those with normal spermatogenesis. A significant increase in PRPS2 expression was observed in Sertoli-cell only syndrome mouse models. PRPS2 over expression significantly inhibited cell apoptosis and promoted cell cycle transition in TM4 Sertoli cells. However, PRPS2 down-regulation showed a reverse effect. Moreover, results revealed that PRPS2 over expression inhibited cell apoptosis via the p53/Bcl-2/caspase-9/caspase-3/caspase-6/caspase-7 signaling pathway. CONCLUSIONS: PRPS2 expression correlates with Sertoli-cell only syndrome and inhibits the apoptosis of TM4 Sertoli cells via the p53/Bcl-2/caspases signaling pathway.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation , RNA/genetics , Ribose-Phosphate Pyrophosphokinase/genetics , Sertoli Cell-Only Syndrome/genetics , Sertoli Cells/pathology , Adult , Animals , Blotting, Western , Cells, Cultured , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Ribose-Phosphate Pyrophosphokinase/biosynthesis , Sertoli Cell-Only Syndrome/metabolism , Sertoli Cell-Only Syndrome/pathology , Sertoli Cells/metabolism , Young Adult
19.
Andrologia ; 47(9): 1083-5, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25395209

ABSTRACT

A loss of function of the murine Sin3A gene resulted in male infertility with Sertoli cell-only syndrome (SCOS) phenotype in mice. Here, we investigated the relevance of this gene to human male infertility with azoospermia caused by SCOS. Mutation analysis of SIN3A in the coding region was performed on 80 Japanese patients. However, no variants could be detected. This study suggests a lack of association of SIN3A gene sequence variants with azoospermia caused by SCOS in humans.


Subject(s)
Azoospermia/genetics , Repressor Proteins/genetics , Sertoli Cell-Only Syndrome/genetics , Adult , Asian People/genetics , Humans , Japan , Male , Mutation , Sin3 Histone Deacetylase and Corepressor Complex
20.
J Urol ; 192(2): 607-12, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24518766

ABSTRACT

PURPOSE: We determined whether Raman spectroscopy could identify spermatogenesis in a Sertoli-cell only rat model. MATERIALS AND METHODS: A partial Sertoli-cell only model was created using a testicular hypothermia-ischemia technique. Bilateral testis biopsy was performed in 4 rats. Raman spectra were acquired with a probe in 1 mm3 samples of testicular tissue. India ink was used to mark the site of spectral acquisition. Comparative histopathology was applied to verify whether Raman spectra were obtained from Sertoli-cell only tubules or seminiferous tubules with spermatogenesis. Principal component analysis and logistic regression were used to develop a mathematical model to evaluate the predictive accuracy of identifying tubules with spermatogenesis vs Sertoli-cell only tubules. RESULTS: Raman peak intensity changes were noted at 1,000 and 1,690 cm(-1) for tubules with spermatogenesis and Sertoli-cell only tubules, respectively. When principal components were used to predict whether seminferous tubules were Sertoli-cell only tubules or showed spermatogenesis, sensitivity and specificity were 96% and 100%, respectively. The ROC AUC to predict tubules with spermatogenesis with Raman spectroscopy was 0.98. CONCLUSIONS: Raman spectroscopy is capable of identifying seminiferous tubules with spermatogenesis in a Sertoli-cell only ex vivo rat model. Future ex vivo studies of human testicular tissue are necessary to confirm whether these findings can be translated to the clinical setting.


Subject(s)
Sertoli Cells , Spectrum Analysis, Raman , Spermatogenesis , Animals , Feasibility Studies , Male , Models, Animal , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL