Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 19.901
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 180(2): 248-262.e21, 2020 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-31978344

RESUMO

The testis expresses the largest number of genes of any mammalian organ, a finding that has long puzzled molecular biologists. Our single-cell transcriptomic data of human and mouse spermatogenesis provide evidence that this widespread transcription maintains DNA sequence integrity in the male germline by correcting DNA damage through a mechanism we term transcriptional scanning. We find that genes expressed during spermatogenesis display lower mutation rates on the transcribed strand and have low diversity in the population. Moreover, this effect is fine-tuned by the level of gene expression during spermatogenesis. The unexpressed genes, which in our model do not benefit from transcriptional scanning, diverge faster over evolutionary timescales and are enriched for sensory and immune-defense functions. Collectively, we propose that transcriptional scanning shapes germline mutation signatures and modulates mutation rates in a gene-specific manner, maintaining DNA sequence integrity for the bulk of genes but allowing for faster evolution in a specific subset.


Assuntos
Expressão Gênica/genética , Mutação em Linhagem Germinativa/genética , Espermatogênese/genética , Adulto , Animais , Sequência de Bases/genética , Perfilação da Expressão Gênica/métodos , Células Germinativas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Taxa de Mutação , Testículo/metabolismo , Transcrição Gênica/genética , Transcriptoma/genética
2.
Cell ; 178(4): 901-918.e16, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31398343

RESUMO

Physiology and metabolism are often sexually dimorphic, but the underlying mechanisms remain incompletely understood. Here, we use the intestine of Drosophila melanogaster to investigate how gut-derived signals contribute to sex differences in whole-body physiology. We find that carbohydrate handling is male-biased in a specific portion of the intestine. In contrast to known sexual dimorphisms in invertebrates, the sex differences in intestinal carbohydrate metabolism are extrinsically controlled by the adjacent male gonad, which activates JAK-STAT signaling in enterocytes within this intestinal portion. Sex reversal experiments establish roles for this male-biased intestinal metabolic state in controlling food intake and sperm production through gut-derived citrate. Our work uncovers a male gonad-gut axis coupling diet and sperm production, revealing that metabolic communication across organs is physiologically important. The instructive role of citrate in inter-organ communication might be significant in more biological contexts than previously recognized.


Assuntos
Metabolismo dos Carboidratos/fisiologia , Drosophila melanogaster/metabolismo , Ingestão de Alimentos/fisiologia , Mucosa Intestinal/metabolismo , Caracteres Sexuais , Maturação do Esperma/fisiologia , Animais , Ácido Cítrico/metabolismo , Proteínas de Drosophila/metabolismo , Feminino , Expressão Gênica , Janus Quinases/metabolismo , Masculino , RNA-Seq , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Açúcares/metabolismo , Testículo/metabolismo
3.
Cell ; 168(5): 916-927.e12, 2017 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-28235201

RESUMO

Regulatory variation influencing gene expression is a key contributor to phenotypic diversity, both within and between species. Unfortunately, RNA degrades too rapidly to be recovered from fossil remains, limiting functional genomic insights about our extinct hominin relatives. Many Neanderthal sequences survive in modern humans due to ancient hybridization, providing an opportunity to assess their contributions to transcriptional variation and to test hypotheses about regulatory evolution. We developed a flexible Bayesian statistical approach to quantify allele-specific expression (ASE) in complex RNA-seq datasets. We identified widespread expression differences between Neanderthal and modern human alleles, indicating pervasive cis-regulatory impacts of introgression. Brain regions and testes exhibited significant downregulation of Neanderthal alleles relative to other tissues, consistent with natural selection influencing the tissue-specific regulatory landscape. Our study demonstrates that Neanderthal-inherited sequences are not silent remnants of ancient interbreeding but have measurable impacts on gene expression that contribute to variation in modern human phenotypes.


Assuntos
Evolução Molecular , Expressão Gênica , Homem de Neandertal/genética , Animais , Encéfalo/metabolismo , Regulação da Expressão Gênica , Humanos , Masculino , Especificidade de Órgãos , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Testículo/metabolismo
4.
Cell ; 171(7): 1559-1572.e20, 2017 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-29245011

RESUMO

Large-scale transcriptome sequencing efforts have vastly expanded the catalog of long non-coding RNAs (lncRNAs) with varying evolutionary conservation, lineage expression, and cancer specificity. Here, we functionally characterize a novel ultraconserved lncRNA, THOR (ENSG00000226856), which exhibits expression exclusively in testis and a broad range of human cancers. THOR knockdown and overexpression in multiple cell lines and animal models alters cell or tumor growth supporting an oncogenic role. We discovered a conserved interaction of THOR with IGF2BP1 and show that THOR contributes to the mRNA stabilization activities of IGF2BP1. Notably, transgenic THOR knockout produced fertilization defects in zebrafish and also conferred a resistance to melanoma onset. Likewise, ectopic expression of human THOR in zebrafish accelerated the onset of melanoma. THOR represents a novel class of functionally important cancer/testis lncRNAs whose structure and function have undergone positive evolutionary selection.


Assuntos
Modelos Animais de Doenças , Melanoma/metabolismo , RNA Longo não Codificante/metabolismo , Peixe-Zebra , Animais , Linhagem Celular Tumoral , Técnicas de Inativação de Genes , Humanos , Masculino , Camundongos , Proteínas de Ligação a RNA/metabolismo , Testículo/metabolismo
5.
Annu Rev Genet ; 56: 339-368, 2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-36070560

RESUMO

Spermatogenesis is a complex differentiation process coordinated spatiotemporally across and along seminiferous tubules. Cellular heterogeneity has made it challenging to obtain stage-specific molecular profiles of germ and somatic cells using bulk transcriptomic analyses. This has limited our ability to understand regulation of spermatogenesis and to integrate knowledge from model organisms to humans. The recent advancement of single-cell RNA-sequencing (scRNA-seq) technologies provides insights into the cell type diversity and molecular signatures in the testis. Fine-grained cell atlases of the testis contain both known and novel cell types and define the functional states along the germ cell developmental trajectory in many species. These atlases provide a reference system for integrated interspecies comparisons to discover mechanistic parallels and to enable future studies. Despite recent advances, we currently lack high-resolution data to probe germ cell-somatic cell interactions in the tissue environment, but the use of highly multiplexed spatial analysis technologies has begun to resolve this problem. Taken together, recent single-cell studies provide an improvedunderstanding of gametogenesis to examine underlying causes of infertility and enable the development of new therapeutic interventions.


Assuntos
Espermatogênese , Transcriptoma , Humanos , Masculino , Transcriptoma/genética , Espermatogênese/genética , Testículo/metabolismo , Perfilação da Expressão Gênica , Diferenciação Celular/genética
6.
Cell ; 163(4): 920-33, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26522592

RESUMO

A long-standing question concerns how stem cells maintain their identity through multiple divisions. Previously, we reported that pre-existing and newly synthesized histone H3 are asymmetrically distributed during Drosophila male germline stem cell (GSC) asymmetric division. Here, we show that phosphorylation at threonine 3 of H3 (H3T3P) distinguishes pre-existing versus newly synthesized H3. Converting T3 to the unphosphorylatable residue alanine (H3T3A) or to the phosphomimetic aspartate (H3T3D) disrupts asymmetric H3 inheritance. Expression of H3T3A or H3T3D specifically in early-stage germline also leads to cellular defects, including GSC loss and germline tumors. Finally, compromising the activity of the H3T3 kinase Haspin enhances the H3T3A but suppresses the H3T3D phenotypes. These studies demonstrate that H3T3P distinguishes sister chromatids enriched with distinct pools of H3 in order to coordinate asymmetric segregation of "old" H3 into GSCs and that tight regulation of H3T3 phosphorylation is required for male germline activity.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Histonas/metabolismo , Espermatogênese , Animais , Proteínas de Drosophila/química , Drosophila melanogaster/citologia , Células Germinativas/citologia , Células Germinativas/metabolismo , Histonas/química , Masculino , Mitose , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Testículo/metabolismo , Treonina/metabolismo
7.
Cell ; 160(4): 715-728, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25679763

RESUMO

AMP-activated protein kinase (AMPK) is a master sensor and regulator of cellular energy status. Upon metabolic stress, AMPK suppresses anabolic and promotes catabolic processes to regain energy homeostasis. Cancer cells can occasionally suppress the growth-restrictive AMPK pathway by mutation of an upstream regulatory kinase. Here, we describe a widespread mechanism to suppress AMPK through its ubiquitination and degradation by the cancer-specific MAGE-A3/6-TRIM28 ubiquitin ligase. MAGE-A3 and MAGE-A6 are highly similar proteins normally expressed only in the male germline but frequently re-activated in human cancers. MAGE-A3/6 are necessary for cancer cell viability and are sufficient to drive tumorigenic properties of non-cancerous cells. Screening for targets of MAGE-A3/6-TRIM28 revealed that it ubiquitinates and degrades AMPKα1. This leads to inhibition of autophagy, activation of mTOR signaling, and hypersensitization to AMPK agonists, such as metformin. These findings elucidate a germline mechanism commonly hijacked in cancer to suppress AMPK.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Antígenos de Neoplasias/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Sequência de Aminoácidos , Animais , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Metabolismo Energético , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Alinhamento de Sequência , Transdução de Sinais , Testículo/metabolismo
8.
Cell ; 163(5): 1252-1266, 2015 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-26548954

RESUMO

In meiosis, telomeres attach to the inner nuclear membrane (INM) and drive the chromosome movement required for homolog pairing and recombination. Here, we address the question of how telomeres are structurally adapted for the meiotic task. We identify a multi-subunit meiotic telomere-complex, TERB1/2-MAJIN, which takes over telomeric DNA from the shelterin complex in mouse germ cells. TERB1/2-MAJIN initially assembles on the INM sequestered by its putative transmembrane subunit MAJIN. In early meiosis, telomere attachment is achieved by the formation of a chimeric complex of TERB1/2-MAJIN and shelterin. The chimeric complex matures during prophase into DNA-bound TERB1/2-MAJIN by releasing shelterin, forming a direct link between telomeric DNA and the INM. These hierarchical processes, termed "telomere cap exchange," are regulated by CDK-dependent phosphorylation and the DNA-binding activity of MAJIN. Further, we uncover a positive feedback between telomere attachment and chromosome movement, revealing a comprehensive regulatory network underlying meiosis-specific telomere function in mammals.


Assuntos
Proteínas de Membrana/metabolismo , Membrana Nuclear/metabolismo , Proteínas de Ligação a Telômeros/metabolismo , Telômero/metabolismo , Sequência de Aminoácidos , Animais , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Humanos , Masculino , Meiose , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/genética , Dados de Sequência Molecular , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Alinhamento de Sequência , Proteínas de Ligação a Telômeros/química , Proteínas de Ligação a Telômeros/genética , Testículo/metabolismo
9.
Nature ; 629(8012): 652-659, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38693261

RESUMO

The gut microbiota operates at the interface of host-environment interactions to influence human homoeostasis and metabolic networks1-4. Environmental factors that unbalance gut microbial ecosystems can therefore shape physiological and disease-associated responses across somatic tissues5-9. However, the systemic impact of the gut microbiome on the germline-and consequently on the F1 offspring it gives rise to-is unexplored10. Here we show that the gut microbiota act as a key interface between paternal preconception environment and intergenerational health in mice. Perturbations to the gut microbiota of prospective fathers increase the probability of their offspring presenting with low birth weight, severe growth restriction and premature mortality. Transmission of disease risk occurs via the germline and is provoked by pervasive gut microbiome perturbations, including non-absorbable antibiotics or osmotic laxatives, but is rescued by restoring the paternal microbiota before conception. This effect is linked with a dynamic response to induced dysbiosis in the male reproductive system, including impaired leptin signalling, altered testicular metabolite profiles and remapped small RNA payloads in sperm. As a result, dysbiotic fathers trigger an elevated risk of in utero placental insufficiency, revealing a placental origin of mammalian intergenerational effects. Our study defines a regulatory 'gut-germline axis' in males, which is sensitive to environmental exposures and programmes offspring fitness through impacting placenta function.


Assuntos
Suscetibilidade a Doenças , Disbiose , Pai , Microbioma Gastrointestinal , Insuficiência Placentária , Lesões Pré-Natais , Espermatozoides , Animais , Feminino , Masculino , Camundongos , Gravidez , Disbiose/complicações , Disbiose/microbiologia , Microbioma Gastrointestinal/fisiologia , Leptina/metabolismo , Camundongos Endogâmicos C57BL , Placenta/metabolismo , Placenta/fisiopatologia , Insuficiência Placentária/etiologia , Insuficiência Placentária/metabolismo , Insuficiência Placentária/fisiopatologia , Resultado da Gravidez , Lesões Pré-Natais/etiologia , Lesões Pré-Natais/metabolismo , Lesões Pré-Natais/fisiopatologia , Transdução de Sinais , Espermatozoides/metabolismo , Testículo/metabolismo , Testículo/fisiopatologia , Suscetibilidade a Doenças/etiologia
10.
Mol Cell ; 81(23): 4826-4842.e8, 2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34626567

RESUMO

In animals, PIWI-interacting RNAs (piRNAs) silence transposons, fight viral infections, and regulate gene expression. piRNA biogenesis concludes with 3' terminal trimming and 2'-O-methylation. Both trimming and methylation influence piRNA stability. Our biochemical data show that multiple mechanisms destabilize unmethylated mouse piRNAs, depending on whether the piRNA 5' or 3' sequence is complementary to a trigger RNA. Unlike target-directed degradation of microRNAs, complementarity-dependent destabilization of piRNAs in mice and flies is blocked by 3' terminal 2'-O-methylation and does not require base pairing to both the piRNA seed and the 3' sequence. In flies, 2'-O-methylation also protects small interfering RNAs (siRNAs) from complementarity-dependent destruction. By contrast, pre-piRNA trimming protects mouse piRNAs from a degradation pathway unaffected by trigger complementarity. In testis lysate and in vivo, internal or 3' terminal uridine- or guanine-rich tracts accelerate pre-piRNA decay. Loss of both trimming and 2'-O-methylation causes the mouse piRNA pathway to collapse, demonstrating that these modifications collaborate to stabilize piRNAs.


Assuntos
Proteínas Argonautas/metabolismo , RNA Interferente Pequeno/metabolismo , Animais , Separação Celular , Drosophila melanogaster , Feminino , Citometria de Fluxo , Expressão Gênica , Inativação Gênica , Técnicas Genéticas , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Processamento de Proteína Pós-Traducional , RNA de Cadeia Dupla , Espermatócitos/metabolismo , Espermatogônias/metabolismo , Testículo/metabolismo
11.
Cell ; 153(5): 1012-24, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23706739

RESUMO

Histone acetylation plays critical roles in chromatin remodeling, DNA repair, and epigenetic regulation of gene expression, but the underlying mechanisms are unclear. Proteasomes usually catalyze ATP- and polyubiquitin-dependent proteolysis. Here, we show that the proteasomes containing the activator PA200 catalyze the polyubiquitin-independent degradation of histones. Most proteasomes in mammalian testes ("spermatoproteasomes") contain a spermatid/sperm-specific α subunit α4 s/PSMA8 and/or the catalytic ß subunits of immunoproteasomes in addition to PA200. Deletion of PA200 in mice abolishes acetylation-dependent degradation of somatic core histones during DNA double-strand breaks and delays core histone disappearance in elongated spermatids. Purified PA200 greatly promotes ATP-independent proteasomal degradation of the acetylated core histones, but not polyubiquitinated proteins. Furthermore, acetylation on histones is required for their binding to the bromodomain-like regions in PA200 and its yeast ortholog, Blm10. Thus, PA200/Blm10 specifically targets the core histones for acetylation-mediated degradation by proteasomes, providing mechanisms by which acetylation regulates histone degradation, DNA repair, and spermatogenesis.


Assuntos
Reparo do DNA , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Espermatogênese , Testículo/metabolismo , Acetilação , Sequência de Aminoácidos , Animais , Quebras de DNA de Cadeia Dupla , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Proteínas Nucleares/química , Estrutura Terciária de Proteína , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Alinhamento de Sequência
12.
Mol Cell ; 79(4): 645-659.e9, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32692974

RESUMO

Stress granules (SGs) are membrane-less ribonucleoprotein condensates that form in response to various stress stimuli via phase separation. SGs act as a protective mechanism to cope with acute stress, but persistent SGs have cytotoxic effects that are associated with several age-related diseases. Here, we demonstrate that the testis-specific protein, MAGE-B2, increases cellular stress tolerance by suppressing SG formation through translational inhibition of the key SG nucleator G3BP. MAGE-B2 reduces G3BP protein levels below the critical concentration for phase separation and suppresses SG initiation. Knockout of the MAGE-B2 mouse ortholog or overexpression of G3BP1 confers hypersensitivity of the male germline to heat stress in vivo. Thus, MAGE-B2 provides cytoprotection to maintain mammalian spermatogenesis, a highly thermosensitive process that must be preserved throughout reproductive life. These results demonstrate a mechanism that allows for tissue-specific resistance against stress and could aid in the development of male fertility therapies.


Assuntos
Grânulos Citoplasmáticos/genética , DNA Helicases/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética , Biossíntese de Proteínas , RNA Helicases/genética , Proteínas com Motivo de Reconhecimento de RNA/genética , Estresse Fisiológico/genética , Regiões 5' não Traduzidas , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Grânulos Citoplasmáticos/metabolismo , Grânulos Citoplasmáticos/patologia , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , DNA Helicases/metabolismo , Feminino , Células HCT116 , Células HeLa , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , RNA Helicases/metabolismo , Proteínas com Motivo de Reconhecimento de RNA/metabolismo , Espermatogônias/citologia , Espermatogônias/patologia , Testículo/citologia , Testículo/metabolismo
13.
Am J Hum Genet ; 111(9): 1953-1969, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39116879

RESUMO

While it is widely thought that de novo mutations (DNMs) occur randomly, we previously showed that some DNMs are enriched because they are positively selected in the testes of aging men. These "selfish" mutations cause disorders with a shared presentation of features, including exclusive paternal origin, significant increase of the father's age, and high apparent germline mutation rate. To date, all known selfish mutations cluster within the components of the RTK-RAS-MAPK signaling pathway, a critical modulator of testicular homeostasis. Here, we demonstrate the selfish nature of the SMAD4 DNMs causing Myhre syndrome (MYHRS). By analyzing 16 informative trios, we show that MYHRS-causing DNMs originated on the paternally derived allele in all cases. We document a statistically significant epidemiological paternal age effect of 6.3 years excess for fathers of MYHRS probands. We developed an ultra-sensitive assay to quantify spontaneous MYHRS-causing SMAD4 variants in sperm and show that pathogenic variants at codon 500 are found at elevated level in sperm of most men and exhibit a strong positive correlation with donor's age, indicative of a high apparent germline mutation rate. Finally, we performed in vitro assays to validate the peculiar functional behavior of the clonally selected DNMs and explored the basis of the pathophysiology of the different SMAD4 sperm-enriched variants. Taken together, these data provide compelling evidence that SMAD4, a gene operating outside the canonical RAS-MAPK signaling pathway, is associated with selfish spermatogonial selection and raises the possibility that other genes/pathways are under positive selection in the aging human testis.


Assuntos
Mutação em Linhagem Germinativa , Deficiência Intelectual , Proteína Smad4 , Humanos , Masculino , Proteína Smad4/genética , Deficiência Intelectual/genética , Contratura/genética , Adulto , Fácies , Espermatozoides/metabolismo , Espermatozoides/patologia , Criptorquidismo/genética , Transtornos do Crescimento/genética , Deformidades Congênitas da Mão/genética , Seleção Genética , Alelos , Idade Paterna , Testículo/patologia , Testículo/metabolismo
14.
Development ; 151(13)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38953252

RESUMO

Spermatogonial stem cell (SSC) self-renewal and differentiation provide foundational support for long-term, steady-state spermatogenesis in mammals. Here, we have investigated the essential role of RNA exosome associated DIS3 ribonuclease in maintaining spermatogonial homeostasis and facilitating germ cell differentiation. We have established male germ-cell Dis3 conditional knockout (cKO) mice in which the first and subsequent waves of spermatogenesis are disrupted. This leads to a Sertoli cell-only phenotype and sterility in adult male mice. Bulk RNA-seq documents that Dis3 deficiency partially abolishes RNA degradation and causes significant increases in the abundance of transcripts. This also includes pervasively transcribed PROMoter uPstream Transcripts (PROMPTs), which accumulate robustly in Dis3 cKO testes. In addition, scRNA-seq analysis indicates that Dis3 deficiency in spermatogonia significantly disrupts RNA metabolism and gene expression, and impairs early germline cell development. Overall, we document that exosome-associated DIS3 ribonuclease plays crucial roles in maintaining early male germ cell lineage in mice.


Assuntos
Fertilidade , Espermatogônias , Testículo , Animais , Masculino , Camundongos , Diferenciação Celular , Complexo Multienzimático de Ribonucleases do Exossomo/metabolismo , Complexo Multienzimático de Ribonucleases do Exossomo/genética , Exossomos/metabolismo , Fertilidade/genética , Infertilidade Masculina/genética , Camundongos Knockout , Estabilidade de RNA/genética , Células de Sertoli/metabolismo , Espermatogênese , Espermatogônias/metabolismo , Espermatogônias/citologia , Testículo/metabolismo
15.
Development ; 151(11)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38832826

RESUMO

Germline maintenance relies on adult stem cells to continually replenish lost gametes over a lifetime and respond to external cues altering the demands on the tissue. Mating worsens germline homeostasis over time, yet a negative impact on stem cell behavior has not been explored. Using extended live imaging of the Drosophila testis stem cell niche, we find that short periods of mating in young males disrupts cytokinesis in germline stem cells (GSCs). This defect leads to failure of abscission, preventing release of differentiating cells from the niche. We find that GSC abscission failure is caused by increased Ecdysone hormone signaling induced upon mating, which leads to disrupted somatic encystment of the germline. Abscission failure is rescued by isolating males from females, but recurs with resumption of mating. Importantly, reiterative mating also leads to increased GSC loss, requiring increased restoration of stem cells via symmetric renewal and de-differentiation. Together, these results suggest a model whereby acute mating results in hormonal changes that negatively impact GSC cytokinesis but preserves the stem cell population.


Assuntos
Citocinese , Drosophila melanogaster , Ecdisona , Células Germinativas , Testículo , Animais , Masculino , Ecdisona/metabolismo , Testículo/metabolismo , Feminino , Drosophila melanogaster/metabolismo , Células Germinativas/metabolismo , Células Germinativas/citologia , Nicho de Células-Tronco , Células-Tronco/metabolismo , Células-Tronco/citologia , Diferenciação Celular , Transdução de Sinais , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética
16.
Development ; 151(18)2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39222051

RESUMO

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.


Assuntos
Homeostase , Espermátides , Espermatogênese , Masculino , Animais , Espermatogênese/genética , Camundongos , Espermátides/metabolismo , Testículo/metabolismo , Histonas/metabolismo , Peptídeo Hidrolases/metabolismo , Peptídeo Hidrolases/genética , Epigênese Genética , Infertilidade Masculina/genética , Camundongos Endogâmicos C57BL , Meiose/genética , Células-Tronco Germinativas Adultas/metabolismo , Camundongos Knockout , Imunidade Inata/genética , Espermatogônias/metabolismo
17.
Development ; 151(12)2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38934417

RESUMO

Spermatogonial stem cells (SSCs) undergo self-renewal division to sustain spermatogenesis. Although it is possible to derive SSC cultures in most mouse strains, SSCs from a 129 background never proliferate under the same culture conditions, suggesting they have distinct self-renewal requirements. Here, we established long-term culture conditions for SSCs from mice of the 129 background (129 mice). An analysis of 129 testes showed significant reduction of GDNF and CXCL12, whereas FGF2, INHBA and INHBB were higher than in testes of C57BL/6 mice. An analysis of undifferentiated spermatogonia in 129 mice showed higher expression of Chrna4, which encodes an acetylcholine (Ach) receptor component. By supplementing medium with INHBA and Ach, SSC cultures were derived from 129 mice. Following lentivirus transduction for marking donor cells, transplanted cells re-initiated spermatogenesis in infertile mouse testes and produced transgenic offspring. These results suggest that the requirements of SSC self-renewal in mice are diverse, which has important implications for understanding self-renewal mechanisms in various animal species.


Assuntos
Camundongos Endogâmicos C57BL , Espermatogênese , Espermatogônias , Testículo , Animais , Masculino , Camundongos , Espermatogônias/citologia , Espermatogônias/metabolismo , Espermatogênese/genética , Espermatogênese/fisiologia , Testículo/metabolismo , Testículo/citologia , Autorrenovação Celular , Células-Tronco Germinativas Adultas/metabolismo , Células-Tronco Germinativas Adultas/citologia , Células Cultivadas , Receptores Nicotínicos/metabolismo , Receptores Nicotínicos/genética , Camundongos Endogâmicos , Diferenciação Celular , Proliferação de Células , Células-Tronco/citologia , Células-Tronco/metabolismo , Camundongos Transgênicos
18.
Nat Methods ; 21(7): 1231-1244, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38844627

RESUMO

Spatially resolved transcriptomics (SRT) studies are becoming increasingly common and large, offering unprecedented opportunities in mapping complex tissue structures and functions. Here we present integrative and reference-informed tissue segmentation (IRIS), a computational method designed to characterize tissue spatial organization in SRT studies through accurately and efficiently detecting spatial domains. IRIS uniquely leverages single-cell RNA sequencing data for reference-informed detection of biologically interpretable spatial domains, integrating multiple SRT slices while explicitly considering correlations both within and across slices. We demonstrate the advantages of IRIS through in-depth analysis of six SRT datasets encompassing diverse technologies, tissues, species and resolutions. In these applications, IRIS achieves substantial accuracy gains (39-1,083%) and speed improvements (4.6-666.0) in moderate-sized datasets, while representing the only method applicable for large datasets including Stereo-seq and 10x Xenium. As a result, IRIS reveals intricate brain structures, uncovers tumor microenvironment heterogeneity and detects structural changes in diabetes-affected testis, all with exceptional speed and accuracy.


Assuntos
Análise de Célula Única , Transcriptoma , Humanos , Animais , Análise de Célula Única/métodos , Perfilação da Expressão Gênica/métodos , Camundongos , Masculino , Biologia Computacional/métodos , Encéfalo/metabolismo , Análise de Sequência de RNA/métodos , Testículo/metabolismo
19.
PLoS Biol ; 22(4): e3002605, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38687805

RESUMO

Although sex chromosomes have evolved from autosomes, they often have unusual regulatory regimes that are sex- and cell-type-specific such as dosage compensation (DC) and meiotic sex chromosome inactivation (MSCI). The molecular mechanisms and evolutionary forces driving these unique transcriptional programs are critical for genome evolution but have been, in the case of MSCI in Drosophila, subject to continuous debate. Here, we take advantage of the younger sex chromosomes in D. miranda (XR and the neo-X) to infer how former autosomes acquire sex-chromosome-specific regulatory programs using single-cell and bulk RNA sequencing and ribosome profiling, in a comparative evolutionary context. We show that contrary to mammals and worms, the X down-regulation through germline progression is most consistent with the shutdown of DC instead of MSCI, resulting in half gene dosage at the end of meiosis for all 3 X's. Moreover, lowly expressed germline and meiotic genes on the neo-X are ancestrally lowly expressed, instead of acquired suppression after sex linkage. For the young neo-X, DC is incomplete across all tissue and cell types and this dosage imbalance is rescued by contributions from Y-linked gametologs which produce transcripts that are translated to compensate both gene and protein dosage. We find an excess of previously autosomal testis genes becoming Y-specific, showing that the neo-Y and its masculinization likely resolve sexual antagonism. Multicopy neo-sex genes are predominantly expressed during meiotic stages of spermatogenesis, consistent with their amplification being driven to interfere with mendelian segregation. Altogether, this study reveals germline regulation of evolving sex chromosomes and elucidates the consequences these unique regulatory mechanisms have on the evolution of sex chromosome architecture.


Assuntos
Drosophila , Células Germinativas , Meiose , RNA-Seq , Cromossomos Sexuais , Análise de Célula Única , Testículo , Animais , Masculino , Testículo/metabolismo , Cromossomos Sexuais/genética , Análise de Célula Única/métodos , Células Germinativas/metabolismo , Drosophila/genética , Drosophila/metabolismo , RNA-Seq/métodos , Meiose/genética , Mecanismo Genético de Compensação de Dose , Evolução Molecular , Feminino , Cromossomo X/genética , Análise da Expressão Gênica de Célula Única
20.
Cell ; 148(3): 596-607, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22304924

RESUMO

Sex hormones such as estrogen and testosterone are essential for sexually dimorphic behaviors in vertebrates. However, the hormone-activated molecular mechanisms that control the development and function of the underlying neural circuits remain poorly defined. We have identified numerous sexually dimorphic gene expression patterns in the adult mouse hypothalamus and amygdala. We find that adult sex hormones regulate these expression patterns in a sex-specific, regionally restricted manner, suggesting that these genes regulate sex typical behaviors. Indeed, we find that mice with targeted disruptions of each of four of these genes (Brs3, Cckar, Irs4, Sytl4) exhibit extremely specific deficits in sex specific behaviors, with single genes controlling the pattern or extent of male sexual behavior, male aggression, maternal behavior, or female sexual behavior. Taken together, our findings demonstrate that various components of sexually dimorphic behaviors are governed by separable genetic programs.


Assuntos
Tonsila do Cerebelo/metabolismo , Perfilação da Expressão Gênica , Hipotálamo/metabolismo , Caracteres Sexuais , Comportamento Sexual Animal , Agressão , Animais , Estro/metabolismo , Feminino , Masculino , Comportamento Materno , Camundongos , Ovário/metabolismo , Testículo/metabolismo , Testosterona/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA