Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26.640
Filtrar
1.
Ceska Gynekol ; 89(4): 282-292, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39242203

RESUMEN

AIM: The aim of the study was to evaluate the influence of a specific diagnosis of infertile women and men on their life quality and psychosexual functioning based on internationally validated questionnaires. MATERIALS AND METHODS: A total of 853 couples seeking treatment for infertility completed the gender-specific batteries comprised of Fertility Quality of Life tool (FertiQoL), Female Sexual Function Index (FSFI) in women, and Brief Sexual Function Inventory (BSFI) in men. Women were followed in the group of primary and secondary infertility and then with specific diagnoses - polycystic ovary syndrome, tubal factor, endometriosis, and idiopathic sterility. Men's categories reflected spermiogram results, i.e., normozoospermia, merged categories of milder disorders of a spermiogram (teratozoospermia, asthenozoospermia, oligozoospermia, and oligoasthenoteratospermia), oligoasthenoteratospermia (OAT) gravis, azoospermia, and when the man was not examined. RESULTS: When evaluating the quality of life in women, we found statistically significant differences between primary and secondary sterility. Primary infertile women scored worse especially in the social area. Worse assessment appeared also in mind-body (area evaluating affliction of the body). Emotional and relational domains included similar results in primary and secondary infertile women. With a specific diagnosis, statistically significant differences were not proved. Using the orientational cut-off score, FertiQoL stated that approximately 10% of women experienced adverse quality of life in relation to fertility. In the domain of sexual functioning, 30% of women demonstrated clinically significant dysfunctions. In men, respondents in the normozoospermic and non-diagnosed categories scored higher than those in the merged category and OAT gravis. Only 2% of men felt their quality of life was poor due to fertility, and clinically significant dysfunctions appeared only in 3% of them. CONCLUSION: In women, impaired fertility-related quality of life and psychosexual functioning are significantly linked to primary sterility, where specifically the social domain is affected. The impact of a specific diagnosis appears to be minimal. We found high levels of sexual dysfunctions in women. In men, we follow the link of evaluated quality of life in connection with their results of the spermiogram. With spermiogram defects, both areas of functioning can be affected.


Asunto(s)
Infertilidad Femenina , Infertilidad Masculina , Calidad de Vida , Humanos , Femenino , Masculino , Infertilidad Femenina/psicología , Infertilidad Femenina/diagnóstico , Adulto , Infertilidad Masculina/psicología , Infertilidad Masculina/diagnóstico , Encuestas y Cuestionarios
2.
Zool Res ; 45(5): 1073-1087, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39245651

RESUMEN

Infertility represents a significant health concern, with sperm quantity and quality being crucial determinants of male fertility. Oligoasthenoteratozoospermia (OAT) is characterized by reduced sperm motility, lower sperm concentration, and morphological abnormalities in sperm heads and flagella. Although variants in several genes have been implicated in OAT, its genetic etiologies and pathogenetic mechanisms remain inadequately understood. In this study, we identified a homozygous nonsense mutation (c.916C>T, p.Arg306*) in the coiled-coil domain containing 146 ( CCDC146) gene in an infertile male patient with OAT. This mutation resulted in the production of a truncated CCDC146 protein (amino acids 1-305), retaining only two out of five coiled-coil domains. To validate the pathogenicity of the CCDC146 mutation, we generated a mouse model ( Ccdc146 mut/mut ) with a similar mutation to that of the patient. Consistently, the Ccdc146 mut/mut mice exhibited infertility, characterized by significantly reduced sperm counts, diminished motility, and multiple defects in sperm heads and flagella. Furthermore, the levels of axonemal proteins, including DNAH17, DNAH1, and SPAG6, were significantly reduced in the sperm of Ccdc146 mut/mut mice. Additionally, both human and mouse CCDC146 interacted with intraflagellar transport protein 20 (IFT20), but this interaction was lost in the mutated versions, leading to the degradation of IFT20. This study identified a novel deleterious homozygous nonsense mutation in CCDC146 that causes male infertility, potentially by disrupting axonemal protein transportation. These findings offer valuable insights for genetic counseling and understanding the mechanisms underlying CCDC146 mutant-associated infertility in human males.


Asunto(s)
Astenozoospermia , Proteínas Asociadas a Microtúbulos , Animales , Humanos , Masculino , Ratones , Astenozoospermia/genética , Codón sin Sentido , Homocigoto , Infertilidad Masculina/genética , Mutación , Oligospermia/genética , Motilidad Espermática/genética , Espermatozoides , Proteínas Asociadas a Microtúbulos/genética
3.
Mol Genet Genomics ; 299(1): 84, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39223386

RESUMEN

Male infertility is a complex multifactorial reproductive disorder with highly heterogeneous phenotypic presentations. Azoospermia is a medically non-manageable cause of male infertility affecting ∼1% of men. Precise etiology of azoospermia is not known in approximately three-fourth of the cases. To explore the genetic basis of azoospermia, we performed whole exome sequencing in two non-obstructive azoospermia affected siblings from a consanguineous Pakistani family. Bioinformatic filtering and segregation analysis of whole exome sequencing data resulted in the identification of a rare homozygous missense variant (c.962G>C, p. Arg321Thr) in YTHDC2, segregating with disease in the family. Structural analysis of the missense variant identified in our study and two previously reported functionally characterized missense changes (p. Glu332Gln and p. His327Arg) in mice showed that all these three variants may affect Mg2+ binding ability and helicase activity of YTHDC2. Collectively, our genetic analyses and experimental observations revealed that missense variant of YTHDC2 can induce azoospermia in humans. These findings indicate the important role of YTHDC2 deficiency for azoospermia and will provide important guidance for genetic counseling of male infertility.


Asunto(s)
Azoospermia , Secuenciación del Exoma , Homocigoto , Mutación Missense , Linaje , Hermanos , Adulto , Animales , Humanos , Masculino , Ratones , Azoospermia/genética , Azoospermia/patología , Consanguinidad , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Pakistán , ARN Helicasas/genética
4.
BMJ Open ; 14(9): e084236, 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39277209

RESUMEN

BACKGROUND: Male factor infertility can affect spermatogenesis, sexual desire, and thus the quality of life of couples. The present study was conducted to investigate the relationship between spermogram parameters, and the score of sexual desire in infertile men. METHODS: This cross-sectional study was conducted on 315 infertile men referred to the Avicenna Infertility Center of Tehran (March 2022 to March 2023). The participants were selected based on the results of previous spermogram and hormonal tests recorded in their medical records. Eligible men completed the demographic information questionnaire and Hurlbert Index of Sexual Desire. A multivariable linear regression model was used to adjust the effect of variables on Hurlbert's score. RESULTS: There was no significant relationship among sperm parameters (count, morphology, motility, vitality, concentration and DNA Fragmentation Index) and with sexual desire of infertile men. Education level, age of men and their partners, the duration of the marriage and duration of infertility did not have a statistically significant effect on sexual desire. However, economic status had an inverse effect on men's sexual desire, with regression coefficients of 7.37 and 7.78 for medium and low socioeconomic levels compared with high (p<0.05). CONCLUSION: Male sexual desire is primarily influenced by social factors rather than organic ones. Further multicentre prospective studies are recommended for more accurate results.


Asunto(s)
Infertilidad Masculina , Libido , Humanos , Masculino , Estudios Transversales , Irán/epidemiología , Adulto , Infertilidad Masculina/psicología , Libido/fisiología , Centros de Atención Terciaria , Recuento de Espermatozoides , Análisis de Semen , Encuestas y Cuestionarios , Motilidad Espermática , Calidad de Vida
5.
Clin Genet ; 106(4): 437-447, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39221575

RESUMEN

Male infertility due to asthenozoospermia is quite frequent, but its etiology is poorly understood. We recruited two infertile brothers, born to first-cousin parents from Pakistan, displaying idiopathic asthenozoospermia with mild stuttering disorder but no ciliary-related symptoms. Whole-exome sequencing identified a splicing variant (c.916+1G>A) in ARMC3, recessively co-segregating with asthenozoospermia in the family. The ARMC3 protein is evolutionarily highly conserved and is mostly expressed in the brain and testicular tissue of human. The ARMC3 splicing mutation leads to the exclusion of exon 8, resulting in a predicted truncated protein (p.Glu245_Asp305delfs*16). Quantitative real-time PCR revealed a significant decrease at mRNA level for ARMC3 and Western blot analysis did not detect ARMC3 protein in the patient's sperm. Individuals homozygous for the ARMC3 splicing variant displayed reduced sperm motility with frequent morphological abnormalities of sperm flagella. Transmission electron microscopy of the affected individual IV: 2 revealed vacuolation in sperm mitochondria at the midpiece and disrupted flagellar ultrastructure in the principal and end piece. Altogether, our results indicate that this novel homozygous ARMC3 splicing mutation destabilizes sperm flagella and leads to asthenozoospermia in our patients, providing a novel marker for genetic counseling and diagnosis of male infertility.


Asunto(s)
Astenozoospermia , Consanguinidad , Homocigoto , Linaje , Empalme del ARN , Cola del Espermatozoide , Adulto , Humanos , Masculino , Astenozoospermia/genética , Astenozoospermia/patología , Secuenciación del Exoma , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Mutación , Empalme del ARN/genética , Motilidad Espermática/genética , Cola del Espermatozoide/patología , Cola del Espermatozoide/ultraestructura , Cola del Espermatozoide/metabolismo , Espermatozoides/ultraestructura , Espermatozoides/patología
6.
Cell Mol Life Sci ; 81(1): 379, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39222270

RESUMEN

As the most abundant small RNAs, piwi-interacting RNAs (piRNAs) have been identified as a new class of non-coding RNAs with 24-32 nucleotides in length, and they are expressed at high levels in male germ cells. PiRNAs have been implicated in the regulation of several biological processes, including cell differentiation, development, and male reproduction. In this review, we focused on the functions and molecular mechanisms of piRNAs in controlling spermatogenesis, including genome stability, regulation of gene expression, and male germ cell development. The piRNA pathways include two major pathways, namely the pre-pachytene piRNA pathway and the pachytene piRNA pathway. In the pre-pachytene stage, piRNAs are involved in chromosome remodeling and gene expression regulation to maintain genome stability by inhibiting transposon activity. In the pachytene stage, piRNAs mediate the development of male germ cells via regulating gene expression by binding to mRNA and RNA cleavage. We further discussed the correlations between the abnormalities of piRNAs and male infertility and the prospective of piRNAs' applications in reproductive medicine and future studies. This review provides novel insights into mechanisms underlying mammalian spermatogenesis and offers new targets for diagnosing and treating male infertility.


Asunto(s)
Infertilidad Masculina , ARN Interferente Pequeño , Espermatogénesis , Espermatogénesis/genética , Masculino , Humanos , Animales , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/genética , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Medicina Reproductiva , Mamíferos/genética , Mamíferos/metabolismo , ARN de Interacción con Piwi
7.
PLoS One ; 19(9): e0309974, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39231187

RESUMEN

Azoospermia (the complete absence of spermatozoa in the semen) is a common cause of male infertility. The etiology of azoospermia is poorly understood. Whole-genome analysis of azoospermic men has identified a number of candidate genes, such as the X-linked testis-expressed 11 (TEX11) gene. Using a comparative genomic hybridization array, an exonic deletion (exons 10-12) of TEX11 had previously been identified in two non-apparent azoospermic patients. However, the putative impact of this genetic alteration on spermatogenesis and the azoospermia phenotype had not been validated functionally. We therefore used a CRISPR/Cas9 system to generate a mouse model (Tex11Ex9-11del/Y) with a partial TEX11 deletion that mimicked the human mutation. Surprisingly, the mutant male Tex11Ex9-11del/Y mice were fertile. The sperm concentration, motility, and morphology were normal. Similarly, the mutant mouse line's testis transcriptome was normal, and the expression of spermatogenesis genes was not altered. These results suggest that the mouse equivalent of the partial deletion observed in two infertile male with azoospermia has no impact on spermatogenesis or fertility in mice, at least of a FVB/N genetic background and until 10 months of age. Mimicking a human mutation does not necessarily lead to the same human phenotype in mice, highlighting significant differences species.


Asunto(s)
Azoospermia , Meiosis , Espermatogénesis , Animales , Masculino , Ratones , Espermatogénesis/genética , Meiosis/genética , Azoospermia/genética , Azoospermia/patología , Infertilidad Masculina/genética , Eliminación de Secuencia , Humanos , Testículo/metabolismo , Testículo/patología , Sistemas CRISPR-Cas
8.
Development ; 151(18)2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39222051

RESUMEN

Male infertility can be caused by chromosomal abnormalities, mutations and epigenetic defects. Epigenetic modifiers pre-program hundreds of spermatogenic genes in spermatogonial stem cells (SSCs) for expression later in spermatids, but it remains mostly unclear whether and how those genes are involved in fertility. Here, we report that Wfdc15a, a WFDC family protease inhibitor pre-programmed by KMT2B, is essential for spermatogenesis. We found that Wfdc15a is a non-canonical bivalent gene carrying both H3K4me3 and facultative H3K9me3 in SSCs, but is later activated along with the loss of H3K9me3 and acquisition of H3K27ac during meiosis. We show that WFDC15A deficiency causes defective spermiogenesis at the beginning of spermatid elongation. Notably, depletion of WFDC15A causes substantial disturbance of the testicular protease-antiprotease network and leads to an orchitis-like inflammatory response associated with TNFα expression in round spermatids. Together, our results reveal a unique epigenetic program regulating innate immunity crucial for fertility.


Asunto(s)
Homeostasis , Espermátides , Espermatogénesis , Masculino , Animales , Espermatogénesis/genética , Ratones , Espermátides/metabolismo , Testículo/metabolismo , Histonas/metabolismo , Péptido Hidrolasas/metabolismo , Péptido Hidrolasas/genética , Epigénesis Genética , Infertilidad Masculina/genética , Ratones Endogámicos C57BL , Meiosis/genética , Células Madre Germinales Adultas/metabolismo , Ratones Noqueados , Inmunidad Innata/genética , Espermatogonias/metabolismo
9.
BMC Med Genomics ; 17(1): 230, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39267058

RESUMEN

BACKGROUND: Spermatogenic failure is one of the leading causes of male infertility and its genetic etiology has not yet been fully understood. METHODS: The study screened a cohort of patients (n = 167) with primary male infertility in contrast to 210 normally fertile men using whole exome sequencing (WES). The expression analysis of the candidate genes based on public single cell sequencing data was performed using the R language Seurat package. RESULTS: No pathogenic copy number variations (CNVs) related to male infertility were identified using the the GATK-gCNV tool. Accordingly, variants of 17 known causative (five X-linked and twelve autosomal) genes, including ACTRT1, ADAD2, AR, BCORL1, CFAP47, CFAP54, DNAH17, DNAH6, DNAH7, DNAH8, DNAH9, FSIP2, MSH4, SLC9C1, TDRD9, TTC21A, and WNK3, were identified in 23 patients. Variants of 12 candidate (seven X-linked and five autosomal) genes were identified, among which CHTF18, DDB1, DNAH12, FANCB, GALNT3, OPHN1, SCML2, UPF3A, and ZMYM3 had altered fertility and semen characteristics in previously described knockout mouse models, whereas MAGEC1,RBMXL3, and ZNF185 were recurrently detected in patients with male factor infertility. The human testis single cell-sequencing database reveals that CHTF18, DDB1 and MAGEC1 are preferentially expressed in spermatogonial stem cells. DNAH12 and GALNT3 are found primarily in spermatocytes and early spermatids. UPF3A is present at a high level throughout spermatogenesis except in elongating spermatids. The testicular expression profiles of these candidate genes underlie their potential roles in spermatogenesis and the pathogenesis of male infertility. CONCLUSION: WES is an effective tool in the genetic diagnosis of primary male infertility. Our findings provide useful information on precise treatment, genetic counseling, and birth defect prevention for male factor infertility.


Asunto(s)
Secuenciación del Exoma , Infertilidad Masculina , Humanos , Masculino , Infertilidad Masculina/genética , Adulto , Variaciones en el Número de Copia de ADN
10.
Hum Genomics ; 18(1): 97, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39256880

RESUMEN

BACKGROUND: Bi-allelic variants in DNAH11 have been identified as causative factors in Primary Ciliary Dyskinesia, leading to abnormal respiratory cilia. Nonetheless, the specific impact of these variants on human sperm flagellar and their involvement in male infertility remain largely unknown. METHODS: A collaborative effort involving two Chinese reproductive centers conducted a study with 975 unrelated infertile men. Whole-exome sequencing was employed for variant screening, and Sanger sequencing confirmed the identified variants. Morphological and ultrastructural analyses of sperm were conducted using Scanning Electron Microscopy and Transmission Electron Microscopy. Western Blot Analysis and Immunofluorescence Analysis were utilized to assess protein levels and localization. ICSI was performed to evaluate its efficacy in achieving favorable pregnancy outcomes for individuals with DNAH11 variants. RESULTS: In this study, we identified seven novel variants in the DNAH11 gene in four asthenoteratozoospermia subjects. These variants led the absence of DNAH11 proteins and ultrastructure defects in sperm flagella, particularly affecting the outer dynein arms (ODAs) and adjacent structures. The levels of ODA protein DNAI2 and axoneme related proteins were down regulated, instead of inner dynein arms (IDA) proteins DNAH1 and DNAH6. Two out of four individuals with DNAH11 variants achieved clinical pregnancies through ICSI. The findings confirm the association between male infertility and bi-allelic deleterious variants in DNAH11, resulting in the aberrant assembly of sperm flagella and contributing to asthenoteratozoospermia. Importantly, ICSI emerges as an effective intervention for overcoming reproductive challenges caused by DNAH11 gene variants.


Asunto(s)
Astenozoospermia , Dineínas Axonemales , Secuenciación del Exoma , Infertilidad Masculina , Humanos , Masculino , Astenozoospermia/genética , Astenozoospermia/patología , Dineínas Axonemales/genética , Femenino , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Adulto , Cola del Espermatozoide/patología , Cola del Espermatozoide/ultraestructura , Cola del Espermatozoide/metabolismo , Inyecciones de Esperma Intracitoplasmáticas , Embarazo , Espermatozoides/ultraestructura , Espermatozoides/patología , Dineínas/genética
11.
BMJ ; 386: e080664, 2024 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-39231578

RESUMEN

OBJECTIVE: To investigate associations between long term residential exposure to road traffic noise and particulate matter with a diameter <2.5 µm (PM2.5) and infertility in men and women. DESIGN: Nationwide prospective cohort study. SETTING: Denmark. PARTICIPANTS: 526 056 men and 377 850 women aged 30-45 years, with fewer than two children, cohabiting or married, and residing in Denmark between 2000 and 2017. MAIN OUTCOME MEASURE: Incident infertility in men and women during follow-up in the Danish National Patient Register. RESULTS: Infertility was diagnosed in 16 172 men and 22 672 women during a mean follow-up of 4.3 years and 4.2 years, respectively. Mean exposure to PM2.5 over five years was strongly associated with risk of infertility in men, with hazard ratios of 1.24 (95% confidence interval 1.18 to 1.30) among men aged 30-36.9 years and 1.24 (1.15 to 1.33) among men aged 37-45 years for each interquartile (2.9 µg/m3) higher PM2.5 after adjustment for sociodemographic variables and road traffic noise. PM2.5 was not associated with infertility in women. Road traffic noise (Lden, most exposed facade of residence) was associated with a higher risk of infertility among women aged 35-45 years, with a hazard ratio of 1.14 (1.10 to 1.18) for each interquartile (10.2 dB) higher five year mean exposure. Noise was not associated with infertility among younger women (30-34.9 years). In men, road traffic noise was associated with higher risk of infertility in the 37-45 age group (1.06, 1.02 to 1.11), but not among those aged 30-36.9 years (0.93, 0.91 to 0.96). CONCLUSIONS: PM2.5 was associated with a higher risk of an infertility diagnosis in men, whereas road traffic noise was associated with a higher risk of an infertility diagnosis in women older than 35 years, and potentially in men older than 37 years. If these results are confirmed in future studies, higher fertility could be added to the list of health benefits from regulating noise and air pollution.


Asunto(s)
Contaminación del Aire , Exposición a Riesgos Ambientales , Material Particulado , Humanos , Dinamarca/epidemiología , Femenino , Masculino , Adulto , Persona de Mediana Edad , Exposición a Riesgos Ambientales/efectos adversos , Contaminación del Aire/efectos adversos , Material Particulado/efectos adversos , Material Particulado/análisis , Estudios Prospectivos , Infertilidad Masculina/epidemiología , Infertilidad Masculina/etiología , Ruido del Transporte/efectos adversos , Ruido del Transporte/estadística & datos numéricos , Factores de Riesgo , Infertilidad Femenina/epidemiología , Infertilidad Femenina/etiología , Infertilidad/epidemiología , Incidencia
12.
PLoS One ; 19(9): e0307396, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39250513

RESUMEN

BACKGROUND: Since the discovery of COVID-19 in December 2019, the novel virus has spread globally causing significant medical and socio-economic burden. Although the pandemic has been curtailed, the virus and its attendant complication live on. A major global concern is its adverse impact on male fertility. AIM: This study was aimed to give an up to date and robust data regarding the effect of COVID-19 on semen variables and male reproductive hormones. MATERIALS AND METHODS: Literature search was performed according to the recommendations of PRISMA. Out of the 852 studies collected, only 40 were eligible for inclusion in assessing the effect SARS-CoV-2 exerts on semen quality and androgens. More so, a SWOT analysis was conducted. RESULTS: The present study demonstrated that SARS-CoV-2 significantly reduced ejaculate volume, sperm count, concentration, viability, normal morphology, and total and progressive motility. Furthermore, SARS-CoV-2 led to a reduction in circulating testosterone level, but a rise in oestrogen, prolactin, and luteinizing hormone levels. These findings were associated with a decline in testosterone/luteinizing hormone ratio. CONCLUSIONS: The current study provides compelling evidence that SARS-CoV-2 may lower male fertility by reducing semen quality through a hormone-dependent mechanism; reduction in testosterone level and increase in oestrogen and prolactin levels.


Asunto(s)
COVID-19 , SARS-CoV-2 , Análisis de Semen , Testosterona , Humanos , Masculino , Testosterona/sangre , SARS-CoV-2/aislamiento & purificación , Fertilidad , Infertilidad Masculina/virología , Infertilidad Masculina/sangre , Hormona Luteinizante/sangre , Recuento de Espermatozoides , Semen/virología , Semen/metabolismo , Motilidad Espermática
13.
Sci Rep ; 14(1): 20889, 2024 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-39244620

RESUMEN

Microtubules, polymers of αß-tubulin heterodimers, are essential for various cellular processes. The incorporation of different tubulin isotypes, each encoded by distinct genes, is proposed to contribute to the functional diversity observed in microtubules. However, the functional roles of each tubulin isotype are not completely understood. In this study, we investigated the role of the ß4B-tubulin isotype (Tubb4b) in spermatogenesis, utilizing a Tubb4b knockout mouse model. We showed that ß4B-tubulin is expressed in the germ cells throughout spermatogenesis. ß4B-tubulin was localized to cytoplasmic microtubules, mitotic spindles, manchette, and axonemes of sperm flagella. We found that the absence of ß4B-tubulin resulted in male infertility and failure to produce sperm cells. Our findings demonstrate that a lack of ß4B-tubulin leads to defects in the initial stages of spermatogenesis. Specifically, ß4B-tubulin is needed for the expansion of differentiating spermatogonia, which is essential for the subsequent progression of spermatogenesis.


Asunto(s)
Diferenciación Celular , Ratones Noqueados , Microtúbulos , Espermatogénesis , Espermatogonias , Tubulina (Proteína) , Animales , Masculino , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/genética , Espermatogonias/metabolismo , Espermatogonias/citología , Espermatogénesis/genética , Ratones , Microtúbulos/metabolismo , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología
14.
Elife ; 132024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39269275

RESUMEN

Transmembrane channel-like (TMC) proteins are a highly conserved ion channel family consisting of eight members (TMC1-TMC8) in mammals. TMC1/2 are components of the mechanotransduction channel in hair cells, and mutations of TMC1/2 cause deafness in humans and mice. However, the physiological roles of other TMC proteins remain largely unknown. Here, we show that Tmc7 is specifically expressed in the testis and that it is required for acrosome biogenesis during spermatogenesis. Tmc7-/- mice exhibited abnormal sperm head, disorganized mitochondrial sheaths, and reduced number of elongating spermatids, similar to human oligo-astheno-teratozoospermia. We further demonstrate that TMC7 is colocalized with GM130 at the cis-Golgi region in round spermatids. TMC7 deficiency leads to aberrant Golgi morphology and impaired fusion of Golgi-derived vesicles to the developing acrosome. Moreover, upon loss of TMC7 intracellular ion homeostasis is impaired and ROS levels are increased, which in turn causes Golgi and endoplasmic reticulum stress. Taken together, these results suggest that TMC7 is required to maintain pH and ion homeostasis, which is needed for acrosome biogenesis. Our findings unveil a novel role for TMC7 in acrosome biogenesis during spermiogenesis.


Asunto(s)
Acrosoma , Infertilidad Masculina , Ratones Noqueados , Espermatogénesis , Animales , Masculino , Acrosoma/metabolismo , Ratones , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Espermatogénesis/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/deficiencia , Aparato de Golgi/metabolismo , Testículo/metabolismo
15.
Cells ; 13(17)2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39273059

RESUMEN

This study aims to investigate the role of ferroptosis, an iron-dependent form of regulated cell death, in male infertility. The motivation behind this research stems from the increasing recognition of oxidative stress and iron metabolism dysregulation as critical factors in male reproductive health. In this study, 28 infertile patients (grouped by the presence of urogenital infections or varicocele) and 19 fertile men were selected. Spermiograms were performed by light microscopy (WHO, 2021). Testosterone, ferritin, transferrin-bound iron, transferrin, and F2-isoprostanes (F2-IsoPs) were detected in seminal plasma. Glutathione peroxidase 4 (GPX4) and acyl coenzyme A synthetase long chain family member 4 (ACSL4) were also assessed in sperm cells using enzyme-linked immunosorbent assays (ELISA). All the variables were correlated (statistically significant Spearman's rank correlations) in the whole population, and then the comparison between variables of the different groups of men were carried out. Seminal ferritin and transferrin positively correlated with seminal F2-IsoPs, which had positive correlations with ACSL4 detected in sperm cells. Ferritin and ACSL4 negatively correlated with the seminal parameters. No correlation was detected for GPX4. Comparing the variables in the three examined groups, elevated levels of ACSL4 were observed in infertile patients with urogenital infections and varicocele; GPX4 levels were similar in the three groups. These results suggested a mechanism of ferroptosis, identified by increased ACSL4 levels and the occurrence of lipid peroxidation. Such events appear to be GPX4-independent in reproductive pathologies such as varicocele and urogenital infections.


Asunto(s)
Biomarcadores , Ferroptosis , Infertilidad Masculina , Semen , Humanos , Masculino , Semen/metabolismo , Adulto , Biomarcadores/metabolismo , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Coenzima A Ligasas/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fertilidad , Espermatozoides/metabolismo , Espermatozoides/patología
16.
Syst Biol Reprod Med ; 70(1): 261-271, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39217625

RESUMEN

Despite that the SARS-CoV-2 pandemic has been controlled, it has affected a large proportion of the population, raising some concerns about potential sequelae in men at reproductive age. To contribute to the clarification of this issue, we performed a retrospective study comparing semen parameters values before and after confirmed SARS-CoV-2 infection in a large cohort of infertile men, compared to a control group that did not undergo SARS-CoV-2 infection. Wilcoxon test on paired samples and general linear regression model showed that SARS-CoV-2 infection has a detrimental effect on semen volume values (p < 0.005). However, semen volume seems to be significantly lower only during the first spermatogenic cycle after SARS-COV-2 infection (p < 0.005) and mainly in unvaccinated patients (p < 0.05). In addition, we detected alterations in progressive motility in patients infected with the alpha SARS-COV-2 strain (p < 0.05). In conclusion, our results show that although SARS-CoV-2 has a small effect on semen volume and sperm motility in infertile men, depending on the infectious strain or vaccination status, pre-infection values of semen parameters appear to be restored over one spermatogenic cycle after infection.


Asunto(s)
COVID-19 , Infertilidad Masculina , SARS-CoV-2 , Análisis de Semen , Semen , Humanos , Masculino , COVID-19/complicaciones , COVID-19/fisiopatología , Estudios Retrospectivos , Adulto , Infertilidad Masculina/virología , Infertilidad Masculina/fisiopatología , Infertilidad Masculina/etiología , Semen/virología , Motilidad Espermática
17.
Sheng Li Xue Bao ; 76(4): 631-642, 2024 Aug 25.
Artículo en Chino | MEDLINE | ID: mdl-39192795

RESUMEN

In recent decades, there has been a consistent decline in semen quality across the globe, with environmental pollution emerging as the predominant factor. Persistent organic pollutants (POPs) have garnered considerable attention due to their potent biological toxicity and resistance to natural degradation. Within this class of pollutants, polycyclic aromatic hydrocarbons (PAHs) and halogenated aromatic hydrocarbons (HAHs) have been identified as detrimental agents that can disrupt cellular physiological functions by activating aryl hydrocarbon receptor (AhR). However, the precise role of AhR in the adverse effects of environmental pollutants on male mammalian fertility remains incompletely understood. This article provides a comprehensive review of the impact of various environmental pollutants, specifically PAHs such as benzo[a]pyrene, 3-methylcholanthrene, and 7,12-dimethylbenzo[a]anthracene, HAHs including 2,3,7,8-tetrachlorodibenzo-p-dioxins, polychlorinated biphenyls, polybrominated diphenyl ethers, and the pollutant complex PM2.5, as well as cigarette smoke condensates, on male mammalian reproductive function. Additionally, this review focuses on the role of the AhR in mediating these effects. The objective of this review is to elucidate the involvement of AhR in the regulation of male mammalian fertility, thereby offering insights for prospective investigations into the interplay between AhR and male reproductive function, as well as the etiology of idiopathic male infertility in clinic.


Asunto(s)
Contaminantes Ambientales , Infertilidad Masculina , Hidrocarburos Policíclicos Aromáticos , Receptores de Hidrocarburo de Aril , Animales , Humanos , Masculino , Contaminantes Ambientales/toxicidad , Contaminantes Ambientales/efectos adversos , Fertilidad/efectos de los fármacos , Éteres Difenilos Halogenados/efectos adversos , Éteres Difenilos Halogenados/toxicidad , Infertilidad Masculina/inducido químicamente , Infertilidad Masculina/etiología , Infertilidad Masculina/metabolismo , Contaminantes Orgánicos Persistentes/efectos adversos , Contaminantes Orgánicos Persistentes/metabolismo , Bifenilos Policlorados/efectos adversos , Bifenilos Policlorados/toxicidad , Hidrocarburos Policíclicos Aromáticos/efectos adversos , Hidrocarburos Policíclicos Aromáticos/toxicidad , Receptores de Hidrocarburo de Aril/metabolismo
18.
Zhonghua Nan Ke Xue ; 30(6): 547-563, 2024 Jun.
Artículo en Chino | MEDLINE | ID: mdl-39212365

RESUMEN

OBJECTIVE: To evaluate the efficacy and safety of traditional Chinese medicine (TCM) in the treatment of male immune infertility (MII) by meta-analysis. METHODS: We retrieved randomized controlled trial (RCT) on the treatment of male immune infertility with traditional Chinese medicine from the databases of WanFang, Chinese Biomedical Literature, Cochrane Library, Weipu, PubMed and CNKI, and performed methodological quality assessment of the RCTs identified and statistical analysis and evaluation of the publication bias using the RevMan5.4 software. RESULTS: Totally, 25 RCTs (2 563 cases) were included in this study. Compared with Western medicine alone in the treatment of MII, TCM achieved a significantly higher total effectiveness rate (OR = 6.35, 95% CI: 4.96-8.13, P<0.000 01), negative conversion rate of seminal plasma anti-sperm antibodies (OR = 4.52, 95% CI: 2.72 - 7.51, P<0.000 01), negative rate of serum anti-sperm antibodies (OR = 2.98, 95% CI: 2.23-3.96, P<0.000 01), sperm concentration (MD = 15.56, 95% CI: 11.32-19.79, P<0.000 01), grade a sperm motility (MD = 3.85, 95% CI: 1.91-5.79, P=0.000 01), grade a+b sperm motility (MD = 13.77, 95% CI: 7.06-20.48, P<0.000 1), sperm viability (MD = 10.32, 95% CI: 6.78-13.86, P<0.000 01) and pregnancy rate (OR = 3.53, 95% CI: 2.68-4.63, P<0.000 01), but a lower rate of adverse reactions (OR = 0.06, 95% CI: 0.01-0.23, P<0.000 01). There was no statistically significant difference in the percentage of morphologically abnormal sperm between TCM and Western medicine alone in the treatment of MII (MD = -7.53, 95% CI: -15.50-0.44, P = 0.06). CONCLUSION: TCM has a definite effectiveness and high safe in the treatment of male immune infertility.


Asunto(s)
Medicamentos Herbarios Chinos , Infertilidad Masculina , Humanos , Masculino , Medicamentos Herbarios Chinos/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Medicina Tradicional China/métodos , Fitoterapia
19.
Front Endocrinol (Lausanne) ; 15: 1412684, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39205681

RESUMEN

Background: In recent years, the decline in sperm quality in men has become a global trend. There is a close relationship between sperm quality and pregnancy outcome. There is a large body of literature supporting the role of plasma lipidome in male infertility, while the complex mechanisms between them and male infertility are still less clear. Systematic study of the causal relationship between plasma lipidome and MI can help to provide new therapeutic ideas and targets for male infertility. Methods: In this study, we used a two-sample Mendelian randomization analysis based on Genome-wide association studies pooled data of 179 causal relationships between plasma lipidome and male infertility. We used employed the inverse variance weighted method as the main analysis to assess causality between exposure and outcome, in addition to MR-Egger, Weighted median as complementary methods, and tests for multiplicity and heterogeneity. Results: We identified 13 plasma lipidome comprising 4 types of plasma lipidome that were associated with male infertility. Among these, 9 plasma lipidome were found to be protective factors, while 4 were risk factors. Notably, the largest proportion of these plasma lipidome were triglyceride types, with Sphingomyelin (d40:1) exhibiting the strongest association with male infertility. Conclusion: These findings contribute to the current better understanding of male infertility and provide new perspectives on the underlying etiology of male infertility as well as prevention and treatment strategies. In addition, clinical trial validation is needed to assess the potential of these plasma lipidome as biomarkers.


Asunto(s)
Estudio de Asociación del Genoma Completo , Infertilidad Masculina , Lipidómica , Análisis de la Aleatorización Mendeliana , Humanos , Masculino , Infertilidad Masculina/sangre , Infertilidad Masculina/genética , Infertilidad Masculina/epidemiología , Lípidos/sangre , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA