Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 943
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 176(6): 1379-1392.e14, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30773315

RESUMEN

Cell fate specification by lateral inhibition typically involves contact signaling through the Delta-Notch signaling pathway. However, whether this is the only signaling mode mediating lateral inhibition remains unclear. Here we show that in zebrafish oogenesis, a group of cells within the granulosa cell layer at the oocyte animal pole acquire elevated levels of the transcriptional coactivator TAZ in their nuclei. One of these cells, the future micropyle precursor cell (MPC), accumulates increasingly high levels of nuclear TAZ and grows faster than its surrounding cells, mechanically compressing those cells, which ultimately lose TAZ from their nuclei. Strikingly, relieving neighbor-cell compression by MPC ablation or aspiration restores nuclear TAZ accumulation in neighboring cells, eventually leading to MPC re-specification from these cells. Conversely, MPC specification is defective in taz-/- follicles. These findings uncover a novel mode of lateral inhibition in cell fate specification based on mechanical signals controlling TAZ activity.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Oogénesis/fisiología , Proteínas de Pez Cebra/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Comunicación Celular/fisiología , Diferenciación Celular/fisiología , Linaje de la Célula , Núcleo Celular/metabolismo , Femenino , Células de la Granulosa/metabolismo , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Oocitos/metabolismo , Oocitos/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Activación Transcripcional/fisiología , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Pez Cebra/metabolismo , Proteínas de Pez Cebra/antagonistas & inhibidores
2.
Cell ; 179(3): 671-686.e17, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31626769

RESUMEN

The molecular events that direct nuclear pore complex (NPC) assembly toward nuclear envelopes have been conceptualized in two pathways that occur during mitosis or interphase, respectively. In gametes and embryonic cells, NPCs also occur within stacked cytoplasmic membrane sheets, termed annulate lamellae (AL), which serve as NPC storage for early development. The mechanism of NPC biogenesis at cytoplasmic membranes remains unknown. Here, we show that during Drosophila oogenesis, Nucleoporins condense into different precursor granules that interact and progress into NPCs. Nup358 is a key player that condenses into NPC assembly platforms while its mRNA localizes to their surface in a translation-dependent manner. In concert, Microtubule-dependent transport, the small GTPase Ran and nuclear transport receptors regulate NPC biogenesis in oocytes. We delineate a non-canonical NPC assembly mechanism that relies on Nucleoporin condensates and occurs away from the nucleus under conditions of cell cycle arrest.


Asunto(s)
Proteínas de Drosophila/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Poro Nuclear/metabolismo , Oogénesis , Transporte Activo de Núcleo Celular , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Microtúbulos/metabolismo , Chaperonas Moleculares/genética , Proteínas de Complejo Poro Nuclear/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína de Unión al GTP ran/genética , Proteína de Unión al GTP ran/metabolismo
3.
Cell ; 174(5): 1082-1094.e12, 2018 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-30057117

RESUMEN

Although animals have evolved multiple mechanisms to suppress transposons, "leaky" mobilizations that cause mutations and diseases still occur. This suggests that transposons employ specific tactics to accomplish robust propagation. By directly tracking mobilization, we show that, during a short and specific time window of oogenesis, retrotransposons achieve massive amplification via a cell-type-specific targeting strategy. Retrotransposons rarely mobilize in undifferentiated germline stem cells. However, as oogenesis proceeds, they utilize supporting nurse cells-which are highly polyploid and eventually undergo apoptosis-as factories to massively manufacture invading products. Moreover, retrotransposons rarely integrate into nurse cells themselves but, instead, via microtubule-mediated transport, they preferentially target the DNA of the interconnected oocytes. Blocking microtubule-dependent intercellular transport from nurse cells significantly alleviates damage to the oocyte genome. Our data reveal that parasitic genomic elements can efficiently hijack a host developmental process to propagate robustly, thereby driving evolutionary change and causing disease.


Asunto(s)
Drosophila melanogaster/genética , Elementos de Nucleótido Esparcido Largo , Oogénesis , ARN Interferente Pequeño , Retroelementos , Retroviridae/genética , Animales , Proteínas de Drosophila , Femenino , Biblioteca de Genes , Silenciador del Gen , Células Germinativas , Proteínas Fluorescentes Verdes/metabolismo , Hibridación Fluorescente in Situ , Masculino , Oocitos/metabolismo , Células Madre/metabolismo
4.
Cell ; 169(3): 457-469.e13, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28431246

RESUMEN

Fat metabolism has been linked to fertility and reproductive adaptation in animals and humans, and environmental sex determination potentially plays a role in the process. To investigate the impact of fatty acids (FA) on sex determination and reproductive development, we examined and observed an impact of FA synthesis and mobilization by lipolysis in somatic tissues on oocyte fate in Caenorhabditis elegans. The subsequent genetic analysis identified ACS-4, an acyl-CoA synthetase and its FA-CoA product, as key germline factors that mediate the role of FA in promoting oocyte fate through protein myristoylation. Further tests indicated that ACS-4-dependent protein myristoylation perceives and translates the FA level into regulatory cues that modulate the activities of MPK-1/MAPK and key factors in the germline sex-determination pathway. These findings, including a similar role of ACS-4 in a male/female species, uncover a likely conserved mechanism by which FA, an environmental factor, regulates sex determination and reproductive development.


Asunto(s)
Acetato CoA Ligasa/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiología , Ácidos Grasos/metabolismo , Ácido Mirístico/metabolismo , Procesamiento Proteico-Postraduccional , Procesos de Determinación del Sexo , Acetato CoA Ligasa/genética , Animales , Proteínas de Caenorhabditis elegans/genética , Mutación , Oocitos/metabolismo
5.
Cell ; 165(2): 396-409, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27020753

RESUMEN

Multiple division cycles without growth are a characteristic feature of early embryogenesis. The female germline loads proteins and RNAs into oocytes to support these divisions, which lack many quality control mechanisms operating in somatic cells undergoing growth. Here, we describe a small RNA-Argonaute pathway that ensures early embryonic divisions in C. elegans by employing catalytic slicing activity to broadly tune, instead of silence, germline gene expression. Misregulation of one target, a kinesin-13 microtubule depolymerase, underlies a major phenotype associated with pathway loss. Tuning of target transcript levels is guided by the density of homologous small RNAs, whose generation must ultimately be related to target sequence. Thus, the tuning action of a small RNA-catalytic Argonaute pathway generates oocytes capable of supporting embryogenesis. We speculate that the specialized nature of germline chromatin led to the emergence of small RNA-catalytic Argonaute pathways in the female germline as a post-transcriptional control layer to optimize oocyte composition.


Asunto(s)
Caenorhabditis elegans/embriología , Caenorhabditis elegans/metabolismo , Embrión no Mamífero/metabolismo , Redes y Vías Metabólicas , Oocitos/metabolismo , Animales , Proteínas Argonautas/metabolismo , Secuencia de Bases , Caenorhabditis elegans/citología , Proteínas de Caenorhabditis elegans/metabolismo , División Celular , Embrión no Mamífero/citología , Desarrollo Embrionario , Femenino , Cinesinas/metabolismo , Microtúbulos/metabolismo , Datos de Secuencia Molecular , Procesamiento Postranscripcional del ARN
6.
Mol Cell ; 82(9): 1678-1690.e12, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35305312

RESUMEN

The functional consequence of N6-methyladenosine (m6A) RNA modification is mediated by "reader" proteins of the YTH family. YTH domain-containing 2 (YTHDC2) is essential for mammalian fertility, but its molecular function is poorly understood. Here, we identify U-rich motifs as binding sites of YTHDC2 on 3' UTRs of mouse testicular RNA targets. Although its YTH domain is an m6A-binder in vitro, the YTH point mutant mice are fertile. Significantly, the loss of its 3'→5' RNA helicase activity causes mouse infertility, with the catalytic-dead mutation being dominant negative. Biochemical studies reveal that the weak helicase activity of YTHDC2 is enhanced by its interaction with the 5'→3' exoribonuclease XRN1. Single-cell transcriptomics indicate that Ythdc2 mutant mitotic germ cells transition into meiosis but accumulate a transcriptome with mixed mitotic/meiotic identity that fail to progress further into meiosis. Finally, our demonstration that ythdc2 mutant zebrafish are infertile highlights its conserved role in animal germ cell development.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Exorribonucleasas/metabolismo , ARN Helicasas , Pez Cebra , Animales , Fertilidad/genética , Mamíferos/metabolismo , Meiosis , Ratones , ARN/genética , ARN Helicasas/genética , ARN Helicasas/metabolismo , Pez Cebra/genética
7.
Annu Rev Cell Dev Biol ; 31: 291-315, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26355592

RESUMEN

Stem cells are necessary for the maintenance of many adult tissues. Signals within the stem cell microenvironment, or niche, regulate the self-renewal and differentiation capability of these cells. Misregulation of these signals through mutation or damage can lead to overgrowth or depletion of different stem cell pools. In this review, we focus on the Drosophila testis and ovary, both of which contain well-defined niches, as well as the mouse testis, which has become a more approachable stem cell system with recent technical advances. We discuss the signals that regulate gonadal stem cells in their niches, how these signals mediate self-renewal and differentiation under homeostatic conditions, and how stress, whether from mutations or damage, can cause changes in cell fate and drive stem cell competition.


Asunto(s)
Autorrenovación de las Células/genética , Autorrenovación de las Células/fisiología , Gónadas/fisiología , Células Madre/fisiología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Drosophila/genética , Drosophila/fisiología , Femenino , Humanos , Masculino , Transducción de Señal/genética , Transducción de Señal/fisiología , Nicho de Células Madre/genética , Nicho de Células Madre/fisiología
8.
Genes Dev ; 35(5-6): 392-409, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33574069

RESUMEN

Nuclear Argonaute proteins, guided by their bound small RNAs to nascent target transcripts, mediate cotranscriptional silencing of transposons and repetitive genomic loci through heterochromatin formation. The molecular mechanisms involved in this process are incompletely understood. Here, we show that the SFiNX complex, a silencing mediator downstream from nuclear Piwi-piRNA complexes in Drosophila, facilitates cotranscriptional silencing as a homodimer. The dynein light chain protein Cut up/LC8 mediates SFiNX dimerization, and its function can be bypassed by a heterologous dimerization domain, arguing for a constitutive SFiNX dimer. Dimeric, but not monomeric SFiNX, is capable of forming molecular condensates in a nucleic acid-stimulated manner. Mutations that prevent SFiNX dimerization result in loss of condensate formation in vitro and the inability of Piwi to initiate heterochromatin formation and silence transposons in vivo. We propose that multivalent SFiNX-nucleic acid interactions are critical for heterochromatin establishment at piRNA target loci in a cotranscriptional manner.


Asunto(s)
Proteínas Argonautas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica/genética , Silenciador del Gen/fisiología , Complejos Multiproteicos/metabolismo , Animales , Dimerización , Proteínas de Drosophila/química , Drosophila melanogaster/metabolismo , Dineínas/metabolismo , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Transporte Nucleocitoplasmático/química , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
9.
EMBO J ; 42(24): e115076, 2023 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-37987153

RESUMEN

In most metazoans, centrioles are lost during oogenesis, ensuring that the zygote is endowed with the correct number of two centrioles, which are paternally contributed. How centriole architecture is dismantled during oogenesis is not understood. Here, we analyze with unprecedent detail the ultrastructural and molecular changes during oogenesis centriole elimination in Caenorhabditis elegans. Centriole elimination begins with loss of the so-called central tube and organelle widening, followed by microtubule disassembly. The resulting cluster of centriolar proteins then disappears gradually, usually moving in a microtubule- and dynein-dependent manner to the plasma membrane. Our analysis indicates that neither Polo-like kinases nor the PCM, which modulate oogenesis centriole elimination in Drosophila, do so in C. elegans. Furthermore, we demonstrate that the central tube protein SAS-1 normally departs initially from the organelle, which loses integrity earlier in sas-1 mutants. Overall, our work provides novel mechanistic insights regarding the fundamental process of oogenesis centriole elimination.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Centriolos/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Microtúbulos/metabolismo , Drosophila/metabolismo , Oogénesis , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
10.
EMBO J ; 42(9): e112962, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-36929479

RESUMEN

Human in vitro oogenesis provides a framework for clarifying the mechanism of human oogenesis. To create its benchmark, it is vital to promote in vitro oogenesis using a model physiologically close to humans. Here, we establish a foundation for in vitro oogenesis in cynomolgus (cy) monkeys (Macaca fascicularis): cy female embryonic stem cells harboring one active and one inactive X chromosome (Xa and Xi, respectively) differentiate robustly into primordial germ cell-like cells, which in xenogeneic reconstituted ovaries develop efficiently into oogonia and, remarkably, further into meiotic oocytes at the zygotene stage. This differentiation entails comprehensive epigenetic reprogramming, including Xi reprogramming, yet Xa and Xi remain epigenetically asymmetric with, as partly observed in vivo, incomplete Xi reactivation. In humans and monkeys, the Xi epigenome in pluripotent stem cells functions as an Xi-reprogramming determinant. We further show that developmental pathway over-activations with suboptimal up-regulation of relevant meiotic genes impede in vitro meiotic progression. Cy in vitro oogenesis exhibits critical homology with the human system, including with respect to bottlenecks, providing a salient model for advancing human in vitro oogenesis.


Asunto(s)
Oocitos , Oogénesis , Animales , Femenino , Humanos , Macaca fascicularis , Oogénesis/fisiología , Ovario , Células Madre Embrionarias
11.
Development ; 151(16)2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39190553

RESUMEN

The size of subcellular structures must be tightly controlled to maintain normal cell function. Despite its importance, few studies have determined how the size of organelles or other structures is maintained during development, when cells are growing, dividing and rearranging. The developing Drosophila egg chamber is a powerful model in which to study the relative growth rates of subcellular structures. The egg chamber contains a cluster of 16 germline cells, which are connected through intercellular bridges called ring canals. As the egg chamber grows, the germline cells and the ring canals that connect them increase in size. Here, we demonstrate that ring canal size scaling is related to lineage; the largest, 'first-born' ring canals increase in size at a relatively slower rate than ring canals derived from subsequent mitotic divisions. This lineage-based scaling relationship is maintained even if directed transport is reduced, ring canal size is altered, or in egg chambers with twice as many germline cells. Analysis of lines that produce larger or smaller mature eggs reveals that different strategies could be used to alter final egg size.


Asunto(s)
Linaje de la Célula , Células Germinativas , Oogénesis , Animales , Oogénesis/fisiología , Femenino , Células Germinativas/citología , Drosophila melanogaster , Drosophila , Óvulo/citología , Tamaño de la Célula
12.
Development ; 151(5)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38471539

RESUMEN

Gametogenesis is the process through which germ cells differentiate into sexually dimorphic gametes, eggs and sperm. In the teleost fish medaka (Oryzias latipes), a germ cell-intrinsic sex determinant, foxl3, triggers germline feminization by activating two genetic pathways that regulate folliculogenesis and meiosis. Here, we identified a pathway involving a dome-shaped microtubule structure that may be the basis of oocyte polarity. This structure was first established in primordial germ cells in both sexes, but was maintained only during oogenesis and was destabilized in differentiating spermatogonia under the influence of Sertoli cells expressing dmrt1. Although foxl3 was dispensable for this pathway, dazl was involved in the persistence of the microtubule dome at the time of gonocyte development. In addition, disruption of the microtubule dome caused dispersal of bucky ball RNA, suggesting the structure may be prerequisite for the Balbiani body. Collectively, the present findings provide mechanistic insight into the establishment of sex-specific polarity through the formation of a microtubule structure in germ cells, as well as clarifying the genetic pathways implementing oocyte-specific characteristics.


Asunto(s)
Oryzias , Animales , Femenino , Masculino , Oryzias/genética , Semen , Células Germinativas/metabolismo , Gametogénesis , Oogénesis/fisiología
13.
Development ; 151(14)2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38881530

RESUMEN

HemK2 is a highly conserved methyltransferase, but the identification of its genuine substrates has been controversial, and its biological importance in higher organisms remains unclear. We elucidate the role of HemK2 in the methylation of eukaryotic Release Factor 1 (eRF1), a process that is essential for female germline development in Drosophila melanogaster. Knockdown of hemK2 in the germline cells (hemK2-GLKD) induces apoptosis, accompanied by a pronounced decrease in both eRF1 methylation and protein synthesis. Overexpression of a methylation-deficient eRF1 variant recapitulates the defects observed in hemK2-GLKD, suggesting that eRF1 is a primary methylation target of HemK2. Furthermore, hemK2-GLKD leads to a significant reduction in mRNA levels in germline cell. These defects in oogenesis and protein synthesis can be partially restored by inhibiting the No-Go Decay pathway. In addition, hemK2 knockdown is associated with increased disome formation, suggesting that disruptions in eRF1 methylation may provoke ribosomal stalling, which subsequently activates translation-coupled mRNA surveillance mechanisms that degrade actively translated mRNAs. We propose that HemK2-mediated methylation of eRF1 is crucial for ensuring efficient protein production and mRNA stability, which are vital for the generation of high-quality eggs.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Oogénesis , Biosíntesis de Proteínas , Estabilidad del ARN , Animales , Oogénesis/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Estabilidad del ARN/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Femenino , Metilación , Metiltransferasas/metabolismo , Metiltransferasas/genética , Factores de Terminación de Péptidos/metabolismo , Factores de Terminación de Péptidos/genética , ARN Mensajero/metabolismo , ARN Mensajero/genética , Apoptosis/genética , ARN Helicasas DEAD-box
14.
Annu Rev Genet ; 53: 1-18, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31794267

RESUMEN

In Drosophila development, the axes of the egg and future embryo are established during oogenesis. To learn about the underlying genetic and molecular pathways that lead to axis formation, I conducted a large-scale genetic screen at the beginning of my independent career. This led to the eventual understanding that both anterior-posterior and dorsal-ventral pattern information is transmitted from the oocyte to the surrounding follicle cells and in turn from the follicle cells back to the oocyte. How I came to conduct this screen and what further insights were gained by studying the mutants isolated in the screen are the topics of this autobiographical article.


Asunto(s)
Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Genética/historia , Óvulo/fisiología , Animales , Tipificación del Cuerpo/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Embrión no Mamífero , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Historia del Siglo XX , Historia del Siglo XXI , Masculino , Oocitos/fisiología , Ovario/crecimiento & desarrollo , Ovario/fisiología , Receptores de Péptidos de Invertebrados/genética , Receptores de Péptidos de Invertebrados/metabolismo , Análisis para Determinación del Sexo , Procesos de Determinación del Sexo , Estados Unidos
15.
Mol Cell ; 73(6): 1127-1137.e5, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30772175

RESUMEN

We have previously proposed that selective inheritance, the limited transmission of damaging mtDNA mutations from mother to offspring, is based on replication competition in Drosophila melanogaster. This model, which stems from our observation that wild-type mitochondria propagate much more vigorously in the fly ovary than mitochondria carrying fitness-impairing mutations, implies that germ cells recognize the fitness of individual mitochondria and selectively boost the propagation of healthy ones. Here, we demonstrate that the protein kinase PINK1 preferentially accumulates on mitochondria enriched for a deleterious mtDNA mutation. PINK1 phosphorylates Larp to inhibit protein synthesis on the mitochondrial outer membrane. Impaired local translation on defective mitochondria in turn limits the replication of their mtDNA and hence the transmission of deleterious mutations to the offspring. Our work confirms that selective inheritance occurs at the organelle level during Drosophila oogenesis and provides molecular entry points to test this model in other systems.


Asunto(s)
Replicación del ADN , ADN Mitocondrial/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Mitocondrias/enzimología , Membranas Mitocondriales/enzimología , Proteínas Mitocondriales/biosíntesis , Mutación , Oocitos/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Animales Modificados Genéticamente , ADN Mitocondrial/biosíntesis , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Femenino , Patrón de Herencia , Mitocondrias/genética , Proteínas Mitocondriales/genética , Oogénesis , Biogénesis de Organelos , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Estabilidad Proteica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Mol Cell ; 74(5): 1053-1068.e8, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31003867

RESUMEN

Double-strand breaks (DSBs) initiate the homologous recombination that is crucial for meiotic chromosome pairing and segregation. Here, we unveil mouse ANKRD31 as a lynchpin governing multiple aspects of DSB formation. Spermatocytes lacking ANKRD31 have altered DSB locations and fail to target DSBs to the pseudoautosomal regions (PARs) of sex chromosomes. They also have delayed and/or fewer recombination sites but, paradoxically, more DSBs, suggesting DSB dysregulation. Unrepaired DSBs and pairing failures-stochastic on autosomes, nearly absolute on X and Y-cause meiotic arrest and sterility in males. Ankrd31-deficient females have reduced oocyte reserves. A crystal structure defines a pleckstrin homology (PH) domain in REC114 and its direct intermolecular contacts with ANKRD31. In vivo, ANKRD31 stabilizes REC114 association with the PAR and elsewhere. Our findings inform a model in which ANKRD31 is a scaffold anchoring REC114 and other factors to specific genomic locations, thereby regulating DSB formation.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Recombinación Homóloga/genética , Meiosis/genética , Recombinasas/química , Animales , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Emparejamiento Cromosómico , Segregación Cromosómica/genética , Cromosomas , Cristalografía por Rayos X , Roturas del ADN de Doble Cadena , Femenino , Masculino , Ratones , Conformación Proteica , Recombinasas/genética , Espermatocitos/química , Espermatocitos/metabolismo
17.
Hum Mol Genet ; 33(16): 1391-1405, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710511

RESUMEN

Fragile X syndrome (FXS) is an inherited neurodevelopmental disorder and the leading genetic cause of autism spectrum disorders. FXS is caused by loss of function mutations in Fragile X mental retardation protein (FMRP), an RNA binding protein that is known to regulate translation of its target mRNAs, predominantly in the brain and gonads. The molecular mechanisms connecting FMRP function to neurodevelopmental phenotypes are well understood. However, neither the full extent of reproductive phenotypes, nor the underlying molecular mechanisms have been as yet determined. Here, we developed new fmr1 knockout zebrafish lines and show that they mimic key aspects of FXS neuronal phenotypes across both larval and adult stages. Results from the fmr1 knockout females also showed that altered gene expression in the brain, via the neuroendocrine pathway contribute to distinct abnormal phenotypes during ovarian development and oocyte maturation. We identified at least three mechanisms underpinning these defects, including altered neuroendocrine signaling in sexually mature females resulting in accelerated ovarian development, altered expression of germ cell and meiosis promoting genes at various stages during oocyte maturation, and finally a strong mitochondrial impairment in late stage oocytes from knockout females. Our findings have implications beyond FXS in the study of reproductive function and female infertility. Dissection of the translation control pathways during ovarian development using models like the knockout lines reported here may reveal novel approaches and targets for fertility treatments.


Asunto(s)
Modelos Animales de Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Síndrome del Cromosoma X Frágil , Ovario , Proteínas de Pez Cebra , Pez Cebra , Animales , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/patología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Femenino , Ovario/metabolismo , Ovario/crecimiento & desarrollo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Oocitos/metabolismo , Oocitos/crecimiento & desarrollo , Encéfalo/metabolismo , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Fenotipo , Humanos , Proteínas de Unión al ARN
18.
Development ; 150(20)2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37306387

RESUMEN

Lipid droplets (LDs), crucial regulators of lipid metabolism, accumulate during oocyte development. However, their roles in fertility remain largely unknown. During Drosophila oogenesis, LD accumulation coincides with the actin remodeling necessary for follicle development. Loss of the LD-associated Adipose Triglyceride Lipase (ATGL) disrupts both actin bundle formation and cortical actin integrity, an unusual phenotype also seen when the prostaglandin (PG) synthase Pxt is missing. Dominant genetic interactions and PG treatment of follicles indicate that ATGL acts upstream of Pxt to regulate actin remodeling. Our data suggest that ATGL releases arachidonic acid (AA) from LDs to serve as the substrate for PG synthesis. Lipidomic analysis detects AA-containing triglycerides in ovaries, and these are increased when ATGL is lost. High levels of exogenous AA block follicle development; this is enhanced by impairing LD formation and suppressed by reducing ATGL. Together, these data support the model that AA stored in LD triglycerides is released by ATGL to drive the production of PGs, which promote the actin remodeling necessary for follicle development. We speculate that this pathway is conserved across organisms to regulate oocyte development and promote fertility.


Asunto(s)
Proteínas de Drosophila , Prostaglandinas , Animales , Gotas Lipídicas , Actinas , Adipogénesis , Drosophila , Lipasa , Peroxidasas , Proteínas de Drosophila/genética
19.
Development ; 150(20)2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37795747

RESUMEN

Obesity is linked to reduced fertility in various species, from Drosophila to humans. Considering that obesity is often induced by changes in diet or eating behavior, it remains unclear whether obesity, diet, or both reduce fertility. Here, we show that Drosophila females on a high-sugar diet become rapidly obese and less fertile as a result of increased death of early germline cysts and vitellogenic egg chambers (or follicles). They also have high glycogen, glucose and trehalose levels and develop insulin resistance in their fat bodies (but not ovaries). By contrast, females with adipocyte-specific knockdown of the anti-obesity genes brummer or adipose are obese but have normal fertility. Remarkably, females on a high-sugar diet supplemented with a separate source of water have mostly normal fertility and glucose levels, despite persistent obesity, high glycogen and trehalose levels, and fat body insulin resistance. These findings demonstrate that a high-sugar diet affects specific processes in oogenesis independently of insulin resistance, that high glucose levels correlate with reduced fertility on a high-sugar diet, and that obesity alone does not impair fertility.


Asunto(s)
Drosophila melanogaster , Resistencia a la Insulina , Animales , Humanos , Femenino , Drosophila melanogaster/genética , Trehalosa , Obesidad/etiología , Dieta , Drosophila , Fertilidad , Glucosa , Glucógeno
20.
Development ; 150(6)2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36807509

RESUMEN

During Drosophila oogenesis, somatic follicle cells (FCs) differentiate to secrete components of the eggshell. Before secretion, the epithelium reorganizes to shape eggshell specializations, including border FC collective cell migration and later dorsal formation. These FC movements provide valuable insights into collective cell migration. However, little is known about centripetal migration, which encloses the oocyte after secretion has begun. Centripetal migration begins with apical extension of a few FCs that move away from the basement membrane to invade between germ cells. We define a timeline of reproducible milestones, using time-lapse imaging of egg chamber explants. Inward migration occurs in two phases. First, leading centripetal FCs ingress, extending apically over the anterior oocyte, and constricting basally. Second, following FCs move collectively toward the anterior, then around the corner to move inward with minimal change in aspect ratio. E-cadherin was required in leading centripetal FCs for their normal ingression, assessed with homozygous shotgun mutant or RNAi knockdown clones; ingression was influenced non-autonomously by mutant following FCs. This work establishes centripetal migration as an accessible model for biphasic E-cadherin-adhesion-mediated collective migration.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Femenino , Drosophila melanogaster/metabolismo , Folículo Ovárico/metabolismo , Drosophila/metabolismo , Oogénesis , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Movimiento Celular , Cadherinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA