Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
PLoS Genet ; 17(10): e1009834, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34644293

RESUMEN

Stem cells have the potential to maintain undifferentiated state and differentiate into specialized cell types. Despite numerous progress has been achieved in understanding stem cell self-renewal and differentiation, many fundamental questions remain unanswered. In this study, we identify dRTEL1, the Drosophila homolog of Regulator of Telomere Elongation Helicase 1, as a novel regulator of male germline stem cells (GSCs). Our genome-wide transcriptome analysis and ChIP-Seq results suggest that dRTEL1 affects a set of candidate genes required for GSC maintenance, likely independent of its role in DNA repair. Furthermore, dRTEL1 prevents DNA damage-induced checkpoint activation in GSCs. Finally, dRTEL1 functions to sustain Stat92E protein levels, the key player in GSC maintenance. Together, our findings reveal an intrinsic role of the DNA helicase dRTEL1 in maintaining male GSC and provide insight into the function of dRTEL1.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/genética , Células Germinativas/fisiología , Células Madre/fisiología , Animales , Autorrenovación de las Células/genética , ADN Helicasas/metabolismo , Reparación del ADN/genética , Femenino , Masculino , Transducción de Señal/genética , Transcriptoma/genética
2.
Hum Genet ; 142(7): 879-907, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37148394

RESUMEN

Premature ovarian insufficiency (POI) is a common cause of infertility in women, characterised by amenorrhea and elevated FSH under the age of 40 years. In some cases, POI is syndromic in association with other features such as sensorineural hearing loss in Perrault syndrome. POI is a heterogeneous disease with over 80 causative genes known so far; however, these explain only a minority of cases. Using whole-exome sequencing (WES), we identified a MRPL50 homozygous missense variant (c.335T > A; p.Val112Asp) shared by twin sisters presenting with POI, bilateral high-frequency sensorineural hearing loss, kidney and heart dysfunction. MRPL50 encodes a component of the large subunit of the mitochondrial ribosome. Using quantitative proteomics and western blot analysis on patient fibroblasts, we demonstrated a loss of MRPL50 protein and an associated destabilisation of the large subunit of the mitochondrial ribosome whilst the small subunit was preserved. The mitochondrial ribosome is responsible for the translation of subunits of the mitochondrial oxidative phosphorylation machinery, and we found patient fibroblasts have a mild but significant decrease in the abundance of mitochondrial complex I. These data support a biochemical phenotype associated with MRPL50 variants. We validated the association of MRPL50 with the clinical phenotype by knockdown/knockout of mRpL50 in Drosophila, which resulted abnormal ovarian development. In conclusion, we have shown that a MRPL50 missense variant destabilises the mitochondrial ribosome, leading to oxidative phosphorylation deficiency and syndromic POI, highlighting the importance of mitochondrial support in ovarian development and function.


Asunto(s)
Disgenesia Gonadal 46 XX , Pérdida Auditiva Sensorineural , Insuficiencia Ovárica Primaria , Femenino , Humanos , Disgenesia Gonadal 46 XX/genética , Pérdida Auditiva Sensorineural/genética , Mitocondrias/genética , Mutación Missense , Insuficiencia Ovárica Primaria/genética , Animales , Drosophila melanogaster
3.
EMBO J ; 34(10): 1319-35, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25759216

RESUMEN

Snail family members regulate epithelial-to-mesenchymal transition (EMT) during invasion of intestinal tumours, but their role in normal intestinal homeostasis is unknown. Studies in breast and skin epithelia indicate that Snail proteins promote an undifferentiated state. Here, we demonstrate that conditional knockout of Snai1 in the intestinal epithelium results in apoptotic loss of crypt base columnar stem cells and bias towards differentiation of secretory lineages. In vitro organoid cultures derived from Snai1 conditional knockout mice also undergo apoptosis when Snai1 is deleted. Conversely, ectopic expression of Snai1 in the intestinal epithelium in vivo results in the expansion of the crypt base columnar cell pool and a decrease in secretory enteroendocrine and Paneth cells. Following conditional deletion of Snai1, the intestinal epithelium fails to produce a proliferative response following radiation-induced damage indicating a fundamental requirement for Snai1 in epithelial regeneration. These results demonstrate that Snai1 is required for regulation of lineage choice, maintenance of CBC stem cells and regeneration of the intestinal epithelium following damage.


Asunto(s)
Mucosa Intestinal/metabolismo , Intestinos/citología , Factores de Transcripción/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Linaje de la Célula , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Transducción de Señal/genética , Transducción de Señal/fisiología , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
4.
J Cell Physiol ; 233(4): 3262-3273, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28884822

RESUMEN

RNA-binding proteins (RBP) are important facilitators of post-transcriptional gene regulation. We have previously established that nuclear overexpression of the RBP Musashi-2 (MSI2) during male germ cell maturation is detrimental to sperm cell development and fertility. Herein we determine the genes and pathways impacted by the upregulation of Msi2. Microarray analysis and qPCR confirmed differential gene expression in factors fundamental to the cell cycle, cellular proliferation, and cell death. Similarly, comparative protein expression analysis via iTRAQ, immunoblot, and immunolocalization, identified differential expression and localization of important regulators of transcription, translation, RNA processing, and spermatogenesis. Specifically, the testis-expressed transcription factor, Tbx1, and the piRNA regulator of gamete development, Piwil1, were both found to be targeted for translational repression by MSI2. This study provides key evidence to support a fundamental role for MSI2 in post-transcriptional regulation during male gamete development.


Asunto(s)
Proteínas Argonautas/metabolismo , Proteínas de Unión al ARN/metabolismo , Espermatogénesis , Proteínas de Dominio T Box/metabolismo , Animales , Proteínas Argonautas/genética , Regulación de la Expresión Génica , Masculino , Ratones Transgénicos , Modelos Biológicos , Proteoma/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Espermátides/metabolismo , Espermatogénesis/genética , Proteínas de Dominio T Box/genética
5.
Cell Tissue Res ; 364(2): 443-51, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26662055

RESUMEN

Tob1 is a member of the BTG/TOB family of proteins with established antiproliferative function. In Danio rerio and Xenopus laevis, the Tob1 gene is expressed from the one-cell stage through to early gastrula stages, followed in later development by discrete expression in many tissues including the notochord and somites. In both mouse and human, Tob1 is expressed in many adult tissues including the testis and ovary; however, the specific cell types are unknown. We examine Tob1 gene expression in mouse in developing germ cells and in sorted male germ cells (gonocytes, spermatogonia, pachytene spermatocytes and round spermatids) by reverse transcription and droplet digital polymerase chain reaction (RT-ddPCR) and in adult ovary and testis by immunofluorescence with anti-Tob1 protein staining. By RT-ddPCR, Tob1 expression was low in developing male germ cells but was highly expressed in round spermatids. In developing female germ cells undergoing entry into meiosis, it increased 10-fold. Tob1 was also highly expressed in round spermatids and in oocytes in all stages of folliculogenesis. Notably, a marker for P-bodies, Dcp-2, was also highly expressed in round spermatids and all oocyte stages examined. The cytoplasmic presence of Tob1 protein in round spermatids and oocytes and the association of Tob1 protein with Dcp2 in both cell types suggest that Tob1 protein plays a role in post-transcriptional mechanisms.


Asunto(s)
Proteínas Portadoras/biosíntesis , Células Germinales Embrionarias/metabolismo , Endorribonucleasas/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Oocitos/metabolismo , Espermátides/metabolismo , Espermatocitos/metabolismo , Espermatogonias/metabolismo , Animales , Biomarcadores/metabolismo , Femenino , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oogénesis/fisiología , Ovario/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espermatogénesis/fisiología , Testículo/metabolismo
6.
FASEB J ; 29(7): 2759-68, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25782991

RESUMEN

Controlled gene regulation during gamete development is vital for maintaining reproductive potential. During the process of gamete development, male germ cells experience extended periods of inactive transcription despite requirements for continued growth and differentiation. Spermatogenesis therefore provides an ideal model to study the effects of posttranscriptional control on gene regulation. During spermatogenesis posttranscriptional regulation is orchestrated by abundantly expressed RNA-binding proteins. One such group of RNA-binding proteins is the Musashi family, previously identified as a critical regulator of testis germ cell development and meiosis in Drosophila and also shown to be vital to sperm development and reproductive potential in the mouse. We focus in depth on the role and function of the vertebrate Musashi ortholog Musashi-1 (MSI1). Through detailed expression studies and utilizing our novel transgenic Msi1 testis-specific overexpression model, we have identified 2 unique RNA-binding targets of MSI1 in spermatogonia, Msi2 and Erh, and have demonstrated a role for MSI1 in translational regulation. We have also provided evidence to suggest that nuclear import protein, IPO5, facilitates the nuclear translocation of MSI1 to the transcriptionally silenced XY chromatin domain in meiotic pachytene spermatocytes, resulting in the release of MSI1 RNA-binding targets. This firmly establishes MSI1 as a master regulator of posttranscriptional control during early spermatogenesis and highlights the significance of the subcellular localization of RNA binding proteins in relation to their function.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Espermatogénesis/fisiología , Factores de Transcripción/metabolismo , beta Carioferinas/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Proteínas de Ciclo Celular/genética , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Transgénicos , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/genética , Espermatocitos/metabolismo , Espermatogonias/metabolismo , Testículo/citología , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Factores de Transcripción/genética , beta Carioferinas/genética
7.
Dev Biol ; 394(2): 217-27, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25169192

RESUMEN

The development of stem cell daughters into the differentiated state normally requires a cascade of proliferation and differentiation steps that are typically regulated by external signals. The germline cells of most animals, in specific, are associated with somatic support cells and depend on them for normal development. In the male gonad of Drosophila melanogaster, germline cells are completely enclosed by cytoplasmic extensions of somatic cyst cells, and these cysts form a functional unit. Signaling from the germline to the cyst cells via the Epidermal Growth Factor Receptor (EGFR) is required for germline enclosure and has been proposed to provide a temporal signature promoting early steps of differentiation. A temperature-sensitive allele of the EGFR ligand Spitz (Spi) provides a powerful tool for probing the function of the EGRF pathway in this context and for identifying other pathways regulating cyst differentiation via genetic interaction studies. Using this tool, we show that signaling via the Ecdysone Receptor (EcR), a known regulator of developmental timing during larval and pupal development, opposes EGF signaling in testes. In spi mutant animals, reducing either Ecdysone synthesis or the expression of Ecdysone signal transducers or targets in the cyst cells resulted in a rescue of cyst formation and cyst differentiation. Despite of this striking effect in the spi mutant background and the expression of EcR signaling components within the cyst cells, activity of the EcR pathway appears to be dispensable in a wildtype background. We propose that EcR signaling modulates the effects of EGFR signaling by promoting an undifferentiated state in early stage cyst cells.


Asunto(s)
Drosophila melanogaster/embriología , Receptores ErbB/metabolismo , Receptores de Esteroides/metabolismo , Transducción de Señal/fisiología , Testículo/citología , Animales , Diferenciación Celular/fisiología , Cartilla de ADN/genética , Proteínas de Drosophila/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Biol Reprod ; 90(5): 92, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24671879

RESUMEN

Spermatogenesis is a complex developmental process whereby diploid spermatogenic stem cells become haploid and undergo a series of morphological changes to produce physically mature spermatozoa. Crucial to this process are a number of RNA-binding proteins, responsible for the posttranscriptional control of essential mRNAs and particularly pertinent to the two periods of inactive transcription that occur in spermatogenesis. One such group of RNA-binding proteins is the Musashi family, specifically Musashi-1 (MSI1) and Musashi-2 (MSI2), which act as key translational regulators in various stem cell populations and have been linked with the induction of tumorigenesis. In the present study, we examined the differential expression of mammalian MSI1 and MSI2 during germ cell development in the mouse testis. MSI1 was found to be predominately localized in mitotic gonocytes and spermatogonia, whereas MSI2 was detected in meiotic spermatocytes and differentiating spermatids. Extensive examination of the function of Musashi in spermatogenesis was achieved through the use of two transgenic mouse models with germ cell-specific overexpression of full-length isoforms of Msi1 or Msi2. These models demonstrated that aberrant expression of either Msi1 or Msi2 has deleterious effects on normal spermatogenesis, with Msi2 overexpression resulting in male sterility. Studies undertaken on human testicular seminoma tumors provide further insights into the relevance of MSI1 and MSI2 overexpression as diagnostic markers to human stem cell cancers. Overall this study provides further evidence for the unique functions that RNA-binding protein isoforms occupy within spermatogenesis, and introduces the potential manipulation of the Musashi family proteins to elucidate the mechanisms of posttranscriptional gene expression during germ cell development.


Asunto(s)
Proteínas de Unión al ARN/fisiología , Espermatocitos/fisiología , Espermatogénesis/fisiología , Espermatogonias/fisiología , Testículo/fisiología , Animales , Western Blotting , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Meiosis/genética , Meiosis/fisiología , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Isoformas de Proteínas , ARN/química , ARN/genética , Proteínas de Unión al ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espermatocitos/ultraestructura , Espermatogonias/ultraestructura , Estadísticas no Paramétricas , Testículo/citología , Testículo/metabolismo
9.
Cells ; 13(6)2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38534338

RESUMEN

Stem cell maintenance and differentiation can be regulated via the differential activity of transcription factors within stem cells and their progeny. For these factors to be active, they need to be transported from their site of synthesis in the cytoplasm into the nucleus. A tissue-specific requirement for factors involved in nuclear importation is a potential mechanism to regulate stem cell differentiation. We have undertaken a characterization of male sterile importin alpha 1 (Dα1) null alleles in Drosophila and found that Dα1 is required for maintaining germline stem cells (GSCs) in the testis niche. The loss of GSCs can be rescued by ectopic expression of Dα1 within the germline but the animals are still infertile, indicating a second role for Dα1 in spermatogenesis. Expression of a Dα1 dominant negative transgene in GSCs confirmed a functional requirement for Dα1 in GSC maintenance but expression of the transgene in differentiating spermatogonia did not exhibit a phenotype indicating a specific role for Dα1 within GSCs. Our data indicate that Dα1 is utilized as a regulatory protein within GSCs to facilitate nuclear importation of proteins that maintain the stem cell pool.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Masculino , Drosophila/metabolismo , Testículo/metabolismo , Proteínas de Drosophila/metabolismo , alfa Carioferinas/metabolismo , Transducción de Señal/fisiología , Células Madre , Factores de Transcripción/metabolismo , Espermatogonias/metabolismo
10.
J Mol Histol ; 55(1): 121-138, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38165569

RESUMEN

Differentiation of lens fiber cells involves a complex interplay of signals from growth factors together with tightly regulated gene expression via transcriptional and post-transcriptional regulators. Various studies have demonstrated that RNA-binding proteins, functioning in ribonucleoprotein granules, have important roles in regulating post-transcriptional expression during lens development. In this study, we examined the expression and localization of two members of the BTG/TOB family of RNA-binding proteins, TOB1 and TOB2, in the developing lens and examined the phenotype of mice that lack Tob1. By RT-PCR, both Tob1 and Tob2 mRNA were detected in epithelial and fiber cells of embryonic and postnatal murine lenses. In situ hybridization showed Tob1 and Tob2 mRNA were most intensely expressed in the early differentiating fibers, with weaker expression in anterior epithelial cells, and both appeared to be downregulated in the germinative zone of E15.5 lenses. TOB1 protein was detected from E11.5 to E16.5 and was predominantly detected in large cytoplasmic puncta in early differentiating fiber cells, often co-localizing with the P-body marker, DCP2. Occasional nuclear puncta were also observed. By contrast, TOB2 was detected in a series of interconnected peri-nuclear granules, in later differentiating fiber cells of the inner cortex. TOB2 did not appear to co-localize with DCP2 but did partially co-localize with an early stress granule marker (EIF3B). These data suggest that TOB1 and TOB2 are involved with different aspects of the mRNA processing cycle in lens fiber cells. In vitro experiments using rat lens epithelial explants treated with or without a fiber differentiating dose of FGF2 showed that both TOB1 and TOB2 were up-regulated during FGF-induced differentiation. In differentiating explants, TOB1 also co-localized with DCP2 in large cytoplasmic granules. Analyses of Tob1-/- mice revealed relatively normal lens morphology but a subtle defect in cell cycle arrest of some cells at the equator and in the lens fiber mass of E13.5 embryos. Overall, these findings suggest that TOB proteins play distinct regulatory roles in RNA processing during lens fiber differentiation.


Asunto(s)
Proteínas de Ciclo Celular , Procesamiento Postranscripcional del ARN , Animales , Ratones , Ratas , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Gránulos de Ribonucleoproteínas Citoplasmáticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética
11.
Biochim Biophys Acta ; 1819(6): 616-30, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22326858

RESUMEN

Gametogenesis is the process by which sperm or ova are produced in the gonads. It is governed by a tightly controlled series of gene expression events, with some common and others distinct for males and females. Nucleocytoplasmic transport is of central importance to the fidelity of gene regulation that is required to achieve the precisely regulated germ cell differentiation essential for fertility. In this review we discuss the physiological importance for gamete formation of the molecules involved in classical nucleocytoplasmic protein transport, including importins/karyopherins, Ran and nucleoporins. To address what functions/factors are conserved or specialized for these developmental processes between species, we compare knowledge from mice, flies and worms. The present analysis provides evidence of the necessity for and specificity of each nuclear transport factor and for nucleoporins during germ cell differentiation. This article is part of a Special Issue entitled: Nuclear Transport and RNA Processing.


Asunto(s)
Gametogénesis/genética , Regulación de la Expresión Génica/genética , Carioferinas , Proteínas de Complejo Poro Nuclear , Transporte Activo de Núcleo Celular/genética , Animales , Citoplasma , Carioferinas/genética , Carioferinas/metabolismo , Ratones , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Transporte de Proteínas/genética
12.
Adv Exp Med Biol ; 786: 1-4, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23696348

RESUMEN

This volume describes the latest findings on transcriptional and translational regulation of stem cells. Both transcriptional activators and repressors have been shown to be crucial for the maintenance of the stem cell state. A key element of stem cell maintenance is repression of differentiation factors or developmental genes - achieved transcriptionally, epigenetically by the Polycomb complex, and post-transcriptionally by RNA-binding proteins and microRNAs. This volume takes two approaches to this topic - (1) illustrating the general principles outlined above through a series of different stem cell examples - embryonic, iPS and adult stem cells, and (2) describing several molecular families that have been shown to have roles in regulation of multiple stem cell populations.


Asunto(s)
Células Madre Adultas/citología , Células Madre Embrionarias/citología , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Células Madre Adultas/metabolismo , Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
13.
Adv Exp Med Biol ; 786: 269-85, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23696362

RESUMEN

The Myc family proteins are key regulators of animal growth and development, which have critical roles in modulating stem cell behaviour. Since the identification of the oncogenic potential of c-Myc in the early 1980s the mammalian Myc family, which is comprised of c-Myc, N-Myc, and L-Myc, has been studied extensively. dMyc, the only Drosophila member of the Myc gene family, is orthologous to the mammalian c-Myc oncoprotein. Here we discuss key studies addressing the function of the Myc family in stem cell behaviour in both Drosophila Models and mammalian systems.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Células Madre/metabolismo , Factores de Transcripción/genética , Animales , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Mamíferos , MicroARNs/genética , MicroARNs/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Fase S , Transducción de Señal , Células Madre/citología , Factores de Transcripción/metabolismo
14.
Adv Exp Med Biol ; 786: 233-45, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23696360

RESUMEN

In order to maintain their unlimited capacity to divide, stem cells require controlled temporal and spatial protein expression. The Musashi family of RNA-binding proteins have been shown to exhibit this necessary translational control through both repression and activation in order to regulate multiple stem cell populations. This chapter looks in depth at the initial discovery and characterisation of Musashi in the model organism Drosophila, and its subsequent emergence as a master regulator in a number of stem cell populations. Furthermore the unique roles for mammalian Musashi-1 and Musashi-2 in different stem cell types are correlated with the perceived diagnostic power of Musashi expression in specific stem cell derived oncologies. In particular the potential role for Musashi in the identification and treatment of human cancer is considered, with a focus on the role of Musashi-2 in leukaemia. Finally, the manipulation of Musashi expression is proposed as a potential avenue towards the targeted treatment of specific aggressive stem cell cancers.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Leucemia/genética , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/genética , Células Madre/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula/genética , Proliferación Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Regulación de la Expresión Génica , Humanos , Leucemia/metabolismo , Leucemia/patología , Terapia Molecular Dirigida , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Células Madre/citología
15.
Nat Commun ; 14(1): 3403, 2023 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-37296101

RESUMEN

Squamous cell carcinoma antigen recognized by T cells 3 (SART3) is an RNA-binding protein with numerous biological functions including recycling small nuclear RNAs to the spliceosome. Here, we identify recessive variants in SART3 in nine individuals presenting with intellectual disability, global developmental delay and a subset of brain anomalies, together with gonadal dysgenesis in 46,XY individuals. Knockdown of the Drosophila orthologue of SART3 reveals a conserved role in testicular and neuronal development. Human induced pluripotent stem cells carrying patient variants in SART3 show disruption to multiple signalling pathways, upregulation of spliceosome components and demonstrate aberrant gonadal and neuronal differentiation in vitro. Collectively, these findings suggest that bi-allelic SART3 variants underlie a spliceosomopathy which we tentatively propose be termed INDYGON syndrome (Intellectual disability, Neurodevelopmental defects and Developmental delay with 46,XY GONadal dysgenesis). Our findings will enable additional diagnoses and improved outcomes for individuals born with this condition.


Asunto(s)
Disgenesia Gonadal , Células Madre Pluripotentes Inducidas , Discapacidad Intelectual , Masculino , Humanos , Testículo/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Antígenos de Neoplasias
16.
Genesis ; 50(12): 914-20, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22926963

RESUMEN

The Drosophila testis has proven to be a valuable model organ for investigation of germline stem cell (GSC) maintenance and differentiation as well as elucidation of the genetic programs that regulate differentiation of daughter spermatogonia. Development of germ cell specific GAL4 driver transgenes has facilitated investigation of gene function in GSCs and spermatogonia but specific GAL4 tools are not available for analysis of postmitotic spermatogonial differentiation into spermatocytes. We have screened publically available pGT1 strains, a GAL4-encoding gene trap collection, to identify lines that can drive gene expression in late spermatogonia and early spermatocytes. While we were unable to identify any germline-specific drivers, we did identify an insertion in the chiffon locus, which drove expression specifically in early spermatocytes within the germline along with the somatic cyst cells of the testis.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/genética , Elementos de Facilitación Genéticos , Marcación de Gen , Espermatocitos/crecimiento & desarrollo , Factores de Transcripción/genética , Transcripción Genética , Transgenes , Animales , Proteínas de Drosophila/metabolismo , Proteínas del Huevo/genética , Masculino , Espermatocitos/metabolismo
17.
Cell Tissue Res ; 350(2): 385-94, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22961348

RESUMEN

p53 family members have been implicated in regulation of genomic integrity and apoptosis in a variety of tissues. The Drosophila family member, Dmp53, primarily functions to regulate apoptosis in developing and regenerating tissues but loss of function mutants are viable and fertile. Dmp53 exhibits a striking expression pattern in the male germline with high levels found in nuclear bodies in pre-meiotic germ cells. The localisation of Dmp53 to nuclear bodies is dependent upon Dmp53 complexes being able to bind DNA, and although dmp53 mutants do not affect germline stem cell (GSC) maintenance or differentiation, GSCs are sensitive to overexpression of Dmp53 but maturing spermatogonia are not. Dmp53 thus has differential effects depending upon the stage of male germline maturation.


Asunto(s)
Proteínas de Drosophila/metabolismo , Espermatogonias/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Animales Modificados Genéticamente , Apoptosis/fisiología , Diferenciación Celular/fisiología , Línea Celular , Núcleo Celular/metabolismo , Drosophila melanogaster , Masculino , Ratones , Espermatogonias/citología
18.
Mol Cell Biochem ; 369(1-2): 135-45, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22773306

RESUMEN

The Drosophila melanogaster orthologue of the c-Cbl proto-oncogene acts to downregulate signalling from receptor tyrosine kinases by enhancing endocytosis of activated receptors. Expression of an analogue of the C-terminally truncated v-Cbl oncogene, Dv-cbl, in the developing Drosophila eye conversely leads to excess signalling and disruption to the well-ordered adult compound eye. Co-expression of activated Ras with Dv-cbl leads to a severe disruption of eye development. We have used a transposon-based inducible expression system to screen for molecules that can suppress the Dv-cbl phenotype and have identified an allele that upregulates the A-kinase anchoring protein, Akap200. Overexpression of Akap200 not only suppresses the phenotype caused by Dv-cbl expression, but also the severe disruption to eye development caused by the combined expression of Dv-cbl and activated Ras. Akap200 is also endogenously expressed in the developing Drosophila eye at a level that modulates the effects of excessive signalling caused by expression of Dv-cbl.


Asunto(s)
Proteínas de Anclaje a la Quinasa A , Proteínas de Drosophila , Ojo/crecimiento & desarrollo , Proteínas de la Membrana , Proteínas Proto-Oncogénicas c-cbl , Proteínas ras , Proteínas de Anclaje a la Quinasa A/genética , Proteínas de Anclaje a la Quinasa A/metabolismo , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas c-cbl/genética , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Transducción de Señal , Proteínas ras/genética , Proteínas ras/metabolismo
19.
Cell Death Discov ; 8(1): 455, 2022 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-36371343

RESUMEN

The Drosophila ovary is regenerated from germline and somatic stem cell populations that have provided fundamental conceptual understanding on how adult stem cells are regulated within their niches. Recent ovarian transcriptomic studies have failed to identify mRNAs that are specific to follicle stem cells (FSCs), suggesting that their fate may be regulated post-transcriptionally. We have identified that the RNA-binding protein, Musashi (Msi) is required for maintaining the stem cell state of FSCs. Loss of msi function results in stem cell loss, due to a change in differentiation state, indicated by upregulation of Lamin C in the stem cell population. In msi mutant ovaries, Lamin C upregulation was also observed in posterior escort cells that interact with newly formed germ cell cysts. Mutant somatic cells within this region were dysfunctional, as evidenced by the presence of germline cyst collisions, fused egg chambers and an increase in germ cell cyst apoptosis. The msi locus produces two classes of mRNAs (long and short). We show that FSC maintenance and escort cell function specifically requires the long transcripts, thus providing the first evidence of isoform-specific regulation in a population of Drosophila epithelial cells. We further demonstrate that although male germline stem cells have previously been shown to require Msi function to prevent differentiation this is not the case for female germline stem cells, indicating that these similar stem cell types have different requirements for Msi, in addition to the differential use of Msi isoforms between soma and germline. In summary, we show that different isoforms of the Msi RNA-binding protein are expressed in specific cell populations of the ovarian stem cell niche where Msi regulates stem cell differentiation, niche cell function and subsequent germ cell survival and differentiation.

20.
World J Psychiatry ; 11(10): 711-735, 2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34733638

RESUMEN

Mental illness remains the greatest chronic health burden globally with few in-roads having been made despite significant advances in genomic knowledge in recent decades. The field of psychiatry is constantly challenged to bring new approaches and tools to address and treat the needs of vulnerable individuals and subpopulations, and that has to be supported by a continuous growth in knowledge. The majority of neuropsychiatric symptoms reflect complex gene-environment interactions, with epigenetics bridging the gap between genetic susceptibility and environmental stressors that trigger disease onset and drive the advancement of symptoms. It has more recently been demonstrated in preclinical models that epigenetics underpins the transgenerational inheritance of stress-related behavioural phenotypes in both paternal and maternal lineages, providing further supporting evidence for heritability in humans. However, unbiased prospective studies of this nature are practically impossible to conduct in humans so preclinical models remain our best option for researching the molecular pathophysiologies underlying many neuropsychiatric conditions. While rodents will remain the dominant model system for preclinical studies (especially for addressing complex behavioural phenotypes), there is scope to expand current research of the molecular and epigenetic pathologies by using invertebrate models. Here, we will discuss the utility and advantages of two alternative model organisms-Caenorhabditis elegans and Drosophila melanogaster-and summarise the compelling insights of the epigenetic regulation of transgenerational inheritance that are potentially relevant to human psychiatry.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA