Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Am J Hum Genet ; 110(3): 516-530, 2023 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-36796361

RESUMO

Primate-specific genes (PSGs) tend to be expressed in the brain and testis. This phenomenon is consistent with brain evolution in primates but is seemingly contradictory to the similarity of spermatogenesis among mammals. Here, using whole-exome sequencing, we identified deleterious variants of X-linked SSX1 in six unrelated men with asthenoteratozoospermia. SSX1 is a PSG expressed predominantly in the testis, and the SSX family evolutionarily expanded independently in rodents and primates. As the mouse model could not be used for studying SSX1, we used a non-human primate model and tree shrews, which are phylogenetically similar to primates, to knock down (KD) Ssx1 expression in the testes. Consistent with the phenotype observed in humans, both Ssx1-KD models exhibited a reduced sperm motility and abnormal sperm morphology. Further, RNA sequencing indicated that Ssx1 deficiency influenced multiple biological processes during spermatogenesis. Collectively, our experimental observations in humans and cynomolgus monkey and tree shrew models highlight the crucial role of SSX1 in spermatogenesis. Notably, three of the five couples who underwent intra-cytoplasmic sperm injection treatment achieved a successful pregnancy. This study provides important guidance for genetic counseling and clinical diagnosis and, significantly, describes the approaches for elucidating the functions of testis-enriched PSGs in spermatogenesis.


Assuntos
Astenozoospermia , Tupaia , Animais , Masculino , Macaca fascicularis , Primatas , Sêmen , Motilidade dos Espermatozoides , Tupaiidae
2.
Clin Genet ; 105(4): 440-445, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38148155

RESUMO

Nonobstructive azoospermia (NOA), the most severe manifestation of male infertility, lacks a comprehensive understanding of its genetic etiology. Here, a bi-allelic loss-of-function variant in REC114 (c.568C > T: p.Gln190*) were identified through whole exome sequencing (WES) in a Chinese NOA patient. Testicular histopathological analysis and meiotic chromosomal spread analysis were conducted to assess the stage of spermatogenesis arrested. Co-immunoprecipitation (Co-IP) and Western blot (WB) were used to investigate the influence of variant in vitro. In addition, our results revealed that the variant resulted in truncated REC114 protein and impaired interaction with MEI4, which was essential for meiotic DNA double-strand break (DSB) formation. As far as we know, this study presents the first report that identifies REC114 as the causative gene for male infertility. Furthermore, our study demonstrated indispensability of the REC114-MEI4 complex in maintaining DSB homoeostasis, and highlighted that the disruption of the complex due to the REC114 variant may underline the mechanism of NOA.


Assuntos
Azoospermia , Infertilidade Masculina , Humanos , Masculino , Azoospermia/genética , Azoospermia/patologia , Perda de Heterozigosidade , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Testículo/patologia , Meiose/genética , Proteínas de Ciclo Celular/genética
3.
J Hum Genet ; 68(6): 383-392, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36759719

RESUMO

Non-obstructive azoospermia (NOA) is characterized by the failure of sperm production due to testicular disorders and represents the most severe form of male infertility. Growing evidences have indicated that gene defects could be the potential cause of NOA via genome-wide sequencing approaches. Here, bi-allelic deleterious variants in meiosis inhibitor protein 1 (MEI1) were identified by whole-exome sequencing in four Chinese patients with NOA. Testicular pathologic analysis and immunohistochemical staining revealed that spermatogenesis is arrested at spermatocyte stage, with defective programmed DNA double-strand breaks (DSBs) homoeostasis and meiotic chromosome synapsis in patients carrying the variants. In addition, our results showed that one missense variant (c.G186C) reduced the expression of MEI1 and one frameshift variant (c.251delT) led to truncated proteins of MEI1 in in vitro. Furthermore, the missense variant (c.T1585A) was assumed to affect the interaction between MEI1 and its partners via bioinformatic analysis. Collectively, our findings provide direct genetic and functional evidences that bi-allelic variants in MEI1 could cause defective DSBs homoeostasis and meiotic chromosome synapsis, which subsequently lead to meiosis arrest and male infertility. Thus, our study deepens our knowledge of the role of MEI1 in male fertility and provides a novel insight to understand the genetic aetiology of NOA.


Assuntos
Azoospermia , Infertilidade Masculina , Humanos , Masculino , Azoospermia/genética , Azoospermia/patologia , Sêmen , Proteínas/genética , Infertilidade Masculina/genética , Meiose/genética , Proteínas de Ciclo Celular/genética
4.
J Hum Genet ; 68(11): 729-735, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37365320

RESUMO

Meiotic arrest is a common pathologic phenotype of non-obstructive azoospermia (NOA), yet its genetic causes require further investigation. Meiotic nuclear divisions 1 (MND1) has been proved to be indispensable for meiotic recombination in many species. To date, only one variant of MND1 has been reported associated with primary ovarian insufficiency (POI), yet there has been no report of variants in MND1 associated with NOA. Herein, we identified a rare homozygous missense variant (NM_032117:c.G507C:p.W169C) of MND1 in two NOA-affected patients from one Chinese family. Histological analysis and immunohistochemistry demonstrated meiotic arrest at zygotene-like stage in prophase I and lack of spermatozoa in the proband's seminiferous tubules. In silico modeling demonstrated that this variant might cause possible conformational change in the leucine zippers 3 with capping helices (LZ3wCH) domain of MND1-HOP2 complex. Altogether, our study demonstrated that the MND1 variant (c.G507C) is likely responsible for human meiotic arrest and NOA. And our study provides new insights into the genetic etiology of NOA and mechanisms of homologous recombination repair in male meiosis.

5.
Clin Genet ; 104(5): 577-581, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37337432

RESUMO

Genetic causation for the majority of non-obstructive azoospermia (NOA) remains unclear. Mutations in synaptonemal complex (SC)-associated genes could cause meiotic arrest and NOA. Previous studies showed that heterozygous truncating variants in SYCP2 encoding a protein essential for SC formation, are associated with non-obstructive azoospermia and severe oligozoospermia. Herein, we showed a homozygous loss-of-function variant in SYCP2 (c.2689_2690insT) in an NOA-affected patient. And this variant was inherited from heterozygous parental carriers by natural reproduction. HE, IF, and meiotic chromosomal spread analyses demonstrated that spermatogenesis was arrested at the zygotene stage in the proband with NOA. Thus, this study revealed that SYCP2 associated with NOA segregates in an autosomal recessive inheritance pattern, rather than an autosomal dominant pattern. Furthermore, our study expanded the knowledge of variants in SYCP2 and provided new insight into understanding the genetic etiology of NOA.


Assuntos
Azoospermia , Masculino , Humanos , Azoospermia/genética , Mutação da Fase de Leitura , Mutação , Espermatogênese/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ciclo Celular/genética
6.
Mol Hum Reprod ; 28(7)2022 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-35674372

RESUMO

KASH5 is an essential component of the LINC (linker of the nucleoskeleton and cytoskeleton) complex that regulates chromosome movements and nuclear envelope (NE) remodeling in mouse spermatocytes during meiosis prophase I, but its expression and function in human cells, as well as its association with male infertility are largely unknown. In this study, a novel heterozygous copy number variation (CNV) (seq [GRCh37] del(19) (19q13.33) chr19: g.49894043-49903011del) and a heterozygous loss of function variant (NM_144688: c.979_980del: p.R327Sfs*21) in human KASH5 were identified in a non-obstructive azoospermia (NOA)-affected patient and in his infertile sister by whole-exome sequencing and CNV array. Spermatogenesis in the proband was arrested at zygotene-like stage with a deficiency in homolog pairing and synapsis. KASH5 protein expression in human spermatocytes was evaluated and reported first in this study. Single-cell RNA sequencing demonstrated that the LINC complex and associated genes in human and mouse shared a similar expression pattern, indicating a conserved mechanism in the regulation of chromosome movements and NE remodeling. Kash5 knockout mouse displayed similar phenotypes, including a meiotic arrest at a zygotene-like stage and impaired pairing and synapsis. Collectively, we have identified novel rare variants within human KASH5 in patients with NOA and meiosis arrest. Our study expands the knowledge of KASH5 and associated proteins in regulating human meiosis prophase I progress and provides new insight into the genetic etiology of NOA.


Assuntos
Azoospermia , Animais , Humanos , Masculino , Camundongos , Azoospermia/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Variações do Número de Cópias de DNA , Meiose/genética , Proteínas/genética
7.
Clin Genet ; 101(5-6): 507-516, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35285020

RESUMO

The genetic causes of idiopathic premature ovarian insufficiency (POI) and nonobstructive azoospermia (NOA) remain unclear. We performed whole-exome sequencing (WES) in members of a consanguineous family with two POI and two NOA patients to screen for potential pathogenic variants for familial POI and NOA. And a homozygous variant in SPATA22 (c.400C>T:p.R134X) was identified. Histological analysis and spermatocyte spreading assay demonstrated that the spermatogenesis was arrested at a zygotene-like stage in the proband with NOA. The candidate gene was further screened in the in-house WES database of idiopathic POI-affected patients. One additional compound heterozygous variant in SPATA22 (c.900+1G>A and c.31C>T:p.R11X) was found in one patient with sporadic POI and validated by minigene assay. Thus, this is the first report identifying SPATA22 as the causative gene for human POI. Combined with the observations in the familial patient with NOA, our findings highlighted the essential role of meiotic HR genes in gametogenesis and gonadal function maintenance.


Assuntos
Azoospermia , Insuficiência Ovariana Primária , Azoospermia/genética , Azoospermia/patologia , Proteínas de Ciclo Celular/genética , Feminino , Humanos , Masculino , Insuficiência Ovariana Primária/genética , Sequenciamento do Exoma
8.
Reprod Biol Endocrinol ; 20(1): 21, 2022 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-35090489

RESUMO

BACKGROUND: Non-obstructive azoospermia (NOA) is one of the most severe type in male infertility, and the genetic causes of NOA with meiotic arrest remain elusive. METHODS: Four Chinese families with NOA participated in the study. We performed whole-exome sequencing (WES) for the four NOA-affected patients in four pedigrees. The candidate causative gene was further verified by Sanger sequencing. Hematoxylin and eosin staining (H&E) and immunohistochemistry (IHC) were carried out to evaluate the stage of spermatogenesis arrested in the patients with NOA. RESULTS: We identified two novel homozygous frameshift mutations of MSH4 and two novel compound heterozygous variants in MSH4 in four pedigrees with NOA. Homozygous loss of function (LoF) variants in MSH4 was identified in the NOA-affected patient (P9359) in a consanguineous Chinese family (NM_002440.4: c.805_812del: p.V269Qfs*15) and one patient with NOA (P21504) in another Chinese family (NM_002440.4: c.2220_2223del:p.K741Rfs*2). Also, compound heterozygous variants in MSH4 were identified in two NOA-affected siblings (P9517 and P9517B) (NM_002440.4: c.G1950A: p.W650X and c.2179delG: p.D727Mfs*11), and the patient with NOA (P9540) (NM_002440.4: c.G244A: p.G82S and c.670delT: p.L224Cfs*3). Histological analysis demonstrated lack of spermatozoa in seminiferous tubules of all patients and IHC showed the spermatogenesis arrested at the meiotic prophase I stage. Consistent with the autosomal recessive mode of inheritance, all of these mutations were inherited from heterozygous parental carriers. CONCLUSIONS: We identified that six novel mutations in MSH4 responsible for meiotic arrest and NOA. And these results provide researchers with a new insight to understand the genetic etiology of NOA and to identify new loci for genetic counselling of NOA.


Assuntos
Azoospermia/genética , Proteínas de Ciclo Celular/genética , Meiose/genética , Adulto , Pontos de Checagem do Ciclo Celular/genética , China , Família , Estudos de Associação Genética , Homozigoto , Humanos , Infertilidade Masculina/diagnóstico , Infertilidade Masculina/genética , Masculino , Mutação , Linhagem , Espermatogênese/genética , Sequenciamento do Exoma
9.
J Sex Med ; 19(10): 1536-1545, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35999130

RESUMO

BACKGROUND: A recent sham-controlled clinical study has shown that low-intensity pulsed ultrasound twice per week can safely and effectively treat patients with mild-to-moderate erectile dysfunction (ED). However, large-scale clinical trials are needed to verify its efficacy and safety and determine a reasonable treatment interval. AIM: To study whether low-intensity pulsed ultrasound therapy thrice per week is non-inferior to twice per week in patients with mild-to-moderate ED. METHODS: A randomized, open-label, parallel-group, non-inferiority clinical trial was conducted in 7 hospitals in China. A total of 323 patients with mild-to-moderate ED were randomized (1:1) into thrice per week (3/W) and twice per week (2/W) groups. Low-intensity pulsed ultrasound was applied on each side of the penis for 16 sessions. OUTCOMES: The primary outcome was response rate using the minimal clinically important difference in the International Index of Erectile Function (IIEF-EF) score at week 12. Secondary outcomes included Erection Hardness Score (EHS), Sexual Encounter Profile, Global Assessment Question, and Self Esteem and Relationship Questionnaire. RESULTS: Response rates in 3/W and 2/W groups were 62.0% and 62.5%, respectively. Treatment effect in the 3/W group was noninferior to that of the 2/W group, with rate difference lower bound of -0.01% [95% confidence interval -0.11 to 0.10%] within the acceptable margin (-14.0%). No significant difference was found among secondary outcomes. IIEF-EF score showed a significant increase from baseline in the 3/W group (16.8 to 20.7) and 2/W group (17.8 to 21.7), and the percentage of patients with EHS ≥3 increased in the 3/W (54.9% to 84.0%) and 2/W (59.5% to 83.5%) groups. There was no significant difference in response rate between the 2 groups after controlling for strata factors and homogeneous tests. No treatment-related adverse events were reported. CLINICAL IMPLICATIONS: Low-intensity pulsed ultrasound therapy displays similar efficacy and safety for mild-to-moderate ED when administered thrice or twice per week for 16 sessions. This study provides two options to suit patients' needs. STRENGTHS & LIMITATIONS: This is a large-sample, randomized, controlled, noninferiority trial study. Short-term follow-up and mostly younger patients are the main limitations. CONCLUSION: Low-intensity pulsed ultrasound therapy thrice and twice per week showed equivalent therapeutic effects and safety for mild-to-moderate ED in a young and generally healthy population. This therapy warrants further investigation of its potential value in rehabilitation of ED. Chen, H., Li Z., Li X., et al. The Efficacy and Safety of Thrice vs Twice per Week Low-Intensity Pulsed Ultrasound Therapy for Erectile Dysfunction: A Randomized Clinical Trial. J Sex Med 2022;19:1536-1545.


Assuntos
Disfunção Erétil , Método Duplo-Cego , Humanos , Masculino , Ereção Peniana , Pênis , Resultado do Tratamento , Ondas Ultrassônicas
10.
J Med Genet ; 58(10): 679-686, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-32900840

RESUMO

BACKGROUND: The genetic causes of human idiopathic non-obstructive azoospermia (NOA) with meiotic arrest remain unclear. METHODS: Two Chinese families with infertility participated in the study. In family 1, two brothers were affected by idiopathic NOA. In family 2, the proband was diagnosed with idiopathic NOA, and his elder sister suffered from infertility. Whole-exome sequencing (WES) was conducted in the two patients in family 1, the proband in family 2 and 362 additional sporadic patients with idiopathic NOA. Sanger sequencing was used to verify the WES results. Periodic acid-Schiff (PAS), immunohistochemistry (IHC) and meiotic chromosomal spread analyses were carried out to evaluate the stage of spermatogenesis arrested in the affected cases. RESULTS: We identified compound heterozygous loss of function (LoF) variants of SHOC1 (c.C1582T:p.R528X and c.231_232del:p.L78Sfs*9, respectively) in both affected cases with NOA from family 1. In family 2, homozygous LoF variant in SHOC1 (c.1194delA:p.L400Cfs*7) was identified in the siblings with infertility. PAS, IHC and meiotic chromosomal spread analyses demonstrated that the spermatogenesis was arrested at zygotene stage in the three patients with NOA. Consistent with the autosomal recessive mode of inheritance, all of these SHOC1 variants were inherited from heterozygous parental carriers. Intriguingly, WES of 362 sporadic NOA cases revealed one additional NOA case with a bi-allelic SHOC1 LoF variant (c.1464delT:p.D489Tfs*13). CONCLUSION: To the best of our knowledge, this is the first report identifying SHOC1 as the causative gene for human NOA. Furthermore, our study showed an autosomal recessive mode of inheritance in the NOA caused by SHOC1 deficiency.


Assuntos
Alelos , Azoospermia/diagnóstico , Azoospermia/genética , Pontos de Checagem do Ciclo Celular/genética , Mutação com Perda de Função , Meiose/genética , Adulto , Sequência de Aminoácidos , Biomarcadores , Bandeamento Cromossômico , Análise Mutacional de DNA , Estudos de Associação Genética , Predisposição Genética para Doença , Genótipo , Humanos , Imuno-Histoquímica , Masculino , Linhagem , Fenótipo , Testículo/metabolismo , Testículo/patologia , Sequenciamento do Exoma
11.
Andrologia ; 54(7): 1592-1597, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35312195

RESUMO

The three-dimension digital image microscope system (3D-DIM) with a better ergonomic design and equipment characteristics can contribute to the achievement of good results during microsurgery. In this study, the safety and efficiency of 3D-DIM assisted varicocelectomy was evaluated. From July 2019 to November 2019, fifteen cases with varicocele (20 sides of varicocele in total) were included, seven cases underwent 3D-DIM-assisted modified microsurgical subinguinal varicocelectomy, and eight cases underwent modified microsurgical subinguinal varicocelectomy under standard operating microscope (SOM). The mean operative time of 3D-DIM group (67 ± 12.3 min) was a little longer than that of SOM group (55 ± 12.9 min) (p < 0.05). There was no significant difference between the two groups in the number of internal spermatic arteries, internal spermatic vein, lymphatics, gubernacular vein, external spermatic vein and post-operation complications. The 3D-DIM showed a significant difference in image definition for nurse (p < 0.01) and in doctor-nurse cooperation (p < 0.05) over SOM. The 3D-DIM with better ergonomic design and image definition can be applied to perform microsurgical subinguinal varicocelectomy, and could improve the surgeon's fatigue and doctor-nurse cooperation. We believe that the 3D-DIM would be widely used in the field of male infertility microsurgery in the near future.


Assuntos
Cordão Espermático , Varicocele , Humanos , Masculino , Microcirurgia/métodos , Cordão Espermático/irrigação sanguínea , Cordão Espermático/cirurgia , Varicocele/cirurgia , Procedimentos Cirúrgicos Vasculares/métodos , Veias/cirurgia
12.
Int J Mol Sci ; 24(1)2022 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-36613865

RESUMO

Aging has a significant negative impact on human testicular function; steroidogenesis is gradually impaired, and testosterone replacement therapy still has many risks. Low-intensity pulsed ultrasound (LIPUS) has been used as a novel non-invasive treatment for male erectile dysfunction and other fields, and has been shown to increase testosterone levels in animal models. Testosterone is synthesized and secreted by Leydig cells (LCs), and the serum testosterone level decreases after aging due to the LCs senescence. However, the effect of LIPUS on human senescent LCs has not been reported. In this study, human senescent LCs were isolated and stimulated with different energy intensities in vitro, and cell morphology, cell apoptosis, cell proliferation, cell senescence levels, lipid droplet number, testosterone and INSL3 secretion levels were tested and analyzed. Quantitative Polymerase Chain Reaction (QPCR) and Western Blot were performed to compare cell senescence characteristics and the expression profile of key pathways of testosterone secretion, and transcriptome analysis was performed to explore the signaling pathways of LCs alteration after LIPUS stimulation. It was safe and effective to stimulate LCs with the 75 mW/cm2 energy of LIPUS in vitro, which not only improved the senescence phenotype, but also effectively enhanced the secretory function of LCs in vitro, and increased the expression of key pathways of the testosterone synthesis pathway. These results suggest that LIPUS could be used as a novel treatment to human senescent LCs with decreased testosterone secretion levels in vitro.


Assuntos
Células Intersticiais do Testículo , Testículo , Humanos , Masculino , Senescência Celular , Células Intersticiais do Testículo/metabolismo , Testículo/metabolismo , Testosterona/metabolismo , Ondas Ultrassônicas
13.
Zhonghua Nan Ke Xue ; 28(2): 122-128, 2022 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-37462483

RESUMO

OBJECTIVE: To quantitatively assess the performance of the new robotic visualization system (Zeiss, KINEVO 900) in terms of visual imaging effect and evaluate its potential application in microscopic vasectomy reversal. METHODS: We made a parallel comparison between the effects of the plane and stereo visual images of KINEVO 900 and optical surgical microscopy (Zeiss, S7), and performed microscopic vasectomy reversal on the rat model using KINEVO 900. RESULTS: Compared with S7, KINEVO 900 provided an even longer working distance (200-625 mm), a 3-4 times larger effective field area and a 1.5-2 times deeper front depth of field with the same working distance of 200 mm. No statistically significant difference was observed in the average anastomosis time and immediate patency rate between the two platforms (P > 0.05). CONCLUSION: The 4K3D video image stream outputted by KINEVO 900 is not inferior to that of the optical surgical microscope represented by S7 and is sufficient for microsurgeries in urology and andrology. More prospective randomized clinical animal experiments are needed to further evaluate its application value in andrology.


Assuntos
Procedimentos Cirúrgicos Robóticos , Urologia , Vasectomia , Vasovasostomia , Masculino , Animais , Ratos , Vasovasostomia/métodos , Estudos Prospectivos , Anastomose Cirúrgica , Microcirurgia/métodos , Vasectomia/métodos
14.
Reprod Biomed Online ; 43(4): 687-699, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34556414

RESUMO

RESEARCH QUESTION: Is vitrification with microinjection of single seminiferous tubules an efficient cryopreservation approach for limited testicular tissue? DESIGN: Testicular tissue from 10 patients with normal spermatogenesis were assigned to a fresh control group or one of the following cryopreservation procedures: uncontrolled slow freezing (USF) using either 1.5 or 2.1 M DMSO combined with sucrose and vitrification with or without single seminiferous tubules microinjection. RESULTS: Single seminiferous tubules microinjected with cryoprotective agents (CPA) enhanced the penetration of CPA compared with CPA-treated testicular tissue fragments. Microinjection of seminiferous tubules (VLP) maintained tubule structural integrity and germ cell numbers, and reduced spermatogonial apoptosis after cryopreservation compared with vitrification without microinjection (apoptosis rate: VLP versus vitrification without microinjection, P = 0.047; VLP versus USF, P= 0.049). Freezing of single seminiferous tubules using 0.25-ml straws and traditional sperm freezing methods protected sperm retrieval and recovery rates, and the progressive motility index. CONCLUSIONS: Vitrification of single seminiferous tubule with microinjection of low CPA concentration is an effective approach to testicular cryopreservation.


Assuntos
Células-Tronco Germinativas Adultas , Criopreservação/métodos , Crioprotetores/administração & dosagem , Túbulos Seminíferos , Espermatogônias , Humanos , Masculino , Microinjeções , Vitrificação
15.
Andrologia ; 53(8): e14133, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34260106

RESUMO

This study aimed to evaluate the efficacy and safety of vasal vessel-sparing modified single-armed 2-suture longitudinal intussusception vasoepididymostomy (SA-LIVE) to epididymal obstructive azoospermia patients. Forty consecutive epididymal obstructive azoospermia cases, who underwent microsurgical vasoepididymostomy in Shanghai General Hospital from January 2019 to October 2019, were included in this study. Twenty cases underwent SA-LIVE (group A), and 20 cases underwent vasal vessel-sparing SA-LIVE (group B). Until March 2021, the mean follow-up period was 16.9 ± 4.1 (12-23) months. The overall patency rate was 82.5%, and 80% and 85% for group A and group B respectively. The mean time to achieve patency was 4.11 ± 2.74 months. The overall natural pregnancy rate was 51.5%(17/33) at the mean follow-up of 16.9 months. The natural pregnancy rate was 50.0% for group A and 52.9% for group B (p > .05). At the time of 6 months post-operation, the patency rate was 70% for group A and 80% for group B (p = .465); the natural pregnancy rate was 0% for group A and 31.3% for group B (p = .022). Vasal vessel-sparing SA-LIVE is safe and effective to achieve favourable patency and pregnancy rates. Preserving vasal vessel would improve natural pregnancy rate at a very early stage.


Assuntos
Azoospermia , Azoospermia/cirurgia , China , Epididimo/cirurgia , Feminino , Humanos , Masculino , Microcirurgia , Gravidez , Estudos Retrospectivos , Resultado do Tratamento , Ducto Deferente/cirurgia
16.
BMC Urol ; 20(1): 121, 2020 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-32787873

RESUMO

BACKGROUND: To evaluate the clinical outcomes and the duration required for the sperm to return to the ejaculate after a modified single-armed 2-suture longitudinal intussusception vasoepididymostomy (SA-LIVE). METHODS: From March 2015 to December 2018, 134 patients with epididymal obstruction azoospermia underwent the modified single-armed vasoepididymostomy at Shanghai General Hospital. The outcomes and clinical findings were documented and evaluated. The mean follow-up period was 17 (range: 3-36) months. RESULTS: Patency was assessed by the return of sperm in the ejaculate. The overall patency rate was 55.2%, and the patency rates were 58.9, 40.7, 36.4, and 58.9% for bilateral surgery, unilateral surgery, proximal anastomosis, and distal anastomosis, respectively. The average time to achieve patency was 4.11 ± 2.74 months. In the first 6 months, 87.8% (65/74) patency patients reported sperm in the ejaculate. The overall pregnancy rate was 40.9% (29/66) at the follow-up of 3-36 months, and the natural pregnancy rate was 30.3% (20/66). The natural pregnancy rate was 32.1% post-bilateral surgery and 33.3% for the site of distal anastomosis; surprisingly, it was 0% for the site of proximal anastomosis. CONCLUSION: Modified SA-LIVE is safe and may achieve favorable patency and pregnancy rates. When double-armed sutures are not accessible, single-armed may be preferable. The expected patency time was within 1 year. Moreover, because of the low natural pregnancy rate for proximal anastomosis, sperm banking is preferred to SA-LIVE.


Assuntos
Azoospermia/cirurgia , Epididimo/cirurgia , Ducto Deferente/cirurgia , Adulto , Anastomose Cirúrgica/métodos , Feminino , Humanos , Período Intraoperatório , Masculino , Microcirurgia , Pessoa de Meia-Idade , Gravidez , Taxa de Gravidez , Estudos Prospectivos , Resultado do Tratamento , Procedimentos Cirúrgicos Urológicos Masculinos/métodos , Adulto Jovem
17.
Zhonghua Nan Ke Xue ; 25(12): 1059-1065, 2019 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-32251555

RESUMO

OBJECTIVE: To investigate the spatio-temporal expression and function of the end-directed KIFC1 (kinesin family member C1) protein during acrosomogenesis. METHODS: The expression and location of KIFC1 were analyzed by flow separation and immunofluorescence, and the small interference RNA (RNAi1) of KIFC1 with high-interference efficiency was screened using in vitro GC2-spd cell lines. The KIFC1 RNAi1 mixed with 0.5% trypan blue solution was microinjected into the testicular seminiferous tubules and negative Con-RNAi1 into the contralateral testis of a 3-week-old Balb/c mouse, followed by morphological analysis of the sperm collected from the testis tail at 3 weeks after injection. RESULTS: The expression of KIFC1 was observed mainly in the cytoplasm of the sperm cells in the early stage of sperm deformation, in the acrosomal vesicle and acrosome in the middle stage, and in the residual body in the late stage, but vanished during sperm maturation. The sperm head deformity rate was significantly higher in the RNAi1 than in the negative control group (ï¼»32.12 ± 0.25ï¼½% vs ï¼»7.06 ± 1.25ï¼½%, P < 0.01). CONCLUSIONS: The KIFC1 protein may play an important role in the formation of spermatozoa, mainly affecting acrosomogenesis.


Assuntos
Acrossomo/fisiologia , Espermatogênese , beta Carioferinas/genética , beta Carioferinas/fisiologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Interferência de RNA , Túbulos Seminíferos , Espermatozoides , Testículo
18.
Zhonghua Nan Ke Xue ; 24(9): 811-818, 2018 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-32212460

RESUMO

OBJECTIVE: To investigate the effect of Qilin Pills (QP) in facilitating the recovery of spermatogenic function in azoospermia (AS) mice and to explore its mechanism of regulating testicular spermatogenesis. METHODS: Fifteen 4-week-old male mice were equally randomized into an AS model control, a low-dose QP and a high-dose QP group. The AS model was established in the mice by intraperitoneal injection of busulfan at 35 mg/kg. After modeling, the animals in the low- and high-dose QP groups were treated with Qilin Pills intragastrically at 2 000 and 8 000 mg/kg/d respectively while those in the model control group fed on a normal diet, all for 28 days. Then, all the mice were sacrificed for examination of the ultrastructures of the epididymis and testis by HE staining, detection of the specific markers of spermatogenic, Sertoli and Leydig cells by Western blot, and determination of the expressions of these markers in the testis tissue by immunofluorescence assay. RESULTS: The number of spermatogenic cells in the testis tissue was significantly decreased in the AS model controls, with no spermatozoa in most of the seminiferous tubules in the epididymis (Johnsen's score: 5.2 ± 0.5). In the high-dose QP group, spermatogenic cells were tightly arranged with distinct layers in the seminiferous tubules, with a large number of spermatozoa but no non-sperm cells in the lumens of the epididymis (Johnsen's score: 9.4 ± 0.6). The number of spermatogenic cells in the testis was increased in the low-dose QP group with some spermatozoa in the seminiferous tubules as compared with that in the model control, but lower than in the high-dose group (Johnsen's score: 7.6 ± 0.6). The Johnsen's score was significantly lower in the model control than in the high- and low-dose QP groups (P < 0.01), and higher in the high-dose than in the low-dose QP group (P < 0.05). The expressions of the specific markers of Sertoli cells SCF, BMP4, SYCP3, DMC1 and Ki67 were also remarkably lower in the model control than in the high- and low-dose QP groups (P < 0.01), and higher in the high-dose than in the low-dose QP group (P < 0.05 or P < 0.01). No statistically significant differences were observed among the three groups of mice in the markers of spermatogonial stem cells (SSC) and undifferentiated SSCs UCHL1, STRA8, NGN3 and PLZF3 (P > 0.05). The expressions of the spermatocyte markers DMC1 and SYCP3 were markedly lower in the model control than in the high- and low-dose QP groups (P < 0.05 or P < 0.01), and higher in the high-dose than in the low-dose QP group (P < 0.05 or P < 0.01). The Ki67 fluorescence signals were distributed in the spermatogonia, with a higher intensity in the model control than in the high- and low-dose QP groups. The acrosome marker PNA was found mainly in the seminiferous tubules, with abundant fluorescence signals in the high- and low-dose QP groups but no obvious dot signals in the model controls. CONCLUSIONS: Qilin Pills may contribute to the meiosis of spermatogonia and promote spermatogenesis by improving the function of Sertoli cells in the testis.

19.
Reproduction ; 150(1): R25-34, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25852155

RESUMO

Spermatogenesis is composed of three distinctive phases, which include self-renewal of spermatogonia via mitosis, spermatocytes undergoing meiosis I/II and post-meiotic development of haploid spermatids via spermiogenesis. Spermatogenesis also involves condensation of chromatin in the spermatid head before transformation of spermatids to spermatozoa. Epigenetic regulation refers to changes of heritably cellular and physiological traits not caused by modifications in the DNA sequences of the chromatin such as mutations. Major advances have been made in the epigenetic regulation of spermatogenesis. In this review, we address the roles and mechanisms of epigenetic regulators, with a focus on the role of microRNAs and DNA methylation during mitosis, meiosis and spermiogenesis. We also highlight issues that deserve attention for further investigation on the epigenetic regulation of spermatogenesis. More importantly, a thorough understanding of the epigenetic regulation in spermatogenesis will provide insightful information into the etiology of some unexplained infertility, offering new approaches for the treatment of male infertility.


Assuntos
Metilação de DNA , Epigênese Genética , Meiose/genética , MicroRNAs/metabolismo , Mitose/genética , Espermatogênese/genética , Animais , Humanos , Masculino , MicroRNAs/genética
20.
Cell Commun Signal ; 13: 20, 2015 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-25880873

RESUMO

BACKGROUND: Sertoli cells play key roles in regulating spermatogenesis and testis development by providing structural and nutritional supports. Recent studies demonstrate that Sertoli cells can be converted into functional neural stem cells. Adult Sertoli cells have previously been considered the terminally differentiated cells with a fixed and unmodifiable population after puberty. However, this concept has been challenged. Since the number of adult human Sertoli cells is limited, it is essential to culture these cells for a long period and expand them to obtain sufficient cells for their basic research and clinic applications. Nevertheless, the studies on human Sertoli cells are restricted, because it is difficult to get access to human testis tissues. RESULTS: Here we isolated adult human Sertoli cells with a high purity and viability from obstructive azoospermia patients with normal spermatogenesis. Adult human Sertoli cells were cultured with DMEM/F12 and fetal bovine serum for 2 months, and they could be expanded with a 59,049-fold increase of cell numbers. Morphology, phenotypic characteristics, and the signaling pathways of adult human Sertoli cells from different passages were compared. Significantly, adult human Sertoli cells assumed similar morphological features, stable global gene expression profiles and numerous proteins, and activation of AKT and SMAD1/5 during long-period culture. CONCLUSIONS: This study demonstrates that adult human Sertoli cells can be cultured for a long period and expanded with remarkable increase of cell numbers whilst maintaining their primary morphology, phenotype and signaling pathways. This study could provide adequate human Sertoli cells for reproductive and regenerative medicine.


Assuntos
Regulação da Expressão Gênica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células de Sertoli/metabolismo , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Adulto , Animais , Azoospermia/metabolismo , Azoospermia/patologia , Bovinos , Técnicas de Cultura de Células , Células Cultivadas , Humanos , Masculino , Células de Sertoli/patologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA