Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.394
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 186(13): 2897-2910.e19, 2023 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-37295417

RESUMO

Sperm motility is crucial for successful fertilization. Highly decorated doublet microtubules (DMTs) form the sperm tail skeleton, which propels the movement of spermatozoa. Using cryo-electron microscopy (cryo-EM) and artificial intelligence (AI)-based modeling, we determined the structures of mouse and human sperm DMTs and built an atomic model of the 48-nm repeat of the mouse sperm DMT. Our analysis revealed 47 DMT-associated proteins, including 45 microtubule inner proteins (MIPs). We identified 10 sperm-specific MIPs, including seven classes of Tektin5 in the lumen of the A tubule and FAM166 family members that bind the intra-tubulin interfaces. Interestingly, the human sperm DMT lacks some MIPs compared with the mouse sperm DMT. We also discovered variants in 10 distinct MIPs associated with a subtype of asthenozoospermia characterized by impaired sperm motility without evident morphological abnormalities. Our study highlights the conservation and tissue/species specificity of DMTs and expands the genetic spectrum of male infertility.


Assuntos
Inteligência Artificial , Infertilidade Masculina , Masculino , Humanos , Microscopia Crioeletrônica , Motilidade dos Espermatozoides/genética , Sêmen , Espermatozoides , Microtúbulos/metabolismo , Cauda do Espermatozoide/química , Cauda do Espermatozoide/metabolismo , Proteínas dos Microtúbulos/química , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo
2.
PLoS Genet ; 20(7): e1011357, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39074078

RESUMO

Hexokinase (HK) catalyzes the first irreversible rate-limiting step in glycolysis that converts glucose to glucose-6-phosphate. HK1 is ubiquitously expressed in the brain, erythrocytes, and other tissues where glycolysis serves as the major source of ATP production. Spermatogenic cell-specific type 1 hexokinase (HK1S) is expressed in sperm but its physiological role in male mice is still unknown. In this study, we generate Hk1s knockout mice using the CRISPR/Cas9 system to study the gene function in vivo. Hk1s mRNA is exclusively expressed in testes starting from postnatal day 18 and continuing to adulthood. HK1S protein is specifically localized in the outer surface of the sperm fibrous sheath (FS). Depletion of Hk1s leads to infertility in male mice and reduces sperm glycolytic pathway activity, yet they have normal motile parameters and ATP levels. In addition, by using in vitro fertilization (IVF), Hk1s deficient sperms are unable to fertilize cumulus-intact or cumulus-free oocytes, but can normally fertilize zona pellucida-free oocytes. Moreover, Hk1s deficiency impairs sperm migration into the oviduct, reduces acrosome reaction, and prevents capacitation-associated increases in tyrosine phosphorylation, which are probable causes of infertility. Taken together, our results reveal that HK1S plays a critical role in sperm function and male fertility in mice.


Assuntos
Fertilidade , Hexoquinase , Infertilidade Masculina , Camundongos Knockout , Capacitação Espermática , Espermatozoides , Tirosina , Animais , Hexoquinase/genética , Hexoquinase/metabolismo , Masculino , Camundongos , Fosforilação , Espermatozoides/metabolismo , Capacitação Espermática/genética , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Fertilidade/genética , Tirosina/metabolismo , Feminino , Testículo/metabolismo , Motilidade dos Espermatozoides/genética , Glicólise , Espermatogênese/genética
3.
PLoS Genet ; 20(6): e1011337, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38935810

RESUMO

Sperm heads contain not only the nucleus but also the acrosome which is a distinctive cap-like structure located anterior to the nucleus and is derived from the Golgi apparatus. The Golgi Associated RAB2 Interactors (GARINs; also known as FAM71) protein family shows predominant expression in the testis and all possess a RAB2-binding domain which confers binding affinity to RAB2, a small GTPase that is responsible for membrane transport and vesicle trafficking. Our previous study showed that GARIN1A and GARIN1B are important for acrosome biogenesis and that GARIN1B is indispensable for male fertility in mice. Here, we generated KO mice of other Garins, namely Garin2, Garin3, Garin4, Garin5a, and Garin5b (Garin2-5b). Using computer-assisted morphological analysis, we found that the loss of each Garin2-5b resulted in aberrant sperm head morphogenesis. While the fertilities of Garin2-/- and Garin4-/- males are normal, Garin5a-/- and Garin5b-/- males are subfertile, and Garin3-/- males are infertile. Further analysis revealed that Garin3-/- males exhibited abnormal acrosomal morphology, but not as severely as Garin1b-/- males; instead, the amounts of membrane proteins, particularly ADAM family proteins, decreased in Garin3 KO spermatozoa. Moreover, only Garin4 KO mice exhibit vacuoles in the sperm head. These results indicate that GARINs assure correct head morphogenesis and some members of the GARIN family function distinctively in male fertility.


Assuntos
Fertilidade , Infertilidade Masculina , Camundongos Knockout , Cabeça do Espermatozoide , Animais , Masculino , Camundongos , Acrossomo/metabolismo , Fertilidade/genética , Complexo de Golgi/metabolismo , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Morfogênese/genética , Proteína rab2 de Ligação ao GTP/metabolismo , Proteína rab2 de Ligação ao GTP/genética , Cabeça do Espermatozoide/metabolismo , Espermatozoides/metabolismo , Testículo/metabolismo , Testículo/crescimento & desenvolvimento
4.
Development ; 150(24)2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37997706

RESUMO

Sperm with normal morphology and motility are essential for successful fertilization, and the strong attachment of the sperm head-tail coupling apparatus to the nuclear envelope during spermatogenesis is required to ensure the integrity of sperm for capacitation and fertilization. Here, we report that Arrdc5 is associated with spermatogenesis. The Arrdc5 knockout mouse model showed male infertility characterized by a high bent-head rate and reduced motility in sperm, which led to capacitation defects and subsequent fertilization failure. Through mass spectrometry, we found that ARRDC5 affects spermatogenesis by affecting NDC1 and SUN5. We further found that ARRDC5 might affect the vesicle-trafficking protein SEC22A-mediated transport and localization of NDC1, SUN5 and other head-tail coupling apparatus-related proteins that are responsible for initiating the attachment of the sperm head and tail. We finally performed intracytoplasmic sperm injection as a way to explore therapeutic strategies. Our findings demonstrate the essential role and the underlying molecular mechanism of ARRDC5 in anchoring the sperm head to the tail during spermatogenesis.


Assuntos
Infertilidade Masculina , Sêmen , Humanos , Animais , Camundongos , Masculino , Sêmen/metabolismo , Espermatogênese/genética , Espermatozoides/metabolismo , Cabeça do Espermatozoide/metabolismo , Proteínas/metabolismo , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Camundongos Knockout , Proteínas de Membrana/metabolismo
5.
EMBO Rep ; 25(6): 2722-2742, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38773322

RESUMO

Alpha, beta, and gamma tubulins are essential building blocks for all eukaryotic cells. The functions of the non-canonical tubulins, delta, epsilon, and zeta, however, remain poorly understood and their requirement in mammalian development untested. Herein we have used a spermatogenesis model to define epsilon tubulin (TUBE1) function in mice. We show that TUBE1 is essential for the function of multiple complex microtubule arrays, including the meiotic spindle, axoneme and manchette and in its absence, there is a dramatic loss of germ cells and male sterility. Moreover, we provide evidence for the interplay between TUBE1 and katanin-mediated microtubule severing, and for the sub-specialization of individual katanin paralogs in the regulation of specific microtubule arrays.


Assuntos
Katanina , Microtúbulos , Espermatogênese , Tubulina (Proteína) , Animais , Masculino , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Camundongos , Katanina/metabolismo , Katanina/genética , Adenosina Trifosfatases/metabolismo , Adenosina Trifosfatases/genética , Células Germinativas/metabolismo , Fuso Acromático/metabolismo , Espermatozoides/metabolismo , Infertilidade Masculina/metabolismo , Infertilidade Masculina/genética , Camundongos Knockout , Axonema/metabolismo
6.
Dev Biol ; 512: 13-25, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38703942

RESUMO

Drosophila melanogaster is an ideal model organism for investigating spermatogenesis due to its powerful genetics, conserved genes and visible morphology of germ cells during sperm production. Our previous work revealed that ocnus (ocn) knockdown resulted in male sterility, and CG9920 was identified as a significantly downregulated protein in fly abdomen after ocn knockdown, suggesting a role of CG9920 in male reproduction. In this study, we found that CG9920 was highly expressed in fly testes. CG9920 knockdown in fly testes caused male infertility with no mature sperms in seminal vesicles. Immunofluorescence staining showed that depletion of CG9920 resulted in scattered spermatid nuclear bundles, fewer elongation cones that did not migrate to the anterior region of the testis, and almost no individualization complexes. Transmission electron microscopy revealed that CG9920 knockdown severely disrupted mitochondrial morphogenesis during spermatogenesis. Notably, we found that CG9920 might not directly interact with Ocn, but rather was inhibited by STAT92E, which itself was indirectly affected by Ocn. We propose a possible novel pathway essential for spermatogenesis in D. melanogaster, whereby Ocn indirectly induces CG9920 expression, potentially counteracting its inhibition by the JAK-STAT signaling pathway.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Mitocôndrias , Espermatogênese , Testículo , Animais , Espermatogênese/genética , Espermatogênese/fisiologia , Masculino , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Mitocôndrias/metabolismo , Testículo/metabolismo , Morfogênese/genética , Transdução de Sinais , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Técnicas de Silenciamento de Genes , Fatores de Transcrição STAT/metabolismo , Espermátides/metabolismo
7.
Hum Mol Genet ; 33(1): 48-63, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37740387

RESUMO

Assisted reproductive technologies (ART) account for 1-6% of births in developed countries. While most children conceived are healthy, increases in birth and genomic imprinting defects have been reported; such abnormal outcomes have been attributed to underlying parental infertility and/or the ART used. Here, we assessed whether paternal genetic and lifestyle factors, that are associated with male infertility and affect the sperm epigenome, can influence ART outcomes. We examined how paternal factors, haploinsufficiency for Dnmt3L, an important co-factor for DNA methylation reactions, and/or diet-induced obesity, in combination with ART (superovulation, in vitro fertilization, embryo culture and embryo transfer), could adversely influence embryo development and DNA methylation patterning in mice. While male mice fed high-fat diets (HFD) gained weight and showed perturbed metabolic health, their sperm DNA methylation was minimally affected by the diet. In contrast, Dnmt3L haploinsufficiency induced a marked loss of DNA methylation in sperm; notably, regions affected were associated with neurodevelopmental pathways and enriched in young retrotransposons, sequences that can have functional consequences in the next generation. Following ART, placental imprinted gene methylation and growth parameters were impacted by one or both paternal factors. For embryos conceived by natural conception, abnormality rates were similar for WT and Dnmt3L+/- fathers. In contrast, paternal Dnmt3L+/- genotype, as compared to WT fathers, resulted in a 3-fold increase in the incidence of morphological abnormalities in embryos generated by ART. Together, the results indicate that embryonic morphological and epigenetic defects associated with ART may be exacerbated in offspring conceived by fathers with sperm epimutations.


Assuntos
Infertilidade Masculina , Placenta , Criança , Gravidez , Masculino , Humanos , Feminino , Animais , Camundongos , Placenta/metabolismo , Incidência , Sêmen , Reprodução/genética , Metilação de DNA , Técnicas de Reprodução Assistida/efeitos adversos , Espermatozoides/metabolismo , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Pai
8.
Development ; 149(11)2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35587122

RESUMO

The sperm flagellum is essential for male fertility, and defects in flagellum biogenesis are associated with male infertility. Deficiency of coiled-coil domain-containing (CCDC) 42 (CCDC42) is specifically associated with malformation of mouse sperm flagella. Here, we find that the testis-specific protein CCDC38 interacts with CCDC42, localizing on the manchette and sperm tail during spermiogenesis. Inactivation of CCDC38 in male mice results in a distorted manchette, multiple morphological abnormalities of the flagella of spermatozoa and eventually male sterility. Furthermore, we find that CCDC38 interacts with intraflagellar transport protein 88 (IFT88), as well as outer dense fibrous 2 (ODF2), and the knockout of Ccdc38 reduces transport of ODF2 to the flagellum. Altogether, our results uncover the essential role of CCDC38 in sperm flagellum biogenesis, and suggest that some mutations of these genes might be associated with male infertility in humans.


Assuntos
Fertilidade , Infertilidade Masculina , Cauda do Espermatozoide , Animais , Fertilidade/genética , Proteínas de Choque Térmico/metabolismo , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Cauda do Espermatozoide/metabolismo , Espermatozoides/metabolismo , Testículo/metabolismo
9.
Development ; 149(12)2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35616329

RESUMO

The perinuclear theca (PT) is a cytoskeletal element encapsulating the sperm nucleus; however, the physiological roles of the PT in sperm are largely uncertain. Here, we reveal that ACTRT1, ACTRT2, ACTL7A and ACTL9 proteins interact to form a multimeric complex and localize to the subacrosomal region of spermatids. Furthermore, we engineered Actrt1-knockout (KO) mice to define the functions of ACTRT1. Despite normal sperm count and motility, Actrt1-KO males were severely subfertile owing to a deficiency in fertilization. Loss of ACTRT1 caused a high incidence of malformed heads and detachment of acrosomes from sperm nuclei, caused by loosened acroplaxome structure during spermiogenesis. Furthermore, Actrt1-KO sperm showed reduced ACTL7A and PLCζ protein content as a potential cause of fertilization defects. Moreover, we reveal that ACTRT1 anchors developing acrosomes to the nucleus, likely by interacting with the inner acrosomal membrane protein SPACA1 and the nuclear envelope proteins PARP11 and SPATA46. Loss of ACTRT1 weakened the interaction between ACTL7A and SPACA1. Our study and recent findings of ACTL7A/ACTL9-deficient sperm together reveal that the sperm PT-specific ARP complex mediates the acrosome-nucleus connection.


Assuntos
Acrossomo , Infertilidade Masculina , Acrossomo/metabolismo , Animais , Humanos , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Espermátides/metabolismo , Espermatogênese/genética , Espermatozoides/metabolismo
10.
Annu Rev Genet ; 51: 265-285, 2017 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-28853925

RESUMO

Sexual reproduction crucially depends on the production of sperm in males and oocytes in females. Both types of gamete arise from the same precursor, the germ cells. We review the events that characterize the development of germ cells during fetal life as they commit to, and prepare for, oogenesis or spermatogenesis. In females, fetal germ cells enter meiosis, whereas in males they delay meiosis and instead lose pluripotency, activate an irreversible program of prospermatogonial differentiation, and temporarily cease dividing. Both pathways involve sex-specific molecular signals from the somatic cells of the developing gonads and a suite of intrinsic receptors, signal transducers, transcription factors, RNA stability factors, and epigenetic modulators that act in complex, interconnected positive and negative regulatory networks. Understanding these networks is important in the contexts of the etiology, diagnosis, and treatment of infertility and gonadal cancers, and in efforts to augment human and animal fertility using stem cell approaches.


Assuntos
Infertilidade Feminina/genética , Infertilidade Masculina/genética , Oogênese/genética , Processos de Determinação Sexual , Diferenciação Sexual/genética , Espermatogênese/genética , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Humanos , Infertilidade Feminina/metabolismo , Infertilidade Feminina/patologia , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Masculino , Meiose , Oócitos/citologia , Oócitos/crescimento & desenvolvimento , Oócitos/metabolismo , Óvulo/citologia , Óvulo/crescimento & desenvolvimento , Óvulo/metabolismo , Transdução de Sinais , Espermatozoides/citologia , Espermatozoides/crescimento & desenvolvimento , Espermatozoides/metabolismo
11.
Biol Cell ; 116(6): e2300127, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38593304

RESUMO

BACKGROUND: Spermatogenesis is a fundamental process crucial for male reproductive health and fertility. Exosomes, small membranous vesicles released by various cell types, have recently garnered attention for their role in intercellular communication. OBJECTIVE: This review aims to comprehensively explore the role of exosomes in regulating spermatogenesis, focusing on their involvement in testicular development and cell-to-cell communication. METHODS: A systematic examination of literature was conducted to gather relevant studies elucidating the biogenesis, composition, and functions of exosomes in the context of spermatogenesis. RESULTS: Exosomes play a pivotal role in orchestrating the complex signaling networks required for proper spermatogenesis. They facilitate the transfer of key regulatory molecules between different cell populations within the testes, including Sertoli cells, Leydig cells, and germ cells. CONCLUSION: The emerging understanding of exosome-mediated communication sheds light on novel mechanisms underlying spermatogenesis regulation. Further research in this area holds promise for insights into male reproductive health and potential therapeutic interventions.


Assuntos
Exossomos , Infertilidade Masculina , Espermatogênese , Masculino , Espermatogênese/fisiologia , Exossomos/metabolismo , Humanos , Infertilidade Masculina/terapia , Infertilidade Masculina/metabolismo , Animais , Comunicação Celular , Células de Sertoli/metabolismo , Testículo/metabolismo , Células Intersticiais do Testículo/metabolismo , Transdução de Sinais
12.
Mol Cell ; 66(1): 89-101.e8, 2017 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-28366643

RESUMO

Histone replacement by transition proteins (TPs) and protamines (Prms) constitutes an essential step for the successful production of functional male gametes, yet nothing is known on the underlying functional interplay between histones, TPs, and Prms. Here, by studying spermatogenesis in the absence of a spermatid-specific histone variant, H2A.L.2, we discover a fundamental mechanism involved in the transformation of nucleosomes into nucleoprotamines. H2A.L.2 is synthesized at the same time as TPs and enables their loading onto the nucleosomes. TPs do not displace histones but rather drive the recruitment and processing of Prms, which are themselves responsible for histone eviction. Altogether, the incorporation of H2A.L.2 initiates and orchestrates a series of successive transitional states that ultimately shift to the fully compacted genome of the mature spermatozoa. Hence, the current view of histone-to-nucleoprotamine transition should be revisited and include an additional step with H2A.L.2 assembly prior to the action of TPs and Prms.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , Histonas/metabolismo , Nucleossomos/metabolismo , Protaminas/metabolismo , Espermatogênese , Espermatozoides/metabolismo , Animais , Células COS , Chlorocebus aethiops , Cromatina/genética , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Biologia Computacional , Bases de Dados Genéticas , Fertilidade , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Genoma , Histonas/deficiência , Histonas/genética , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Infertilidade Masculina/fisiopatologia , Masculino , Camundongos da Linhagem 129 , Camundongos Knockout , Nucleossomos/genética , Fenótipo , Espermatogênese/genética , Espermatozoides/patologia , Transfecção
13.
Cell Mol Life Sci ; 81(1): 212, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38724675

RESUMO

Leydig cells are essential components of testicular interstitial tissue and serve as a primary source of androgen in males. A functional deficiency in Leydig cells often causes severe reproductive disorders; however, the transcriptional programs underlying the fate decisions and steroidogenesis of these cells have not been fully defined. In this study, we report that the homeodomain transcription factor PBX1 is a master regulator of Leydig cell differentiation and testosterone production in mice. PBX1 was highly expressed in Leydig cells and peritubular myoid cells in the adult testis. Conditional deletion of Pbx1 in Leydig cells caused spermatogenic defects and complete sterility. Histological examinations revealed that Pbx1 deletion impaired testicular structure and led to disorganization of the seminiferous tubules. Single-cell RNA-seq analysis revealed that loss of Pbx1 function affected the fate decisions of progenitor Leydig cells and altered the transcription of genes associated with testosterone synthesis in the adult testis. Pbx1 directly regulates the transcription of genes that play important roles in steroidogenesis (Prlr, Nr2f2 and Nedd4). Further analysis demonstrated that deletion of Pbx1 leads to a significant decrease in testosterone levels, accompanied by increases in pregnenolone, androstenedione and luteinizing hormone. Collectively, our data revealed that PBX1 is indispensable for maintaining Leydig cell function. These findings provide insights into testicular dysgenesis and the regulation of hormone secretion in Leydig cells.


Assuntos
Infertilidade Masculina , Células Intersticiais do Testículo , Fator de Transcrição 1 de Leucemia de Células Pré-B , Testículo , Testosterona , Animais , Masculino , Células Intersticiais do Testículo/metabolismo , Células Intersticiais do Testículo/patologia , Fator de Transcrição 1 de Leucemia de Células Pré-B/metabolismo , Fator de Transcrição 1 de Leucemia de Células Pré-B/genética , Camundongos , Testosterona/metabolismo , Testículo/metabolismo , Testículo/patologia , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Infertilidade Masculina/metabolismo , Diferenciação Celular/genética , Espermatogênese/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout
14.
Cell Mol Life Sci ; 81(1): 317, 2024 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-39066891

RESUMO

Inner dynein arms (IDAs) are formed from a protein complex that is essential for appropriate flagellar bending and beating. IDA defects have previously been linked to the incidence of asthenozoospermia (AZS) and male infertility. The testes-enriched ZMYND12 protein is homologous with an IDA component identified in Chlamydomonas. ZMYND12 deficiency has previously been tied to infertility in males, yet the underlying mechanism remains uncertain. Here, a CRISPR/Cas9 approach was employed to generate Zmynd12 knockout (Zmynd12-/-) mice. These Zmynd12-/- mice exhibited significant male subfertility, reduced sperm motile velocity, and impaired capacitation. Through a combination of co-immunoprecipitation and mass spectrometry, ZMYND12 was found to interact with TTC29 and PRKACA. Decreases in the levels of PRKACA were evident in the sperm of these Zmynd12-/- mice, suggesting that this change may account for the observed drop in male fertility. Moreover, in a cohort of patients with AZS, one patient carrying a ZMYND12 variant was identified, expanding the known AZS-related variant spectrum. Together, these findings demonstrate that ZMYND12 is essential for flagellar beating, capacitation, and male fertility.


Assuntos
Infertilidade Masculina , Camundongos Knockout , Motilidade dos Espermatozoides , Animais , Masculino , Motilidade dos Espermatozoides/genética , Camundongos , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Dineínas/metabolismo , Dineínas/genética , Espermatozoides/metabolismo , Humanos , Astenozoospermia/genética , Astenozoospermia/metabolismo , Astenozoospermia/patologia , Capacitação Espermática/genética , Camundongos Endogâmicos C57BL , Sistemas CRISPR-Cas
15.
Mol Cell Proteomics ; 22(6): 100556, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37087050

RESUMO

Non-obstructive azoospermia (NOA), the most severe form of male infertility, could be treated with intracytoplasmic sperm injection, providing spermatozoa were retrieved with the microdissection testicular sperm extraction (mTESE). We hypothesized that testis-specific and germ cell-specific proteins would facilitate flow cytometry-assisted identification of rare spermatozoa in semen cell pellets of NOA patients, thus enabling non-invasive diagnostics prior to mTESE. Data mining, targeted proteomics, and immunofluorescent microscopy identified and verified a panel of highly testis-specific proteins expressed at the continuum of germ cell differentiation. Late germ cell-specific proteins AKAP4_HUMAN and ASPX_HUMAN (ACRV1 gene) revealed exclusive localization in spermatozoa tails and acrosomes, respectively. A multiplex imaging flow cytometry assay facilitated fast and unambiguous identification of rare but morphologically intact AKAP4+/ASPX+/Hoechst+ spermatozoa within debris-laden semen pellets of NOA patients. While the previously suggested markers for spermatozoa retrieval suffered from low diagnostic specificity, the multistep gating strategy and visualization of AKAP4+/ASPX+/Hoechst+ cells with elongated tails and acrosome-capped nuclei facilitated fast and unambiguous identification of the mature intact spermatozoa. AKAP4+/ASPX+/Hoechst+ assay may emerge as a noninvasive test to predict retrieval of morphologically intact spermatozoa by mTESE, thus improving diagnostics and treatment of severe forms of male infertility.


Assuntos
Azoospermia , Infertilidade Masculina , Masculino , Humanos , Azoospermia/genética , Azoospermia/metabolismo , Azoospermia/terapia , Sêmen/metabolismo , Espermatozoides/metabolismo , Testículo/metabolismo , Infertilidade Masculina/metabolismo , Estudos Retrospectivos , Proteínas de Ancoragem à Quinase A/metabolismo
16.
Mol Cell Proteomics ; 22(6): 100564, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37146716

RESUMO

Spermatogenesis defects are important for male infertility; however, the etiology and pathogenesis are still unknown. Herein, we identified two loss-of-function mutations of STK33 in seven individuals with non-obstructive azoospermia. Further functional studies of these frameshift and nonsense mutations revealed that Stk33-/KI male mice were sterile, and Stk33-/KI sperm were abnormal with defects in the mitochondrial sheath, fibrous sheath, outer dense fiber, and axoneme. Stk33KI/KI male mice were subfertile and had oligoasthenozoospermia. Differential phosphoproteomic analysis and in vitro kinase assay identified novel phosphorylation substrates of STK33, fibrous sheath components A-kinase anchoring protein 3 and A-kinase anchoring protein 4, whose expression levels decreased in testis after deletion of Stk33. STK33 regulated the phosphorylation of A-kinase anchoring protein 3/4, affected the assembly of fibrous sheath in the sperm, and played an essential role in spermiogenesis and male infertility.


Assuntos
Proteínas de Ancoragem à Quinase A , Infertilidade Masculina , Humanos , Masculino , Camundongos , Animais , Proteínas de Ancoragem à Quinase A/metabolismo , Sêmen/metabolismo , Espermatozoides/metabolismo , Espermatogênese/fisiologia , Cauda do Espermatozoide/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Flagelos/metabolismo
17.
Nucleic Acids Res ; 51(7): 3150-3165, 2023 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-36869674

RESUMO

DNA double-strand breaks (DSBs) are functionally linked to genomic instability in spermatocytes and to male infertility. The heavy metal cadmium (Cd) is known to induce DNA damage in spermatocytes by unknown mechanisms. Here, we showed that Cd ions impaired the canonical non-homologous end-joining (NHEJ) repair pathway, but not the homologous recombination (HR) repair pathway, through stimulation of Ser2056 and Thr2609 phosphorylation of DNA-PKcs at DSB sites. Hyper-phosphorylation of DNA-PKcs led to its premature dissociation from DNA ends and the Ku complex, preventing recruitment of processing enzymes and further ligation of DNA ends. Specifically, this cascade was initiated by the loss of PP5 phosphatase activity, which results from the dissociation of PP5 from its activating ions (Mn), that is antagonized by Cd ions through a competitive mechanism. In accordance, in a mouse model Cd-induced genomic instability and consequential male reproductive dysfunction were effectively reversed by a high dosage of Mn ions. Together, our findings corroborate a protein phosphorylation-mediated genomic instability pathway in spermatocytes that is triggered by exchange of heavy metal ions.


Assuntos
Cádmio , Instabilidade Genômica , Infertilidade Masculina , Espermatócitos , Animais , Humanos , Masculino , Camundongos , Cádmio/toxicidade , DNA/metabolismo , Reparo do DNA por Junção de Extremidades , Reparo do DNA , Instabilidade Genômica/efeitos dos fármacos , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Íons/metabolismo , Fosforilação , Reparo de DNA por Recombinação , Espermatócitos/efeitos dos fármacos
18.
PLoS Genet ; 18(6): e1010272, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35763544

RESUMO

Protamines are unique sperm-specific proteins that package and protect paternal chromatin until fertilization. A subset of mammalian species expresses two protamines (PRM1 and PRM2), while in others PRM1 is sufficient for sperm chromatin packaging. Alterations of the species-specific ratio between PRM1 and PRM2 are associated with infertility. Unlike PRM1, PRM2 is generated as a precursor protein consisting of a highly conserved N-terminal domain, termed cleaved PRM2 (cP2), which is consecutively trimmed off during chromatin condensation. The carboxyterminal part, called mature PRM2 (mP2), interacts with DNA and together with PRM1, mediates chromatin-hypercondensation. The removal of the cP2 domain is believed to be imperative for proper chromatin condensation, yet, the role of cP2 is not yet understood. We generated mice lacking the cP2 domain while the mP2 is still expressed. We show that the cP2 domain is indispensable for complete sperm chromatin protamination and male mouse fertility. cP2 deficient sperm show incomplete protamine incorporation and a severely altered protamine ratio, retention of transition proteins and aberrant retention of the testis specific histone variant H2A.L.2. During epididymal transit, cP2 deficient sperm seem to undergo ROS mediated degradation leading to complete DNA fragmentation. The cP2 domain therefore seems to be a key aspect in the complex crosstalk between histones, transition proteins and protamines during sperm chromatin condensation. Overall, we present the first step towards understanding the role of the cP2 domain in paternal chromatin packaging and open up avenues for further research.


Assuntos
Histonas , Infertilidade Masculina , Animais , Cromatina/genética , Cromatina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Masculino , Mamíferos/genética , Camundongos , Protaminas/genética , Protaminas/metabolismo , Sêmen/metabolismo , Espermatozoides/metabolismo
19.
Proc Natl Acad Sci U S A ; 119(40): e2207805119, 2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-36161911

RESUMO

Tmem95 encodes a sperm acrosomal membrane protein, whose knockout has a male-specific sterility phenotype in mice. Tmem95 knockout murine sperm can bind to, but do not fuse with, eggs. How TMEM95 plays a role in membrane fusion of sperm and eggs has remained elusive. Here, we utilize a sperm penetration assay as a model system to investigate the function of human TMEM95. We show that human TMEM95 binds to hamster egg membranes, providing evidence for a TMEM95 receptor on eggs. Using X-ray crystallography, we reveal an evolutionarily conserved, positively charged region of TMEM95 as a putative receptor-binding surface. Amino acid substitutions within this region of TMEM95 ablate egg-binding activity. We identify monoclonal antibodies against TMEM95 that reduce the number of human sperm fused with hamster eggs in sperm penetration assays. Strikingly, these antibodies do not block binding of sperm to eggs. Taken together, these results provide strong evidence for a specific, receptor-mediated interaction of sperm TMEM95 with eggs and suggest that this interaction may have a role in facilitating membrane fusion during fertilization.


Assuntos
Infertilidade Masculina , Fusão de Membrana , Proteínas de Membrana , Óvulo , Proteínas de Plasma Seminal , Interações Espermatozoide-Óvulo , Espermatozoides , Substituição de Aminoácidos , Animais , Anticorpos Monoclonais , Cricetinae , Humanos , Infertilidade Masculina/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Óvulo/metabolismo , Sêmen/metabolismo , Proteínas de Plasma Seminal/genética , Proteínas de Plasma Seminal/metabolismo , Espermatozoides/metabolismo
20.
Genomics ; 116(2): 110813, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38402914

RESUMO

Azoospermia and asthenospermia are common manifestations of male infertility, but it needs further studies to understand the intrinsic regulation mechanism. As a popular model organism, zebrafish is often used to assess reproductive complications. In this study, by analyzing miRNA transcriptome of the mature triploid zebrafish testis afflicted with spermatogenic dysfunctions, leading to the identification of 36 miRNAs that are differentially expressed in comparison with diploid, which are predicted to target 2737 genes. Subsequent functional annotation of these genes pinpointed two miRNAs might association with spermatogenesis. Inhibitory experiments showed that NC_007115.7.7_998413 inhibited conducts a substantial decline in sperm density, and conducted lower embryo fertilization rate than control. And putative target genes qRT-PCR evaluation showed that spata2 was significant down-regulate upon inhibited NC_007115.7.7_998413. In summary, this research positions newly identified miRNA NC_007115.7.998413 as a regulatory factor in male zebrafish reproductive development, enhancing our comprehension of the molecular regulated pathways involved in spermatogenesis.


Assuntos
Infertilidade Masculina , MicroRNAs , Humanos , Animais , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Peixe-Zebra/genética , Sêmen/metabolismo , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Testículo/metabolismo , Espermatogênese/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA