Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.420
Filter
1.
Cell Mol Life Sci ; 81(1): 379, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39222270

ABSTRACT

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.


Subject(s)
Infertility, Male , RNA, Small Interfering , Spermatogenesis , Spermatogenesis/genetics , Male , Humans , Animals , RNA, Small Interfering/metabolism , RNA, Small Interfering/genetics , Infertility, Male/genetics , Infertility, Male/metabolism , Reproductive Medicine , Mammals/genetics , Mammals/metabolism , Piwi-Interacting RNA
2.
Cells ; 13(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39273059

ABSTRACT

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.


Subject(s)
Biomarkers , Ferroptosis , Infertility, Male , Semen , Humans , Male , Semen/metabolism , Adult , Biomarkers/metabolism , Infertility, Male/metabolism , Infertility, Male/pathology , Coenzyme A Ligases/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Fertility , Spermatozoa/metabolism , Spermatozoa/pathology
3.
Elife ; 132024 Sep 13.
Article in English | MEDLINE | ID: mdl-39269275

ABSTRACT

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.


Subject(s)
Acrosome , Infertility, Male , Mice, Knockout , Spermatogenesis , Animals , Male , Acrosome/metabolism , Mice , Infertility, Male/genetics , Infertility, Male/metabolism , Spermatogenesis/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/deficiency , Golgi Apparatus/metabolism , Testis/metabolism
4.
Sci Rep ; 14(1): 20889, 2024 09 07.
Article in English | MEDLINE | ID: mdl-39244620

ABSTRACT

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.


Subject(s)
Cell Differentiation , Mice, Knockout , Microtubules , Spermatogenesis , Spermatogonia , Tubulin , Animals , Male , Tubulin/metabolism , Tubulin/genetics , Spermatogonia/metabolism , Spermatogonia/cytology , Spermatogenesis/genetics , Mice , Microtubules/metabolism , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology
5.
Genes (Basel) ; 15(9)2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39336792

ABSTRACT

Blood-testis barrier (BTB) genes are crucial for the cellular mechanisms of spermatogenesis as they protect against detrimental cytotoxic agents, chemicals, and pathogens, thereby maintaining a sterile environment necessary for sperm development. BTB proteins predominantly consist of extensive tight and gap junctions formed between Sertoli cells. These junctions form a crucial immunological barrier restricting the intercellular movement of substances and molecules within the adluminal compartment. Epithelial tight junctions are complex membrane structures composed of various integral membrane proteins, including claudins, zonula occludens-1, and occludin. Inter-testicular cell junction proteins undergo a constant process of degradation and renewal. In addition, the downregulation of genes crucial to the development and preservation of cell junctions could disrupt the functionality of the BTB, potentially leading to male infertility. Oxidative stress and inflammation may contribute to disrupted spermatogenesis, resulting in male infertility. L-cysteine is a precursor to glutathione, a crucial antioxidant that helps mitigate damage and inflammation resulting from oxidative stress. Preclinical research indicates that L-cysteine may offer protective benefits against testicular injury and promote the expression of BTB genes. This review emphasizes various BTB genes essential for preserving its structural integrity and facilitating spermatogenesis and male fertility. Furthermore, it consolidates various research findings suggesting that L-cysteine may promote the expression of BTB-associated genes, thereby aiding in the maintenance of testicular functions.


Subject(s)
Blood-Testis Barrier , Cysteine , Spermatogenesis , Male , Blood-Testis Barrier/metabolism , Humans , Animals , Cysteine/metabolism , Tight Junctions/metabolism , Oxidative Stress , Infertility, Male/genetics , Infertility, Male/metabolism , Sertoli Cells/metabolism , Sertoli Cells/drug effects , Testis/metabolism
6.
Int J Mol Sci ; 25(18)2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39337482

ABSTRACT

Mature, vital, and motile spermatozoa are essential for reaching the oocyte and binding to hyaluronic acid (HA) in the cumulus oophorus matrix. This study aims to determine the relationship between sperm-migration ability and HA-binding potential, as well as the relationship between sperm concentration and motility. Semen samples were collected from 702 men aged 20-56 years (median 34.8). We evaluated the sperm concentration and motility from basic semen analysis, the swim-up test (expressed as millions per mL and the migration efficiency percentage), and the hyaluronan-binding assay (HBA). A moderate positive correlation was found between the migration test results and HBA (R = 0.48). The highest correlation was observed between the concentration of motile spermatozoa and the migration test results (R = 0.85) and HBA (R = 0.4). The sperm migration efficiency strongly correlated with progressive motility (R = 0.6). Although significantly higher sperm migration was observed in patients with normal HBA results, the results of the functional tests were found to differ in some cases. For infertility treatment, the current diagnostic algorithm should be enhanced with more comprehensive seminological methods that assess the sperm-migration ability and HA-binding potential. We also recommend incorporating the swim-up method into the diagnostic protocol before planning assisted reproductive technology (ART) treatment.


Subject(s)
Hyaluronic Acid , Infertility, Male , Sperm Motility , Spermatozoa , Humans , Male , Hyaluronic Acid/metabolism , Adult , Spermatozoa/metabolism , Middle Aged , Infertility, Male/metabolism , Semen Analysis/methods , Sperm Count , Young Adult , Fertility
7.
Front Endocrinol (Lausanne) ; 15: 1427069, 2024.
Article in English | MEDLINE | ID: mdl-39324123

ABSTRACT

Hydrogen sulfide (H2S) is an endogenously produced signaling molecule that belongs to the group of gasotransmitters along with nitric oxide (NO) and carbon monoxide (CO). H2S plays a pivotal role in male reproductive processes. It is produced in various tissues and cells of the male reproductive system, including testicular tissue, Leydig and Sertoli cells, epididymis, seminal plasma, prostate, penile tissues, and sperm cells. This review aims to summarize the knowledge about the presence and effects of H2S in male reproductive tissues and outline possible therapeutic strategies in pathological conditions related to male fertility, e. g. spermatogenetic disorders and erectile dysfunction (ED). For instance, H2S supports spermatogenesis by maintaining the integrity of the blood-testicular barrier (BTB), stimulating testosterone production, and providing cytoprotective effects. In spermatozoa, H2S modulates sperm motility, promotes sperm maturation, capacitation, and acrosome reaction, and has significant cytoprotective effects. Given its vasorelaxant effects, it supports the erection of penile tissue. These findings suggest the importance and therapeutic potential of H2S in male reproduction, paving the way for further research and potential clinical applications.


Subject(s)
Hydrogen Sulfide , Reproduction , Spermatogenesis , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Male , Humans , Animals , Reproduction/drug effects , Reproduction/physiology , Spermatogenesis/drug effects , Erectile Dysfunction/drug therapy , Erectile Dysfunction/metabolism , Genitalia, Male/metabolism , Genitalia, Male/drug effects , Spermatozoa/drug effects , Spermatozoa/metabolism , Infertility, Male/metabolism , Infertility, Male/drug therapy , Testis/metabolism , Testis/drug effects
8.
Theranostics ; 14(14): 5621-5642, 2024.
Article in English | MEDLINE | ID: mdl-39310107

ABSTRACT

Rationale: Spermatogenesis is a highly organized cell differentiation process in mammals, involving mitosis, meiosis, and spermiogenesis. DIS3L2, which is primarily expressed in the cytoplasm, is an RNA exosome-independent ribonuclease. In female mice, Dis3l2-deficient oocytes fail to resume meiosis, resulting in arrest at the germinal vesicle stage and complete infertility. However, the role of DIS3L2 in germ cell development in males has remained largely unexplored. Methods: We established a pre-meiotic germ cell conditional knockout mouse model and investigated the biological function of DIS3L2 in spermatogenesis and male fertility through bulk RNA-seq and scRNA-seq analyses. Results: This study unveils that conditional ablation of Dis3l2 in pre-meiotic germ cells with Stra8-Cre mice impairs spermatogonial differentiation and hinders spermatocyte meiotic progression coupled with cell apoptosis. Such conditional ablation leads to defective spermatogenesis and sterility in adults. Bulk RNA-seq analysis revealed that Dis3l2 deficiency significantly disrupted the transcriptional expression pattern of genes related to the cell cycle, spermatogonial differentiation, and meiosis in Dis3l2 conditional knockout testes. Additionally, scRNA-seq analysis indicated that absence of DIS3L2 in pre-meiotic germ cells causes disrupted RNA metabolism, downregulated expression of cell cycle genes, and aberrant expression of spermatogonial differentiation genes, impeding spermatogonial differentiation. In meiotic spermatocytes, loss of DIS3L2 results in disturbed RNA metabolism, abnormal translation, and disrupted meiotic genes that perturb meiotic progression and induce cell apoptosis, leading to subsequent failure of spermatogenesis and male infertility. Conclusions: Collectively, these findings highlight the critical role of DIS3L2 ribonuclease-mediated RNA degradation in safeguarding the correct transcriptome during spermatogonial differentiation and spermatocyte meiotic progression, thus ensuring normal spermatogenesis and male fertility.


Subject(s)
Infertility, Male , Meiosis , Mice, Knockout , Spermatogenesis , Animals , Male , Spermatogenesis/genetics , Mice , Meiosis/genetics , Infertility, Male/genetics , Infertility, Male/metabolism , Cell Differentiation , Testis/metabolism , Spermatocytes/metabolism , Apoptosis/genetics , Spermatogonia/metabolism , Ribonucleases/metabolism , Ribonucleases/genetics , Female , Mice, Inbred C57BL , Germ Cells/metabolism
9.
Pak J Pharm Sci ; 37(3): 663-667, 2024 May.
Article in English | MEDLINE | ID: mdl-39340857

ABSTRACT

The study aimed to determine the in-vitro effect of metformin on total antioxidant capacity (TAC) of seminal samples of infertile male subjects. It was conducted from January to June 2022 on forty-four seminal plasma samples collected from male infertile patients, age ranging from 18 to 55 years. All 44 semen samples were treated as three distinct groups: (i) a control group (ii) a study group subjected to oxidative stress (OS) induction and (iii) a test group exposed to OS induction and subsequent treatment with metformin. OS was introduced by using commercially available 100µM hydrogen peroxide (H2O2) and incubated for twenty-four hours at 37ºC. After that 1 ml of 100 mmol/l concentration of commercially available Metformin (PHR 1331, CAS: 461-58-5) was administered to test group samples for additional 24h at 37ºC. Low levels of TAC were observed after OS induction in comparison to the control group (p=0.01). In test samples (after treatment with Metformin), a positive correlation of TAC with sperm count, normal sperm morphology and sperm motility were observed however, results were not significant. The antioxidant effect of Metformin was shown to improve the antioxidant capacity of OS induced samples and their sperm parameters in seminal plasma of infertile male subjects.


Subject(s)
Antioxidants , Infertility, Male , Metformin , Oxidative Stress , Semen , Spermatozoa , Male , Metformin/pharmacology , Oxidative Stress/drug effects , Humans , Infertility, Male/drug therapy , Infertility, Male/metabolism , Adult , Antioxidants/pharmacology , Young Adult , Spermatozoa/drug effects , Spermatozoa/metabolism , Middle Aged , Semen/drug effects , Semen/metabolism , Adolescent , Sperm Motility/drug effects , Hydrogen Peroxide/metabolism , Sperm Count
10.
Sheng Li Xue Bao ; 76(4): 631-642, 2024 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-39192795

ABSTRACT

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.


Subject(s)
Environmental Pollutants , Infertility, Male , Polycyclic Aromatic Hydrocarbons , Receptors, Aryl Hydrocarbon , Animals , Humans , Male , Environmental Pollutants/toxicity , Environmental Pollutants/adverse effects , Fertility/drug effects , Halogenated Diphenyl Ethers/adverse effects , Halogenated Diphenyl Ethers/toxicity , Infertility, Male/chemically induced , Infertility, Male/etiology , Infertility, Male/metabolism , Persistent Organic Pollutants/adverse effects , Persistent Organic Pollutants/metabolism , Polychlorinated Biphenyls/adverse effects , Polychlorinated Biphenyls/toxicity , Polycyclic Aromatic Hydrocarbons/adverse effects , Polycyclic Aromatic Hydrocarbons/toxicity , Receptors, Aryl Hydrocarbon/metabolism
11.
Int J Mol Sci ; 25(16)2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39201677

ABSTRACT

Zinc (Zn) is an essential trace element; it exhibits a plethora of physiological properties and biochemical functions. It plays a pivotal role in regulating the cell cycle, apoptosis, and DNA organization, as well as in protein, lipid, and carbohydrate metabolism. Among other important processes, Zn plays an essential role in reproductive health. The ZIP and ZnT proteins are responsible for the mobilization of Zn within the cell. Zn is an inert antioxidant through its interaction with a variety of proteins and enzymes to regulate the redox system, including metallothioneins (MTs), metalloenzymes, and gene regulatory proteins. The role of Zn in the reproductive system is of great importance; processes, such as spermatogenesis and sperm maturation that occur in the testicle and epididymis, respectively, depend on this element for their development and function. Zn modulates the synthesis of androgens, such as testosterone, for these reproductive processes, so Zn deficiency is related to alterations in sperm parameters that lead to male infertility.


Subject(s)
Epididymis , Testis , Zinc , Male , Zinc/metabolism , Epididymis/metabolism , Humans , Testis/metabolism , Animals , Spermatogenesis , Spermatozoa/metabolism , Infertility, Male/metabolism , Sperm Maturation/physiology
12.
Nat Commun ; 15(1): 6637, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122675

ABSTRACT

piRNAs are crucial for transposon silencing, germ cell maturation, and fertility in male mice. Here, we report on the genetic landscape of piRNA dysfunction in humans and present 39 infertile men carrying biallelic variants in 14 different piRNA pathway genes, including PIWIL1, GTSF1, GPAT2, MAEL, TDRD1, and DDX4. In some affected men, the testicular phenotypes differ from those of the respective knockout mice and range from complete germ cell loss to the production of a few morphologically abnormal sperm. A reduced number of pachytene piRNAs was detected in the testicular tissue of variant carriers, demonstrating impaired piRNA biogenesis. Furthermore, LINE1 expression in spermatogonia links impaired piRNA biogenesis to transposon de-silencing and serves to classify variants as functionally relevant. These results establish the disrupted piRNA pathway as a major cause of human spermatogenic failure and provide insights into transposon silencing in human male germ cells.


Subject(s)
DNA Transposable Elements , Infertility, Male , RNA, Small Interfering , Spermatogenesis , Testis , Male , Humans , Spermatogenesis/genetics , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , RNA, Small Interfering/metabolism , RNA, Small Interfering/genetics , DNA Transposable Elements/genetics , Animals , Testis/metabolism , Mice , Adult , Gene Silencing , Mice, Knockout , Argonaute Proteins/metabolism , Argonaute Proteins/genetics , Long Interspersed Nucleotide Elements/genetics , Spermatogonia/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Piwi-Interacting RNA
13.
J Cell Biol ; 223(11)2024 Nov 04.
Article in English | MEDLINE | ID: mdl-39158699

ABSTRACT

Ciliary beat and intraflagellar transport depend on dynein and kinesin motors. The kinesin-9 family members Kif6 and Kif9 are implicated in motile cilia motilities across protists and mammals. How they function and whether they act redundantly, however, remain unclear. Here, we show that Kif6 and Kif9 play distinct roles in mammals. Kif6 forms puncta that move bidirectionally along axonemes, whereas Kif9 appears to oscillate regionally on the ciliary central apparatus. Consistently, only Kif6 displays microtubule-based motor activity in vitro, and its ciliary localization requires its ATPase activity. Kif6 deficiency in mice disrupts coordinated ciliary beat across ependymal tissues and impairs cerebrospinal fluid flow, resulting in severe hydrocephalus and high mortality. Kif9 deficiency causes mild hydrocephalus without obviously affecting the ciliary beat or the lifespan. Kif6-/- and Kif9-/- males are infertile but exhibit oligozoospermia with poor sperm motility and defective forward motion of sperms, respectively. These results suggest Kif6 as a motor for cargo transport and Kif9 as a central apparatus regulator.


Subject(s)
Cilia , Kinesins , Mice, Knockout , Animals , Kinesins/metabolism , Kinesins/genetics , Cilia/metabolism , Male , Mice , Protein Transport , Sperm Motility/genetics , Hydrocephalus/metabolism , Hydrocephalus/genetics , Hydrocephalus/pathology , Mice, Inbred C57BL , Axoneme/metabolism , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Humans , Microtubules/metabolism
14.
EMBO J ; 43(19): 4197-4227, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39160277

ABSTRACT

In mammals, the transition from mitosis to meiosis facilitates the successful production of gametes. However, the regulatory mechanisms that control meiotic initiation remain unclear, particularly in the context of complex histone modifications. Herein, we show that KDM2A, acting as a lysine demethylase targeting H3K36me3 in male germ cells, plays an essential role in modulating meiotic entry and progression. Conditional deletion of Kdm2a in mouse pre-meiotic germ cells results in complete male sterility, with spermatogenesis ultimately arrested at the zygotene stage of meiosis. KDM2A deficiency disrupts H3K36me2/3 deposition in c-KIT+ germ cells, characterized by a reduction in H3K36me2 but a dramatic increase in H3K36me3. Furthermore, KDM2A recruits the transcription factor E2F1 and its co-factor HCFC1 to the promoters of key genes required for meiosis entry and progression, such as Stra8, Meiosin, Spo11, and Sycp1. Collectively, our study unveils an essential role for KDM2A in mediating H3K36me2/3 deposition and controlling the programmed gene expression necessary for the transition from mitosis to meiosis during spermatogenesis.


Subject(s)
E2F1 Transcription Factor , Jumonji Domain-Containing Histone Demethylases , Meiosis , Spermatogenesis , Animals , Male , Mice , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Spermatogenesis/genetics , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/genetics , Host Cell Factor C1/metabolism , Host Cell Factor C1/genetics , Histones/metabolism , Histones/genetics , Mice, Knockout , Infertility, Male/genetics , Infertility, Male/metabolism , Histone Demethylases
15.
Elife ; 132024 Aug 20.
Article in English | MEDLINE | ID: mdl-39163107

ABSTRACT

Ribonucleoprotein (RNP) granules are membraneless electron-dense structures rich in RNAs and proteins, and involved in various cellular processes. Two RNP granules in male germ cells, intermitochondrial cement and the chromatoid body (CB), are associated with PIWI-interacting RNAs (piRNAs) and are required for transposon silencing and spermatogenesis. Other RNP granules in male germ cells, the reticulated body and CB remnants, are also essential for spermiogenesis. In this study, we disrupted FBXO24, a testis-enriched F-box protein, in mice and found numerous membraneless electron-dense granules accumulated in sperm flagella. Fbxo24 knockout (KO) mice exhibited malformed flagellar structures, impaired sperm motility, and male infertility, likely due to the accumulation of abnormal granules. The amount and localization of known RNP granule-related proteins were not disrupted in Fbxo24 KO mice, suggesting that the accumulated granules were distinct from known RNP granules. Further studies revealed that RNAs and two importins, IPO5 and KPNB1, abnormally accumulated in Fbxo24 KO spermatozoa and that FBXO24 could ubiquitinate IPO5. In addition, IPO5 and KPNB1 were recruited to stress granules, RNP complexes, when cells were treated with oxidative stress or a proteasome inhibitor. These results suggest that FBXO24 is involved in the degradation of IPO5, disruption of which may lead to the accumulation of abnormal RNP granules in sperm flagella.


Subject(s)
F-Box Proteins , Infertility, Male , Mice, Knockout , Sperm Tail , Male , Animals , Infertility, Male/genetics , Infertility, Male/metabolism , Mice , Sperm Tail/metabolism , F-Box Proteins/metabolism , F-Box Proteins/genetics , Cytoplasmic Granules/metabolism , Spermatozoa/metabolism
16.
Reprod Sci ; 31(10): 2943-2956, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39090335

ABSTRACT

Research into the impacts of oxidative stress (OS), and hormonal balance on reproductive potential has increased over the last 40 years possibly due to rising male infertility. Decreased antioxidant levels and increased OS in tissues result from hormonal imbalance, which in turn leads to male infertility. Increased reactive oxygen species (ROS) generation in seminal plasma has been linked to many lifestyle factors such as alcohol and tobacco use, toxicant exposure, obesity, varicocele, stress, and aging. This article provides an overview of the crosslink between OS and gonadal hormone disruption, as well as a potential mode of action in male infertility. Disrupting the equilibrium between ROS generation and the antioxidant defense mechanism in the male reproductive system may affect key hormonal regulators of male reproductive activities. Unchecked ROS production may cause direct injury on reproductive tissues or could disrupt normal regulatory mechanisms of the hypothalamic-pituitary-gonadal (HPG) axis and its interaction with other endocrine axes, both of which have negative effects on male reproductive health and can lead to male infertility.


Subject(s)
Infertility, Male , Oxidative Stress , Reproduction , Male , Humans , Oxidative Stress/physiology , Infertility, Male/metabolism , Infertility, Male/etiology , Infertility, Male/physiopathology , Animals , Reproduction/physiology , Reactive Oxygen Species/metabolism , Hypothalamo-Hypophyseal System/metabolism
17.
Reprod Sci ; 31(10): 2957-2971, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39168918

ABSTRACT

Subclinical hyperthyroidism (SCH) is a subtle thyroid dysfunction marked by decreased serum thyroid-stimulating hormone (TSH) levels while maintaining a normal thyroid hormone profile. Despite its mild nature, SCH can significantly impact various physiological functions, including male reproductive health. However, the effects of SCH on reproductive hormones and semen quality are less understood compared to overt thyroid disorders. This study employed extensive search methods across various databases from January 2000 to February 2024 to explore the relationship between SCH and Hormonal and Seminal Perspectives. Effect sizes, estimated using the standardized mean difference (SMD) and pooled with a Random-effect model, provided significant insights from 748 participants. Included studies adhered to the following criteria: Patients (male individuals with SCH), Intervention (assessment of reproductive hormones and semen quality), Comparison (SCH patients versus healthy controls), and Outcome (changes in reproductive factors). Significant alterations in reproductive hormones were observed in SCH patients, including reduced LH levels (SMD = - 0.20; p = 0.007), elevated FSH levels (SMD = 0.25; p = 0.002), and stable testosterone levels (SMD = - 0.05; p = 0.50). Regarding thyroid profile, SCH was associated with increased FT3 (SMD = 0.15; p < 0.001) and FT4 (SMD = 0.08; p = 0.002) levels, along with decreased TSH levels (SMD = - 2.00; p < 0.001). Adverse effects on semen quality were also observed. These findings underscore the need to incorporate thyroid health assessments in the evaluation of male infertility, recognizing the impact of minor thyroid hormone deviations on reproductive outcomes.


Subject(s)
Hyperthyroidism , Reproductive Health , Semen Analysis , Humans , Male , Hyperthyroidism/blood , Thyroid Hormones/blood , Testosterone/blood , Infertility, Male/blood , Infertility, Male/metabolism , Infertility, Male/diagnosis , Luteinizing Hormone/blood , Semen/metabolism
18.
Int Braz J Urol ; 50(5): 530-560, 2024.
Article in English | MEDLINE | ID: mdl-39106113

ABSTRACT

Varicocele can reduce male fertility potential through various oxidative stress mechanisms. Excessive production of reactive oxygen species may overwhelm the sperm's defenses against oxidative stress, damaging the sperm chromatin. Sperm DNA fragmentation, in the form of DNA strand breaks, is recognized as a consequence of the oxidative stress cascade and is commonly found in the ejaculates of men with varicocele and fertility issues. This paper reviews the current knowledge regarding the association between varicocele, oxidative stress, sperm DNA fragmentation, and male infertility, and examines the role of varicocele repair in alleviating oxidative-sperm DNA fragmentation in these patients. Additionally, we highlight areas for further research to address knowledge gaps relevant to clinical practice.


Subject(s)
DNA Fragmentation , Infertility, Male , Oxidative Stress , Spermatozoa , Varicocele , Humans , Male , Varicocele/physiopathology , Varicocele/complications , Oxidative Stress/physiology , Infertility, Male/etiology , Infertility, Male/genetics , Infertility, Male/physiopathology , Infertility, Male/metabolism , Spermatozoa/physiology , Spermatozoa/metabolism , Reactive Oxygen Species/metabolism
19.
Cytokine ; 182: 156701, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39089215

ABSTRACT

Among the many factors with a proven relation to semen quality and male fertility, the determination of seminal plasma cytokines provides a promising direction for research into the identification of factors connected with male infertility. The interleukins: IL-1α, -1ß, -2, -4, -6, -8, -10, -12p40, -12p70, -18, IFNγ, and GM-CSF, total oxidant (TOS) and antioxidant (TAS) status, were simultaneously examined in seminal plasmas and blood sera in terato- (n = 32), asthenoterato- (n = 33), and oligoasthenoteratozoospermic (n = 29) infertile men and in normozoospermic fertile men (n = 20). Our research shows different cytokine composition of the sera and seminal plasmas in all studied groups, along with much higher concentrations of seminal plasma GM-CSF, IFNγ, IL-1α, IL-4, IL-6, and IL-8 and lower IL-18 and TOS in the comparison to their sera levels. The seminal plasma concentrations of GM-CSF, IFNγ, IL-1α, -4, and -6 differ significantly between fertile and infertile as well as between teratozoospermic, asthenoteratozoospermic, and oligoasthenoteratozoospermic groups. The indication of the cause of different concentrations of cytokines in seminal plasmas of infertile men, and their associations with semen parameters and oxidative status, may be a promising direction for the search for new therapeutic targets that would directly affect the cells and tissues of male reproductive organs.


Subject(s)
Antioxidants , Biomarkers , Cytokines , Infertility, Male , Semen , Humans , Male , Semen/metabolism , Cytokines/metabolism , Cytokines/blood , Antioxidants/metabolism , Adult , Biomarkers/metabolism , Biomarkers/blood , Infertility, Male/metabolism , Infertility, Male/blood , Oxidative Stress , Semen Analysis/methods , Spermatozoa/metabolism , Asthenozoospermia/metabolism
20.
Reprod Biol Endocrinol ; 22(1): 83, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39020374

ABSTRACT

BACKGROUND: Besides adenine triphosphate (ATP) production for sustaining motility, the mitochondria of sperm also host other critical cellular functions during germ cell development and fertilization including calcium homeostasis, generation of reactive oxygen species (ROS), apoptosis, and in some cases steroid hormone biosynthesis. Normal mitochondrial membrane potential with optimal mitochondrial performance is essential for sperm motility, capacitation, acrosome reaction, and DNA integrity. RESULTS: Defects in the sperm mitochondrial function can severely harm the fertility potential of males. The role of sperm mitochondria in fertilization and its final fate after fertilization is still controversial. Here, we review the current knowledge on human sperm mitochondria characteristics and their physiological and pathological conditions, paying special attention to improvements in assistant reproductive technology and available treatments to ameliorate male infertility. CONCLUSION: Although mitochondrial variants associated with male infertility have potential clinical use, research is limited. Further understanding is needed to determine how these characteristics lead to adverse pregnancy outcomes and affect male fertility potential.


Subject(s)
Fertility , Infertility, Male , Mitochondria , Spermatozoa , Humans , Male , Infertility, Male/physiopathology , Infertility, Male/metabolism , Spermatozoa/metabolism , Spermatozoa/physiology , Mitochondria/metabolism , Mitochondria/physiology , Fertility/physiology , Sperm Motility/physiology , Female , Reactive Oxygen Species/metabolism , Animals
SELECTION OF CITATIONS
SEARCH DETAIL