Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Andrologia ; 54(10): e14548, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36177775

RESUMEN

Spermatogenesis associated 16 (SPATA16) gene plays an important role in acrosome formation. In this study, we analysed SPATA16 promoter methylation in 29 oligozoospermic infertile and 16 normozoospermic fertile sperm samples and in testicular biopsy from 16 non-obstructive azoospermic and 2 obstructive azoospermic individuals. Next, we analysed SPATA16 level in sperm from 8 oligozoospermic infertile, 6 normozoospermic fertile, 9 IVF failed normozoospermic and 10 IVF successful normozoospermic samples by immunoblotting. This was followed by the analysis of SPATA16 expression in testicular biopsy from azoospermic individuals (n = 8) in comparison to normozoospermic individuals (n = 2). Oligozoospermic infertile sperm samples showed significantly higher methylation in the SPATA16 promoter region. Similarly, testicular biopsy from azoospermic men also showed significantly higher level of DNA methylation. Sub-group analysis of infertile sperm and testicular biopsy samples showed a direct correlation between DNA methylation and the degree of spermatogenic impairment. Oligozoospermic infertile samples and IVF failed samples showed reduced SPATA16 expression in comparison to normozoospermic fertile and IVF successful samples, respectively. Human biopsy analysis showed a significant decrease in SPATA16 expression in hypospermatogenesis, maturation arrest and Sertoli cell only syndrome. In conclusion, hypermethylation in SPATA16 promoter shows a highly significant correlation with infertility, which is consistent with its down-regulation in infertility.


Asunto(s)
Azoospermia , Infertilidad Masculina , Oligospermia , Proteínas de Transporte Vesicular , Azoospermia/genética , Metilación de ADN , Regulación hacia Abajo , Humanos , Infertilidad Masculina/genética , Masculino , Oligospermia/genética , Regiones Promotoras Genéticas , Semen , Proteínas de Transporte Vesicular/genética
2.
Biol Reprod ; 107(4): 881-901, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-35878972

RESUMEN

Endometriosis that afflicts one in 10 women of reproductive age is characterized by growth of endometrial tissue in the extra-uterine sites and encompasses metabolic-, immunologic-, and endocrine-disruption. Importantly, several comorbidities are associated with endometriosis, especially autoimmune disorders such as inflammatory bowel disease. Primarily thought of as a condition arising from retrograde menstruation, emerging evidence uncovered a functional link between the gut microbiota and endometriosis. Specifically, recent findings revealed altered gut microbiota profiles in endometriosis and in turn this altered microbiota appears to be causal in the disease progression, implying a bidirectional crosstalk. In this review, we discuss the complex etiology and pathogenesis of endometriosis, emphasizing on this recently recognized role of gut microbiome. We review the gut microbiome structure and functions and its complex network of interactions with the host for maintenance of homeostasis that is crucial for disease prevention. We highlight the underlying mechanisms on how some bacteria promote disease progression and others protect against endometriosis. Furthermore, we highlight the areas that require future emphases in the gut microbiome-endometriosis nexus and the potential microbiome-based therapies for amelioration of endometriosis.


Asunto(s)
Endometriosis , Microbioma Gastrointestinal , Microbiota , Progresión de la Enfermedad , Endometriosis/patología , Femenino , Homeostasis , Humanos
3.
J Mol Endocrinol ; 69(3): R81-R94, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35900833

RESUMEN

The gut microbiome is considered an endocrine organ that can influence distant organs and associated biological pathways. Recent advances suggest that gut microbial homeostasis is essential for reproductive health and that perturbations in the gut microbiota can lead to reproductive pathologies. This review provides an updated overview of the relationship between the gut microbiome and female reproductive diseases. Specifically, we highlight the most recent findings on the gut microbiome in gynecological pathologies including polycystic ovarian syndrome, endometriosis, and endometrial cancer. Most studies revealed associations between altered gut microbial compositions and these reproductive diseases, though few have suggested cause-effect relationships. Future studies should focus on determining the molecular mechanisms underlying associations between gut microbiota and reproductive diseases. Understanding this bidirectional relationship could lead to the development of novel and effective strategies to prevent, diagnose, and treat female reproductive organ-related diseases.


Asunto(s)
Endometriosis , Microbioma Gastrointestinal , Síndrome del Ovario Poliquístico , Femenino , Homeostasis , Humanos , Síndrome del Ovario Poliquístico/metabolismo , Reproducción
4.
Andrologia ; 54(7): e14437, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35437806

RESUMEN

The aim of the present study was to identify RNA-based signatures of male infertility by sperm transcriptome analysis. In this study, deep sequencing analyses of coding (mRNA) and regulatory (miRNA) transcriptomes were performed by pooling 15 oligo/oligoasthenozoospermic infertile sperm and 9 normozoospermic fertile sperm samples. Furthermore, interesting candidates were selected for validation by real-time PCR. The comparison of miRNAs between cases and controls identified 94 differentially expressed miRNAs, of which at least 38 have known functions in spermatogenesis. In transcriptome (mRNA) data, a total of 60,505 transcripts were obtained. The comparison of coding RNAs between cases and controls revealed 11,688 differentially expressed genes. miRNA-mRNA paired analysis revealed that 94 differentially expressed miRNAs could potentially target 13,573 genes, of which 6419 transcripts were actually differentially expressed in our data. Out of these, 3303 transcripts showed inverse correlation with their corresponding regulatory miRNAs. Moreover, we found that most of the genes of miRNA-mRNA pairs were involved in male germ cell differentiation, apoptosis, meiosis, spermiogenesis and male infertility. In conclusion, we found that a number of sperm transcripts (miRNAs and mRNAs) have a very high potential of serving as infertility/sperm quality markers.


Asunto(s)
Infertilidad Masculina , MicroARNs , Humanos , Infertilidad Masculina/genética , Masculino , MicroARNs/genética , ARN Mensajero/genética , Semen , Espermatozoides , Transcriptoma
5.
Nutr Res ; 91: 57-66, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34134041

RESUMEN

One-carbon metabolism is crucial for the maintenance of healthy pregnancy and alterations in this pathway have been associated with various pregnancy-related complications. Therefore, the present study was conducted to test the hypothesis that the altered folic acid, vitamin B12 and homocysteine levels are associated with the risk of early pregnancy loss (EPL). Plasma folic acid, vitamin B12 and homocysteine levels were analyzed in 83 females with EPL and 70 healthy pregnant females in their first trimester. Further, meta-analyses of folic acid, vitamin B12 and homocysteine were also performed involving various eligible studies. Results from our case-control study and meta-analysis showed that folic acid deficiency is not associated with the risk of EPL. On the other hand, low vitamin B12 and hyperhomocysteinemia were individually found to be significant risk factors for EPL in the present study (P < .01, P < .05, respectively) and meta-analysis as well (P < .001, P < .05, respectively). Vitamin B12 deficiency in combination with hyperhomocysteinemia was a more serious risk factor for EPL (Odds Ratio = 4.98, P = 0.002). Therefore, we conclude that vitamin B12 deficiency and elevated homocysteine levels are independent risk factors for EPL, and of higher risk when combined. The assessment of vitamin B12 and homocysteine levels may serve as a good screening marker for EPL risk.


Asunto(s)
Aborto Espontáneo/etiología , Homocisteína/sangre , Hiperhomocisteinemia/complicaciones , Estado Nutricional , Complicaciones del Embarazo/sangre , Deficiencia de Vitamina B 12/complicaciones , Vitamina B 12/sangre , Aborto Espontáneo/sangre , Adulto , Estudios de Casos y Controles , Femenino , Ácido Fólico/sangre , Deficiencia de Ácido Fólico , Humanos , Hiperhomocisteinemia/sangre , Fenómenos Fisiologicos Nutricionales Maternos , Metaanálisis como Asunto , Oportunidad Relativa , Embarazo , Factores de Riesgo , Deficiencia de Vitamina B 12/sangre , Adulto Joven
6.
Reprod Sci ; 28(3): 617-638, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32748224

RESUMEN

Lifestyle factors, which include the practices we adopt in our daily life, have a significant role in shaping our overall health. These lifestyle choices are mainly centered on personal preferences and our surrounding social environment. In addition to lifestyle factors, we continuously interact with our environment, which impacts physiology. Several factors have been claimed to affect women's fertility; lifestyle-related factors, in particular, have received great attention in the last decade. Due to societal and professional pressure, childbearing age in women has gradually shifted to the 30s. Delayed age of childbearing along with modern lifestyle offers a wider window of opportunity for various lifestyle and genetic perturbations to penetrate to affect fertility. While clinical studies have strengthened a direct correlation between lifestyle, environment, and female reproductive health; experimental studies on animal models have investigated their mechanism of action. In most instances, these factors target the neuroendocrine pathways, resulting in metabolic derangements. This review aims to dissect the plausible interconnection of lifestyle and environmental factors with various neuroendocrine pathways and to discuss how it can affect the female physiology in the long-term, resulting in reproductive incompetence.


Asunto(s)
Exposición a Riesgos Ambientales/efectos adversos , Fertilidad , Infertilidad Femenina/etiología , Estilo de Vida , Salud Reproductiva , Determinantes Sociales de la Salud , Adulto , Comorbilidad , Femenino , Estado de Salud , Humanos , Infertilidad Femenina/diagnóstico , Infertilidad Femenina/fisiopatología , Medición de Riesgo , Factores de Riesgo
7.
Health Care Women Int ; 42(4-6): 580-597, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32701388

RESUMEN

We investigated the impact of socio-demographic variables on antenatal care (ANC) utilization and the low birth weight of a child. Data were collected from 300 pregnant females. Only 22.5% of females received full antenatal care (≥4 visits). Our results showed that female's age at marriage and education plays a significant role in improving ANC. We observed an overall decrease in the utilization of services provided during each antenatal visit. ANC visits from the first trimester decrease the risk of having a baby with low birth weight. Awareness programs and educating families about pregnancy care are recommended to improve ANC utilization.


Asunto(s)
Aceptación de la Atención de Salud , Atención Prenatal , Niño , Femenino , Humanos , India , Matrimonio , Embarazo , Mujeres Embarazadas
8.
Reprod Toxicol ; 96: 128-140, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32593569

RESUMEN

Approximately 2 billion people worldwide are susceptible to iodine deficiency. Iodine deficiency has largely been tackled by iodine fortification in salt; however indiscriminate use of iodine raises the risk of iodine toxicity. In this study, we aimed to investigate the molecular mechanisms underlying adverse effect of excess iodine on spermatogenesis. Sprague Dawley (SD) rats were orally administered with 0.7 mg potassium iodide (KI)/100 g Bw and 3.5 mg potassium iodide (KI)/100 g Bw for a period of 60 days. This resulted in significant loss of sperm count and motility. Molecular investigations provided evidence for the generation of oxidative stress with high SOD levels, reduced Nrf2, HO-1 and increased NF-kB and Follistatin. Further investigations showed increased apoptosis evidenced by reduced expression of anti-apoptotic (BCL-2, Survivin), increased expression of pro-apoptotic (Bid, Bax) markers, and increased expression of p53 and other modulators/effectors of apoptosis (cytochrome c, cleaved PARP, caspase3 and caspase9). Analysis of the blood testis barrier proteins showed reduced expression of tight junction (JAM-A, Tricellulin), ectoplasmic specialization (Integrin- ß1), adherens junction (N-Cadherin, E-cadherin, ß-catenin) proteins, and reduced expression of other junction protein coding genes (Claudin1, Claudin 5, Occludin, ZO-1, Testin, Fibronectin, CAR-F). Focal adhesion kinase (FAK) and key regulators of spermatogenesis (c-Kit receptor, androgen receptor) were also parallelly decreased. Further investigation showed reduced expression of germ cell proliferation and differentiation markers (PCNA, Cyclin D1, c-Kit, Cdk-4). These findings collectively explain the loss of spermatogenesis under excess iodine conditions. In conclusion, excess iodine causes loss of spermatogenesis by inducing oxidative stress and disrupting the blood testis barrier and cytoskeleton.


Asunto(s)
Barrera Hematotesticular/efectos de los fármacos , Yodo/toxicidad , Estrés Oxidativo/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Ciclo Celular/metabolismo , Masculino , Ratas Sprague-Dawley , Recuento de Espermatozoides , Motilidad Espermática/efectos de los fármacos , Testículo/efectos de los fármacos , Testículo/metabolismo , Testículo/patología , Proteínas de Uniones Estrechas/genética , Proteínas de Uniones Estrechas/metabolismo
9.
J Assist Reprod Genet ; 37(2): 459-469, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31919744

RESUMEN

PURPOSE: To identify the frequency of Y chromosome microdeletions in Indian populations and to quantitatively estimate the significance of association between these deletions and male infertility. METHODS: A total of 379 infertile males (302 azoospermic and 77 oligozoospermic infertile males) and 265 normozoospermic fertile males were evaluated for Y chromosome microdeletions (YCD) using PCR amplification and gel electrophoresis. Meta-analyses were performed on AZFa (2079 cases and 1217 controls), AZFb (2212 cases and 1267 controls), AZFc (4131 cases and 2008 controls), and AZFb+c (1573 cases and 942 controls) deletions data to quantitatively estimate the significance of association between these deletions and male infertility in Indian populations. RESULTS: The results revealed that out of 379 infertile azoospermic and oligozoospermic males, 38 (10.02%) had AZF deletions. No deletion was found in control samples. The highest percentage of deletions was observed in the AZFc region, followed by AZFa and AZFb. Qualitative analysis showed that AZF deletions were present in 0.59 to 32.62% (average 13.48%) of infertile cases in Indian populations. Meta-analysis revealed a significant association of AZFa (OR = 6.74, p value = 0.001), AZFb (OR = 4.694, p value = 0.004), AZFc (OR = 13.575, p value = 0.000), and AZFb+c (OR = 5.946, p value = 0.018) deletions with male infertility. CONCLUSION: AZF deletions were seen in 10.02% of azoospermic and oligozoospermic cases with the highest frequency of AZFc deletions. Pooled analysis for all studies showed deletion frequency from 0.59 to 32.62% (average = 13.48%). Meta-analysis showed significant association of AZFa, AZFb, and AZFb+c deletions with male infertility. Analysis of Y chromosome microdeletions should be reckoned as an essential testing for diagnostic and therapeutic purposes.


Asunto(s)
Azoospermia/genética , Enfermedades Genéticas Ligadas al Cromosoma Y/genética , Infertilidad Masculina/genética , Oligospermia/genética , Trastornos de los Cromosomas Sexuales del Desarrollo Sexual/genética , Adulto , Azoospermia/epidemiología , Azoospermia/patología , Deleción Cromosómica , Cromosomas Humanos Y/genética , Enfermedades Genéticas Ligadas al Cromosoma Y/epidemiología , Enfermedades Genéticas Ligadas al Cromosoma Y/fisiopatología , Humanos , India/epidemiología , Infertilidad Masculina/epidemiología , Infertilidad Masculina/patología , Infertilidad Masculina/fisiopatología , Masculino , Oligospermia/epidemiología , Oligospermia/fisiopatología , Reacción en Cadena de la Polimerasa , Aberraciones Cromosómicas Sexuales , Trastornos de los Cromosomas Sexuales del Desarrollo Sexual/epidemiología , Trastornos de los Cromosomas Sexuales del Desarrollo Sexual/patología , Adulto Joven
10.
Andrology ; 8(3): 602-609, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31838782

RESUMEN

BACKGROUND: Spermatogenesis-associated (SPATA) family of genes plays important roles in spermatogenesis, sperm maturation or fertilization. The knockout studies in mice have demonstrated that SPATA genes are crucial for fertility. Gene expression and genetic polymorphism studies have further suggested their correlation with infertility; however, methylation analysis of SPATA genes in human male infertility has not yet been undertaken. OBJECTIVES: To analyze the methylation status of SPATA4, SPATA5 and SPATA6 genes in oligozoospermic male infertility. MATERIALS AND METHODS: In the present study, we have analyzed DNA methylation pattern in the promoter regions of SPATA4, SPATA5 and SPATA6 genes in oligozoospermic patients and compared it with normozoospermic fertile controls. Semen samples were obtained from 30 oligozoospermic infertile and 19 normozoospermic fertile controls, and DNA methylation levels of the target gene promoters were analyzed by amplicon based deep sequencing methylation analysis using MiSeq. RESULTS: SPATA4 (P < 0.0008), SPATA5 (P = 0.009) and SPATA6 (Promoter, P < 0.0005; Exon 1, P = 0.0128) genes were significantly hypermethylated in oligozoospermic patients in comparison to controls. This is the first study reporting a higher methylation in the promoters of SPATA4, SPATA5 and SPATA6 in oligozoospermic infertile individuals in comparison to the normozoospermic fertile controls. DISCUSSION: Altered methylation of SPATA genes would affect pathways involved in sperm production or affect various processes linked to sperm fertility. CONCLUSION: In conclusion, hypermethylation in the SPATA4, SPATA5 and SPATA6 genes correlates with oligozoospermic infertility.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/genética , Proteínas del Citoesqueleto/genética , Metilación de ADN/genética , Infertilidad Masculina/genética , Proteínas/genética , Adulto , Humanos , Masculino , Regiones Promotoras Genéticas/genética
11.
J Assist Reprod Genet ; 36(10): 2171-2179, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31418107

RESUMEN

PURPOSE: To identify genomic imbalances and candidate loci in idiopathic male infertility. METHODS: Affymetrix CytoScan 750K Array was used to analyze genomic imbalances and candidate loci in 34 idiopathic infertile cases of different phenotypes (hypo-spermatogenesis, n = 8; maturation arrest, n = 7; and Sertoli cell-only syndrome, n = 13, severe oligozoospermia, n = 6, and 10 normozoospermic fertile men). Ten ethnically matched controls were screened for comparison. RESULTS: The cytogenetic array analysis detected a genomic gain at the 19p13.3 region in 9 (26.47%) cases, with the highest frequency in patients with Sertoli cell-only syndrome (SCOS) (38%). Its complete absence in the control group suggests its likely pathogenic nature. In addition to Y-classical, micro, and partial deletions, the duplication in 19p13.3 could serve as a unique biomarker for evaluation of infertility risk. The common region across the individuals harboring the duplication identified STK11, ATP5D, MIDN, CIRBP, and EFNA2 genes which make them strong candidates for further investigations. The largest duplicated region identified in this study displayed a major network of 7 genes, viz., CIRBP, FSTL3, GPX4, GAMT, KISS1R, STK11, and PCSK4, associated with reproductive system development and function. The role of chance was ruled out by screening of ethnically matched controls. CONCLUSION: The result clearly indicates the significance of 19p13.3 duplication in infertile men with severe testicular phenotypes. The present study underlines the utility and significance of whole genomic analysis in the cases of male infertility which goes undiagnosed due to limitations in the conventional cytogenetic techniques and for identifying genes that are essential for spermatogenesis.


Asunto(s)
Duplicación Cromosómica/genética , Cromosomas Humanos Par 19/genética , Infertilidad Masculina/genética , Síndrome de Sólo Células de Sertoli/genética , Adulto , Aberraciones Cromosómicas , Variaciones en el Número de Copia de ADN/genética , Inestabilidad Genómica/genética , Humanos , Infertilidad Masculina/patología , Masculino , Persona de Mediana Edad , Síndrome de Sólo Células de Sertoli/patología , Espermatogénesis/genética
12.
Fertil Steril ; 112(1): 61-72.e1, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31103287

RESUMEN

OBJECTIVE: To study peripheral blood DNA differential methylation in oligozoospermic infertile men in comparison with normozoospermic fertile controls. DESIGN: Case-control study. SETTING: Reproductive biology laboratory. PATIENTS(S): Azoospermic and oligozoospermic infertile patients (n = 6) and normozoospermic fertile controls (n = 6) in the discovery phase, and oligo/asthenozoospermic infertile men (n = 11) and normozoospermic fertile controls (n = 10) in the validation phase. INTERVENTION(S): Blood samples drawn from all participants, DNA isolation and methylation analysis. MAIN OUTCOME MEASURE(S): DNA methylation values analyzed using genomewide methylation 450K BeadChip array, followed by deep sequencing of selected regions for methylation analysis in the neighborhood regions of differentially methylated CpGs. RESULT(S): We found 329 differentially methylated CpG spots, out of which 245 referred to the genes, representing 170 genes. Deep-sequencing analysis confirmed the methylation pattern suggested by 450K array. A thorough literature search suggested that 38 genes play roles in spermatogenesis (PDHA2, PARP12, FHIT, RPTOR, GSTM1, GSTM5, MAGI2, BCAN, DDB2, KDM4C, AGPAT3, CAMTA1, CCR6, CUX1, DNAH17, ELMO1, FNDC3B, GNRHR, HDAC4, IRS2, LIF, SMAD3, SOD3, TALDO1, TRIM27, GAA, PAX8, RNF39, HLA-C, HLA-DRB6), are testis enriched (NFATC1, NMNAT3, PIAS2, SRPK2, WDR36, WWP2), or show methylation differences between infertile cases and controls (PTPRN2, RPH3AL). CONCLUSION(S): We found a statistically significant correlation between peripheral blood DNA methylation and male infertility, raising the hope that epigenome-based blood markers can be used for screening male infertility risk. The study also identified new candidates for spermatogenesis and fertility.


Asunto(s)
Azoospermia/diagnóstico , Metilación de ADN , Fertilidad/genética , Perfilación de la Expresión Génica/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligospermia/diagnóstico , Azoospermia/sangre , Azoospermia/genética , Azoospermia/fisiopatología , Estudios de Casos y Controles , Islas de CpG , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Oligospermia/sangre , Oligospermia/genética , Oligospermia/fisiopatología , Fenotipo , Valor Predictivo de las Pruebas
13.
Artículo en Inglés | MEDLINE | ID: mdl-30744807

RESUMEN

High fidelity DNA repair is critical to sustain the genomic integrity and quality of developing germ cells. Deficiencies in DNA repair machinery may result in increased DNA damage in germ cell leading to abnormal spermatogenesis and infertility. X-ray repair cross-complementing group 1 (XRCC1) is a testis enriched protein that plays a crucial role in the DNA base excision repair (BER) pathway. The aim of this study was to analyze the level of XRCC1 transcript and protein in infertile men and its association with DNA damage in sperm. A total of eighty infertile patients with different infertile phenotypes (Azoospermia, n = 30; Severe oligozoospermia, n = 25; Severe oligoasthenozoospermia, n = 25) and age-matched controls (normal spermatogenesis [NS], n = 15 and fertile controls, n = 10) were recruited. γ-H2 AX protein levels were analyzed to estimate the DNA damage in sperm. XRCC1 transcript levels in cases and controls were determined by qRT-PCR. XRCC1 and γ-H2 AX proteins were immunohistochemically analyzed in testicular biopsy sections obtained from NOA patients and OA controls. The determination of XRCC1 and γ-H2 AX protein levels was performed with Western blots. The results revealed reduced expression of XRCC1 mRNA and protein in infertile individuals as compared to controls (p < 0.001). γ-H2 AX levels were significantly increased in infertile cases as compared to controls, indicating increased DNA damage in infertile men. The results indicate decreased expression of the XRCC1 gene in infertile patients which may be one of the factors associated with impaired spermatogenesis and infertility.


Asunto(s)
Daño del ADN , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Polimorfismo de Nucleótido Simple , Espermatozoides/patología , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/metabolismo , Adulto , Estudios de Casos y Controles , Reparación del ADN , Humanos , Infertilidad Masculina/metabolismo , Masculino , Pronóstico , Espermatozoides/metabolismo , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/genética
14.
DNA Repair (Amst) ; 75: 39-47, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30711825

RESUMEN

Compelling evidence suggest that germs cells are predominantly sensitive to DNA damaging agents in comparison to other cells. High fidelity DNA repair in testicular cells thus becomes indispensable to preserve the genomic integrity for passing on to the progeny. Compromised DNA repair machinery in the testicular cells may result in impaired spermatogenesis and infertility. It remains unclear if the alterations in the expression of DNA repair genes correlate with azoospermia and male infertility. In the present study, 54 non-obstructive azoospermic infertile patients with hypospermatogenesis (HS, n = 26), maturation arrest (MA, n = 15), Sertoli cell only syndrome (SCOS, n = 13) and 14 controls with obstructive azoospermia, but normal spermatogenesis were recruited. Expression profiling of 84 DNA repair genes in testicular biopsy samples was performed using PCR array. Out of 84 genes, 27, 64 and 28 genes showed >5 fold down-regulation in the HS, MA and SCOS groups, respectively. On the basis of differential expression and their functional significance in spermatogenesis, ten genes (MSH2, BRIP1, CCNH, LIG4, MGMT, NTHL1, PMS1, DMC1, POLB and XPA) were selected for validation of transcript levels in a higher number of cases using RT-PCR, which corroborated the findings of array. Four genes (MSH2, LIG4, PMS1 and DMC1) were analyzed for protein levels using immunohistochemistry, which further validated the loss of DNA repair gene expression. Caspase-3 immunostaining showed that the loss of DNA repair correlated with increased testicular apoptosis in patients. Maturation arrest showed the highest apoptotic index with maximum number of downregulated genes. We conclude that the loss of DNA repair genes expression in testis correlates with increased apoptosis, azoospermia and infertility.


Asunto(s)
Azoospermia/genética , Reparación del ADN/genética , Transcriptoma/genética , Adulto , Apoptosis/genética , Azoospermia/enzimología , Azoospermia/patología , Estudios de Casos y Controles , Caspasa 3/metabolismo , Humanos , Masculino , Espermatogénesis/genética
15.
J Assist Reprod Genet ; 36(1): 79-90, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30390177

RESUMEN

PURPOSE: We investigated if substitutions in the ERCC1, ERCC2, and XRCC1 genes of the DNA repair pathway correlate with non-obstructive azoospermia and male infertility. METHODS: A total of 548 azoospermic infertile males and 410 fertile controls were genotyped for XRCC1 399A > G, 280G > A, and ERCC1 C > A 3' UTR and 541 azoospermic infertile males and 416 fertile controls were genotyped for ERCC2 751A > C using iPLEX Gold Assay. Meta-analyses were performed on XRCC1 399A > G (1022 cases and 1004 controls), ERCC1 C > A 3' UTR (879 cases and 1059 controls), and ERCC2 751A > C (914 cases and 850 controls) polymorphisms to quantitatively estimate the significance of the association between these polymorphisms and the risk of infertility. RESULTS: Statistically significant association between ERCC2 751A > C SNP and male infertility was found using the codominant model (p = 0.03). Results of meta-analysis suggested a lack of correlation with male infertility risk, which could be due to pooling of studies from different ethnic populations. Due to limited the number of studies, a stratified analysis for different ethnic groups could not be performed. CONCLUSION (S): In conclusion, AA genotype of 751A > C SNP in ERCC2 correlated with a higher risk of male infertility and may contribute to an increased risk of azoospermia and male infertility in Indian men.


Asunto(s)
Pueblo Asiatico/genética , Infertilidad Masculina/genética , Polimorfismo de Nucleótido Simple , Proteína de la Xerodermia Pigmentosa del Grupo D/genética , Asia/epidemiología , Estudios de Casos y Controles , Reparación del ADN , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Humanos , Infertilidad Masculina/epidemiología , Infertilidad Masculina/patología , Masculino , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/genética
16.
Hum Reprod ; 33(12): 2256-2267, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30358834

RESUMEN

STUDY QUESTION: Do methylation changes in sperm DNA correlate with infertility? STUDY ANSWER: Loss of spermatogenesis and fertility was correlated with 1680 differentially-methylated CpGs (DMCs) across 1052 genes. WHAT IS KNOWN ALREADY: Methylation changes in a number of genes have been correlated with reduced sperm count and motility. STUDY DESIGN, SIZE, DURATION: This case-control study used spermatozoal DNA from 38 oligo-/oligoastheno-zoospermic infertile patients and 26 normozoospermic fertile men. PARTICIPANTS/MATERIALS, SETTINGS, METHODS: Genome-wide methylation analysis was undertaken using 450 K BeadChip on spermatozoal DNA from six infertile and six fertile men to identify DMCs. This was followed by deep sequencing of spermatozoal DNA from 32 infertile patients and 20 fertile controls. MAIN RESULTS AND THE ROLE OF CHANCE: A total of 1680 DMCs were identified, out of which 1436 were hypermethylated and 244 were hypomethylated. Classification of DMCs according to the genes identified BCAN, CTNNA3, DLGAP2, GATA3, MAGI2 and TP73 among imprinted genes, SPATA5, SPATA7, SPATA16 and SPATA22 among spermatogenesis-associated genes, KDM4C and JMJD1C, EZH2 and HDAC4 among genes which regulate methylation and gene expression, HLA-C, HLA-DRB6 and HLA-DQA1 among complementation and immune response genes, and CRISPLD1, LPHN3 and CPEB2 among other genes. Genes showing significant differential methylation in deep sequencing, i.e. HOXB1, GATA3, EBF3, BCAN and TCERG1L, are strong candidates for further investigations. The role of chance was ruled out by deep sequencing of select genes. LARGE-SCALE DATA: N/A. LIMITATIONS, REASON FOR CAUTION: Genome-wide analyses are fairly accurate, but may not be exactly validated in replication studies across all DMCs. We used the 't' test in the genome-wide methylation analysis, whereas other tests could provide a more robust and powerful analysis. WIDER IMPLICATIONS OF THE FINDINGS: DMCs can serve as markers for inclusion in infertility screening panels, particularly those in the genes showing differential methylation consistent with previous studies. The genes validated by deep sequencing are strong candidates for investigations of their roles in spermatogenesis. STUDY FUNDING/COMPETING INTEREST(S): The study was funded by the Council of Scientific and Industrial Research (CSIR), Govt. of India with grant number BSC0101 awarded to Rajender Singh. None of the authors has any competing interest to declare.


Asunto(s)
Islas de CpG/genética , Metilación de ADN , Infertilidad Masculina/genética , Espermatogénesis/genética , Adulto , Estudios de Casos y Controles , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Oligospermia/genética , Recuento de Espermatozoides , Motilidad Espermática/genética
17.
Front Biosci (Schol Ed) ; 9(3): 395-419, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28410126

RESUMEN

Fertilization is a hallmark event of sexual reproduction marked by the fusion of male and female gamete to form zygote. It is a highly complex, yet a robust process that is intricately regulated by various signalling molecules. A healthy fertilization is determined by the quality of zygote which is contingent on the health of egg and sperm. The relationship between infertility and gametic health can be reciprocal. On one hand gametogenesis has to be dynamic and unremitting to sustain the reproductive health, while on the other hand it has to be error free for proper embryonic development. Complex cellular interactions make gametogenesis highly vulnerable to extrinsic as well as intrinsic intrusions. Molecular disparities during these phases may result in complete fertilization failure. Present review provides an overview of the regulation of gametogenesis, determinants of healthy gamete, players at fertilization window and what may go wrong during the development of zygote to embryo leading to implantation failure. We have outlined different 'windows' of vulnerability during gametogenesis supported by evidences affecting the fertility potential of both the partners.


Asunto(s)
Fertilización/fisiología , Óvulo/fisiología , Espermatozoides/fisiología , Animales , Femenino , Humanos , Infertilidad/patología , Infertilidad/fisiopatología , Masculino , Oogénesis , Espermatogénesis
18.
PLoS One ; 11(3): e0151510, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26983014

RESUMEN

BACKGROUND: Optimum efficiency of the folate pathway is considered essential for adequate ovarian function. 677 C>T substitution in the 5, 10-methylene tertrahydrofolatereductase (MTHFR) gene compromises activity of the MTHFR enzyme by about 50%. The significance of correlation between 677C>T substitution and PCOS remains dubious due to the low power of published studies. METHODS AND RESULTS: We analyzed MTHFR 677 C>T site in ethnically two different PCOS case-control groups (total 261 cases and 256 controls) from India. The data analysis revealed a lack of association between this polymorphism and PCOS [OR = 1.11 (95%CI = 0.71-1.72), P = 0.66]. Group-wise analysis on the basis of ethnicity also revealed no association in any of the ethnic groups [Indo-Europeans, P = 1; Dravidians, P = 0.70]. Homocysteine levels did not differ significantly between cases (15.51 µmol/L, SD = 2.89) and controls (15.89 µmol/L, SD = 2.23). We also undertook a meta-analysis on 960 cases and 1028 controls, which suggested a significant association of the substitution with PCOS in the dominant model of analysis (OR = 1.47 (95%CI = 1.04-2.09), P = 0.032]. Trial sequential analysis corroborated findings of the traditional meta-analysis. However, we found that the conclusions of meta-analysis were strongly influenced by studies that deviated from the Hardy Weinberg equilibrium. A careful investigation of each study and a trial sequential analysis suggested that 677 C>T substitution holds no clinical significance in PCOS in most of the populations. CONCLUSION: In conclusion, MTHFR 677 C>T polymorphism does not affect PCOS risk in India. The association seen in the meta-analysis is due to an outlier study and studies showing deviation from the Hardy Weinberg equilibrium.


Asunto(s)
Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Síndrome del Ovario Poliquístico/genética , Polimorfismo Genético , Estudios de Casos y Controles , Femenino , Humanos
19.
Gene ; 542(2): 263-5, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24690399

RESUMEN

We present the case of two brothers who were referred to a male infertility clinic for infertility workup. Conventional chromosome analysis and Y chromosome microdeletions did not reveal any genetic alterations. We utilized the chromosome microarray analysis (CMA) to identify novel and common variations associated with this severely impaired spermatogenesis cases. CMA specific results showed a common deletion in the 15q15.3 region that harbors genes like CATSPER2, STRC and PPIP5K1 in both cases (M18 and M19). In addition we identified small duplication in X and 11 chromosomes of M19. This is the first familial case report from India on occurrence of CATSPER gene deletion in human male infertility.


Asunto(s)
Canales de Calcio/genética , Eliminación de Gen , Infertilidad Masculina/genética , Adulto , Deleción Cromosómica , Cromosomas Humanos Par 15 , Cromosomas Humanos Y , Humanos , India , Péptidos y Proteínas de Señalización Intercelular , Masculino , Proteínas de la Membrana/genética , Análisis por Micromatrices , Fosfotransferasas (Aceptor del Grupo Fosfato)/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA