Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Cell Dev Biol ; 34: 59-84, 2018 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-30074816

RESUMEN

In recent years, thin membrane protrusions such as cytonemes and tunneling nanotubes have emerged as a novel mechanism of intercellular communication. Protrusion-based cellular interactions allow for specific communication between participating cells and have a distinct spectrum of advantages compared to secretion- and diffusion-based intercellular communication. Identification of protrusion-based signaling in diverse systems suggests that this mechanism is a ubiquitous and prevailing means of communication employed by many cell types. Moreover, accumulating evidence indicates that protrusion-based intercellular communication is often involved in pathogenesis, including cancers and infections. Here we review our current understanding of protrusion-based intercellular communication.


Asunto(s)
Comunicación Celular/genética , Linaje de la Célula/genética , Extensiones de la Superficie Celular/genética , Endocitosis/genética , Humanos , Nanotubos/química , Transducción de Señal/genética
2.
Annu Rev Genet ; 57: 181-199, 2023 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-37552892

RESUMEN

Germ cells are the only cell type that is capable of transmitting genetic information to the next generation, which has enabled the continuation of multicellular life for the last 1.5 billion years. Surprisingly little is known about the mechanisms supporting the germline's remarkable ability to continue in this eternal cycle, termed germline immortality. Even unicellular organisms age at a cellular level, demonstrating that cellular aging is inevitable. Extensive studies in yeast have established the framework of how asymmetric cell division and gametogenesis may contribute to the resetting of cellular age. This review examines the mechanisms of germline immortality-how germline cells reset the aging of cells-drawing a parallel between yeast and multicellular organisms.


Asunto(s)
División Celular Asimétrica , Saccharomyces cerevisiae , División Celular Asimétrica/genética , Saccharomyces cerevisiae/genética , Células Germinativas , Células Madre
3.
PLoS Genet ; 20(5): e1011136, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38758955

RESUMEN

Ribosomal DNA (rDNA), which encodes ribosomal RNA, is an essential but unstable genomic element due to its tandemly repeated nature. rDNA's repetitive nature causes spontaneous intrachromatid recombination, leading to copy number (CN) reduction, which must be counteracted by a mechanism that recovers CN to sustain cells' viability. Akin to telomere maintenance, rDNA maintenance is particularly important in cell types that proliferate for an extended time period, most notably in the germline that passes the genome through generations. In Drosophila, the process of rDNA CN recovery, known as 'rDNA magnification', has been studied extensively. rDNA magnification is mediated by unequal sister chromatid exchange (USCE), which generates a sister chromatid that gains the rDNA CN by stealing copies from its sister. However, much remains elusive regarding how germ cells sense rDNA CN to decide when to initiate magnification, and how germ cells balance between the need to generate DNA double-strand breaks (DSBs) to trigger USCE vs. avoiding harmful DSBs. Recently, we identified an rDNA-binding Zinc-finger protein Indra as a factor required for rDNA magnification, however, the underlying mechanism of action remains unknown. Here we show that Indra is a negative regulator of rDNA magnification, balancing the need of rDNA magnification and repression of dangerous DSBs. Mechanistically, we show that Indra is a repressor of RNA polymerase II (Pol II)-dependent transcription of rDNA: Under low rDNA CN conditions, Indra protein amount is downregulated, leading to Pol II-mediated transcription of rDNA. This results in the expression of rDNA-specific retrotransposon, R2, which we have shown to facilitate rDNA magnification via generation of DBSs at rDNA. We propose that differential use of Pol I and Pol II plays a critical role in regulating rDNA CN expansion only when it is necessary.


Asunto(s)
ADN Ribosómico , ARN Polimerasa II , Transcripción Genética , Animales , ADN Ribosómico/genética , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Roturas del ADN de Doble Cadena , Drosophila melanogaster/genética , Intercambio de Cromátides Hermanas/genética , Células Germinativas/metabolismo , Variaciones en el Número de Copia de ADN
4.
PLoS Genet ; 20(6): e1011241, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38870220

RESUMEN

Although introns are typically tens to thousands of nucleotides, there are notable exceptions. In flies as well as humans, a small number of genes contain introns that are more than 1000 times larger than typical introns, exceeding hundreds of kilobases (kb) to megabases (Mb). It remains unknown why gigantic introns exist and how cells overcome the challenges associated with their transcription and RNA processing. The Drosophila Y chromosome contains some of the largest genes identified to date: multiple genes exceed 4Mb, with introns accounting for over 99% of the gene span. Here we demonstrate that co-transcriptional splicing of these gigantic Y-linked genes is important to ensure successful transcription: perturbation of splicing led to the attenuation of transcription, leading to a failure to produce mature mRNA. Cytologically, defective splicing of the Y-linked gigantic genes resulted in disorganization of transcripts within the nucleus suggestive of entanglement of transcripts, likely resulting from unspliced long RNAs. We propose that co-transcriptional splicing maintains the length of nascent transcripts of gigantic genes under a critical threshold, preventing their entanglement and ensuring proper gene expression. Our study reveals a novel biological significance of co-transcriptional splicing.


Asunto(s)
Drosophila melanogaster , Intrones , Empalme del ARN , Transcripción Genética , Empalme del ARN/genética , Animales , Intrones/genética , Drosophila melanogaster/genética , Cromosoma Y/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Masculino , Humanos
5.
Semin Cell Dev Biol ; 156: 152-159, 2024 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-37852904

RESUMEN

Abundant tandemly repeated satellite DNA is present in most eukaryotic genomes. Previous limitations including a pervasive view that it was uninteresting junk DNA, combined with challenges in studying it, are starting to dissolve - and recent studies have found important functions for satellite DNAs. The observed rapid evolution and implied instability of satellite DNA now has important significance for their functions and maintenance within the genome. In this review, we discuss the processes that lead to satellite DNA copy number instability, and the importance of mechanisms to manage the potential negative effects of instability. Satellite DNA is vulnerable to challenges during replication and repair, since it forms difficult-to-process secondary structures and its homology within tandem arrays can result in various types of recombination. Satellite DNA instability may be managed by DNA or chromatin-binding proteins ensuring proper nuclear localization and repair, or by proteins that process aberrant structures that satellite DNAs tend to form. We also discuss the pattern of satellite DNA mutations from recent mutation accumulation (MA) studies that have tracked changes in satellite DNA for up to 1000 generations with minimal selection. Finally, we highlight examples of satellite evolution from studies that have characterized satellites across millions of years of Drosophila fruit fly evolution, and discuss possible ways that selection might act on the satellite DNA composition.


Asunto(s)
ADN Satélite , Drosophila , Animales , ADN Satélite/genética , Drosophila/genética , Mutación , Evolución Molecular
6.
Proc Natl Acad Sci U S A ; 120(23): e2221613120, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37252996

RESUMEN

Ribosomal DNA (rDNA) loci contain hundreds of tandemly repeated copies of ribosomal RNA genes needed to support cellular viability. This repetitiveness makes it highly susceptible to copy number (CN) loss due to intrachromatid recombination between rDNA copies, threatening multigenerational maintenance of rDNA. How this threat is counteracted to avoid extinction of the lineage has remained unclear. Here, we show that the rDNA-specific retrotransposon R2 is essential for restorative rDNA CN expansion to maintain rDNA loci in the Drosophila male germline. The depletion of R2 led to defective rDNA CN maintenance, causing a decline in fecundity over generations and eventual extinction. We find that double-stranded DNA breaks created by the R2 endonuclease, a feature of R2's rDNA-specific retrotransposition, initiate the process of rDNA CN recovery, which relies on homology-dependent repair of the DNA break at rDNA copies. This study reveals that an active retrotransposon provides an essential function for its host, contrary to transposable elements' reputation as entirely selfish. These findings suggest that benefiting host fitness can be an effective selective advantage for transposable elements to offset their threat to the host, which may contribute to retrotransposons' widespread success throughout taxa.


Asunto(s)
Drosophila , Retroelementos , Animales , Retroelementos/genética , ADN Ribosómico/genética , Drosophila/genética , Elementos Transponibles de ADN
7.
Proc Natl Acad Sci U S A ; 120(47): e2314440120, 2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-37967216

RESUMEN

Ribosomal DNA (rDNA) encodes ribosomal RNA and exists as tandem repeats of hundreds of copies in the eukaryotic genome to meet the high demand of ribosome biogenesis. Tandemly repeated DNA elements are inherently unstable; thus, mechanisms must exist to maintain rDNA copy number (CN), in particular in the germline that continues through generations. A phenomenon called rDNA magnification was discovered over 50 y ago in Drosophila as a process that recovers the rDNA CN on chromosomes that harbor minimal CN. Our recent studies indicated that rDNA magnification is the mechanism to maintain rDNA CN under physiological conditions to counteract spontaneous CN loss that occurs during aging. Our previous studies that explored the mechanism of rDNA magnification implied that asymmetric division of germline stem cells (GSCs) may be particularly suited to achieve rDNA magnification. However, it remains elusive whether GSCs are the unique cell type that undergoes rDNA magnification or differentiating germ cells are also capable of magnification. In this study, we provide empirical evidence that suggests that rDNA magnification operates uniquely in GSCs, but not in differentiating germ cells. We further provide computer simulation that suggests that rDNA magnification is only achievable through asymmetric GSC divisions. We propose that despite known plasticity and transcriptomic similarity between GSCs and differentiating germ cells, GSCs' unique ability to divide asymmetrically serves a critical role of maintaining rDNA CN through generations, supporting germline immortality.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , ADN Ribosómico/genética , ADN Ribosómico/metabolismo , Simulación por Computador , Drosophila/genética , Drosophila/metabolismo , Células Germinativas/metabolismo , Células Madre/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo
8.
Proc Natl Acad Sci U S A ; 120(16): e2220576120, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-37036962

RESUMEN

Across species, sperm maturation involves the dramatic reconfiguration of chromatin into highly compact nuclei that enhance hydrodynamic ability and ensure paternal genomic integrity. This process is mediated by the replacement of histones by sperm nuclear basic proteins, also referred to as protamines. In humans, a carefully balanced dosage between two known protamine genes is required for optimal fertility. However, it remains unknown how their proper balance is regulated and how defects in balance may lead to compromised fertility. Here, we show that a nucleolar protein, modulo, a homolog of nucleolin, mediates the histone-to-protamine transition during Drosophila spermatogenesis. We find that modulo mutants display nuclear compaction defects during late spermatogenesis due to decreased expression of autosomal protamine genes (including Mst77F) and derepression of Y-linked multicopy Mst77F homologs (Mst77Y), leading to the mutant's known sterility. Overexpression of Mst77Y in a wild-type background is sufficient to cause nuclear compaction defects, similar to modulo mutant, indicating that Mst77Y is a dominant-negative variant interfering with the process of histone-to-protamine transition. Interestingly, ectopic overexpression of Mst77Y caused decompaction of X-bearing spermatids nuclei more frequently than Y-bearing spermatid nuclei, although this did not greatly affect the sex ratio of offspring. We further show that modulo regulates these protamine genes at the step of transcript polyadenylation. We conclude that the regulation of protamines mediated by modulo, ensuring the expression of functional ones while repressing dominant-negative ones, is critical for male fertility.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Humanos , Animales , Masculino , Drosophila melanogaster/metabolismo , Histonas/genética , Histonas/metabolismo , Protaminas/genética , Protaminas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Semen/metabolismo , Espermatozoides/metabolismo , Cromatina/metabolismo , Espermatogénesis/genética , Drosophila/genética
9.
Semin Cell Dev Biol ; 128: 69-77, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35469677

RESUMEN

Satellite DNAs are arrays of tandem repeats found in the eukaryotic genome. They are mainly found in pericentromeric heterochromatin and have been believed to be mostly inert, leading satellite DNAs to be erroneously regarded as junk. Recent studies have started to elucidate the function of satellite DNA, yet little is known about the peculiar case where satellite DNA is found within the introns of protein coding genes, resulting in incredibly large introns, a phenomenon termed intron gigantism. Studies in Drosophila demonstrated that satellite DNA-containing introns are transcribed with the gene and require specialized mechanisms to overcome the burdens imposed by the extremely long stretches of repetitive DNA. Whether intron gigantism confers any benefit or serves any functional purpose for cells and/or organisms remains elusive. Here we review our current understanding of intron gigantism: where it is found, the challenges it imposes, how it is regulated and what purpose it may serve.


Asunto(s)
ADN Satélite , Gigantismo , Animales , ADN Satélite/genética , Drosophila/genética , Gigantismo/genética , Heterocromatina/genética , Intrones
10.
PLoS Comput Biol ; 19(2): e1010875, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36821548

RESUMEN

From insects to mammals, oocytes and sperm develop within germline cysts comprising cells connected by intercellular bridges (ICBs). In numerous insects, formation of the cyst is accompanied by growth of the fusome-a membranous organelle that permeates the cyst. Fusome composition and function are best understood in Drosophila melanogaster: during oogenesis, the fusome dictates cyst topology and size and facilitates oocyte selection, while during spermatogenesis, the fusome synchronizes the cyst's response to DNA damage. Despite its distinct and sex-specific roles during insect gametogenesis, elucidating fusome growth and inheritance in females and its structure and connectivity in males has remained challenging. Here, we take advantage of advances in three-dimensional (3D) confocal microscopy and computational image processing tools to reconstruct the topology, growth, and distribution of the fusome in both sexes. In females, our experimental findings inform a theoretical model for fusome assembly and inheritance and suggest that oocyte selection proceeds through an 'equivalency with a bias' mechanism. In males, we find that cell divisions can deviate from the maximally branched pattern observed in females, leading to greater topological variability. Our work consolidates existing disjointed experimental observations and contributes a readily generalizable computational approach for quantitative studies of gametogenesis within and across species.


Asunto(s)
Drosophila melanogaster , Semen , Animales , Femenino , Masculino , Drosophila melanogaster/genética , Oocitos , Oogénesis/genética , División Celular , Mamíferos
11.
Mol Biol Evol ; 39(11)2022 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-36223491

RESUMEN

Karyotype refers to the configuration of the genome into a set of chromosomes. The karyotype difference between species is expected to impede various biological processes, such as chromosome segregation and meiotic chromosome pairing, potentially contributing to incompatibility. Karyotypes can rapidly change between closely related species and even among populations of the same species. However, the forces driving karyotype evolution are poorly understood. Here we describe a unique karyotype of a Drosophila melanogaster strain isolated from the Seychelles archipelago. This strain has lost the ribosomal DNA (rDNA) locus on the X chromosome. Because the Y chromosome is the only other rDNA-bearing chromosome, all females carry at least one Y chromosome as the source of rDNA. Interestingly, we found that the strain also carries a truncated Y chromosome (YS) that is stably maintained in the population despite its inability to support male fertility. Our modeling and cytological analysis suggest that the Y chromosome has a larger negative impact on female fitness than the YS chromosome. Moreover, we generated an independent strain that lacks X rDNA and has a karyotype of XXY females and XY males. This strain quickly evolved multiple karyotypes: two new truncated Y chromosomes (similar to YS), as well as two independent X chromosome fusions that contain the Y-derived rDNA fragment, eliminating females' dependence on the Y chromosome. Considering that Robertsonian fusions frequently occur at rDNA loci in humans, we propose that rDNA loci instability may be one of driving forces of karyotype evolution.


Asunto(s)
Drosophila melanogaster , Cromosoma Y , Animales , Masculino , Femenino , Humanos , ADN Ribosómico/genética , Drosophila melanogaster/genética , Cariotipo , Cromosoma Y/genética , Cromosoma X/genética
12.
J Cell Sci ; 134(14)2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34164657

RESUMEN

Tissue-specific stem cells maintain tissue homeostasis by providing a continuous supply of differentiated cells throughout the life of organisms. Differentiated/differentiating cells can revert back to a stem cell identity via dedifferentiation to help maintain the stem cell pool beyond the lifetime of individual stem cells. Although dedifferentiation is important for maintaining the stem cell population, it is speculated that it underlies tumorigenesis. Therefore, this process must be tightly controlled. Here, we show that a translational regulator, me31B, plays a critical role in preventing excess dedifferentiation in the Drosophila male germline: in the absence of me31B, spermatogonia dedifferentiate into germline stem cells (GSCs) at a dramatically elevated frequency. Our results show that the excess dedifferentiation is likely due to misregulation of nos, a key regulator of germ cell identity and GSC maintenance. Taken together, our data reveal negative regulation of dedifferentiation to balance stem cell maintenance with differentiation.


Asunto(s)
ARN Helicasas DEAD-box , Proteínas de Drosophila , Drosophila , Células Germinativas , Células Madre , Animales , Diferenciación Celular , ARN Helicasas DEAD-box/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Homeostasis , Masculino , Espermatogonias
13.
PLoS Genet ; 16(3): e1008648, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32168327

RESUMEN

The piRNA pathway protects germline genomes from selfish genetic elements (e.g. transposons) through their transcript cleavage in the cytoplasm and/or their transcriptional silencing in the nucleus. Here, we describe a mechanism by which the nuclear and cytoplasmic arms of the piRNA pathway are linked. We find that during mitosis of Drosophila spermatogonia, nuclear Piwi interacts with nuage, the compartment that mediates the cytoplasmic arm of the piRNA pathway. At the end of mitosis, Piwi leaves nuage to return to the nucleus. Dissociation of Piwi from nuage occurs at the depolymerizing microtubules of the central spindle, mediated by a microtubule-depolymerizing kinesin, Klp10A. Depletion of klp10A delays the return of Piwi to the nucleus and affects piRNA production, suggesting the role of nuclear-cytoplasmic communication in piRNA biogenesis. We propose that cell cycle-dependent communication between the nuclear and cytoplasmic arms of the piRNA pathway may play a previously unappreciated role in piRNA regulation.


Asunto(s)
Proteínas Argonautas/metabolismo , Proteínas de Drosophila/metabolismo , Cinesinas/metabolismo , ARN Interferente Pequeño/genética , Animales , Proteínas Argonautas/genética , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , División Celular/fisiología , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Elementos Transponibles de ADN , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Silenciador del Gen , Células Germinativas , Cinesinas/genética , Masculino , Ovario/metabolismo , ARN Interferente Pequeño/metabolismo
14.
Mol Biol Evol ; 38(11): 4977-4986, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34302471

RESUMEN

Although rapid evolution of pericentromeric satellite DNA repeats is theorized to promote hybrid incompatibility (HI) (Yunis and Yasmineh 1971; Henikoff et al. 2001; Ferree and Barbash 2009; Sawamura 2012; Jagannathan and Yamashita 2017), how divergent repeats affect hybrid cells remains poorly understood. Recently, we demonstrated that sequence-specific DNA-binding proteins cluster satellite DNA from multiple chromosomes into "chromocenters," thereby bundling chromosomes to maintain the entire genome in a single nucleus (Jagannathan et al. 2018, 2019). Here, we show that ineffective clustering of divergent satellite DNA in the cells of Drosophila hybrids results in chromocenter disruption, associated micronuclei formation, and tissue atrophy. We further demonstrate that previously identified HI factors trigger chromocenter disruption and micronuclei in hybrids, linking their function to a conserved cellular process. Together, we propose a unifying framework that explains how the widely observed satellite DNA divergence between closely related species can cause reproductive isolation.


Asunto(s)
ADN Satélite , Drosophila , Animales , Cromosomas , Análisis por Conglomerados , ADN Satélite/genética , Drosophila/genética , Aislamiento Reproductivo
15.
Trends Genet ; 35(10): 734-742, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31395390

RESUMEN

rDNA, the genes encoding the RNA components of ribosomes (rRNA), are highly repetitive in all eukaryotic genomes, containing 100s to 1000s of copies, to meet the demand for ribosome biogenesis. rDNA genes are arranged in large stretches of tandem repeats, forming loci that are highly susceptible to copy loss due to their repetitiveness and active transcription throughout the cell cycle. Despite this inherent instability, rDNA copy number is generally maintained within a particular range in each species, pointing to the presence of mechanisms that maintain rDNA copy number in a homeostatic range. In this review, we summarize the current understanding of these maintenance mechanisms and how they sustain rDNA copy number throughout populations.


Asunto(s)
Variaciones en el Número de Copia de ADN , ADN Ribosómico , Dosificación de Gen , Animales , Sitios Genéticos , Células Germinativas/metabolismo , Humanos , Especificidad de la Especie
16.
PLoS Genet ; 15(5): e1008028, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31071079

RESUMEN

Intron gigantism, where genes contain megabase-sized introns, is observed across species, yet little is known about its purpose or regulation. Here we identify a unique gene expression program utilized for the proper expression of genes with intron gigantism. We find that two Drosophila genes with intron gigantism, kl-3 and kl-5, are transcribed in a spatiotemporal manner over the course of spermatocyte differentiation, which spans ~90 hours. The introns of these genes contain megabases of simple satellite DNA repeats that comprise over 99% of the gene loci, and these satellite-DNA containing introns are transcribed. We identify two RNA-binding proteins that specifically localize to kl-3 and kl-5 transcripts and are needed for the successful transcription or processing of these genes. We propose that genes with intron gigantism require a unique gene expression program, which may serve as a platform to regulate gene expression during cellular differentiation.


Asunto(s)
ADN Satélite/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Intrones , Espermatocitos/metabolismo , Espermatogénesis/genética , Animales , ADN Satélite/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Exones , Regulación del Desarrollo de la Expresión Génica , Masculino , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Espermatocitos/citología , Espermatocitos/crecimiento & desarrollo , Transcripción Genética , Cromosoma Y/química
17.
Nature ; 523(7560): 329-32, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26131929

RESUMEN

Stem cell niches provide resident stem cells with signals that specify their identity. Niche signals act over a short range such that only stem cells but not their differentiating progeny receive the self-renewing signals. However, the cellular mechanisms that limit niche signalling to stem cells remain poorly understood. Here we show that the Drosophila male germline stem cells form previously unrecognized structures, microtubule-based nanotubes, which extend into the hub, a major niche component. Microtubule-based nanotubes are observed specifically within germline stem cell populations, and require intraflagellar transport proteins for their formation. The bone morphogenetic protein (BMP) receptor Tkv localizes to microtubule-based nanotubes. Perturbation of microtubule-based nanotubes compromises activation of Dpp signalling within germline stem cells, leading to germline stem cell loss. Moreover, Dpp ligand and Tkv receptor interaction is necessary and sufficient for microtubule-based nanotube formation. We propose that microtubule-based nanotubes provide a novel mechanism for selective receptor-ligand interaction, contributing to the short-range nature of niche-stem-cell signalling.


Asunto(s)
Drosophila melanogaster/citología , Microtúbulos/química , Microtúbulos/metabolismo , Nanotubos , Transducción de Señal , Nicho de Células Madre/fisiología , Testículo/citología , Animales , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Ligandos , Masculino , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Superficie Celular/metabolismo
18.
Trends Genet ; 33(12): 971-978, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28947158

RESUMEN

Germ cells develop as a cyst of interconnected sibling cells in a broad range of organisms in both sexes. A well-established function of intercellular connectivity is to transport cytoplasmic materials from 'nurse' cells to oocytes, a critical process for developing functional oocytes in ovaries of many species. However, there are situations where connectivity exists without a nursing mechanism, and the biological meaning of such connectivity remains obscure. In this review, we summarize current knowledge on the formation of intercellular connectivity, and discuss its meaning by visiting multiple examples of germ cell connectivity observed in evolutionarily distant species.


Asunto(s)
Células Germinativas/fisiología , Animales , Citoplasma/fisiología , Femenino , Oocitos/fisiología , Oogénesis/fisiología , Ovario/fisiología
19.
Semin Cell Dev Biol ; 128: 1, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35577697
20.
Nature ; 498(7453): 251-4, 2013 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-23644460

RESUMEN

Adult stem cells undergo asymmetric cell division to self-renew and give rise to differentiated cells that comprise mature tissue. Sister chromatids may be distinguished and segregated nonrandomly in asymmetrically dividing stem cells, although the underlying mechanism and the purpose it may serve remain elusive. Here we develop the CO-FISH (chromosome orientation fluorescence in situ hybridization) technique with single-chromosome resolution and show that sister chromatids of X and Y chromosomes, but not autosomes, are segregated nonrandomly during asymmetric divisions of Drosophila male germline stem cells. This provides the first direct evidence, to our knowledge, that two sister chromatids containing identical genetic information can be distinguished and segregated nonrandomly during asymmetric stem-cell divisions. We further show that the centrosome, SUN-KASH nuclear envelope proteins and Dnmt2 (also known as Mt2) are required for nonrandom sister chromatid segregation. Our data indicate that the information on X and Y chromosomes that enables nonrandom segregation is primed during gametogenesis in the parents. Moreover, we show that sister chromatid segregation is randomized in germline stem cell overproliferation and dedifferentiated germline stem cells. We propose that nonrandom sister chromatid segregation may serve to transmit distinct information carried on two sister chromatids to the daughters of asymmetrically dividing stem cells.


Asunto(s)
Cromátides/metabolismo , Segregación Cromosómica , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Células Madre/citología , Animales , Secuencia de Bases , Desdiferenciación Celular , División Celular , Centrosoma/metabolismo , Cromátides/genética , ADN (Citosina-5-)-Metiltransferasas , Proteínas de Drosophila , Drosophila melanogaster/genética , Masculino , Datos de Secuencia Molecular , Espermatogonias/citología , Células Madre/metabolismo , Testículo/citología , Cromosoma X/genética , Cromosoma X/metabolismo , Cromosoma Y/genética , Cromosoma Y/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA