Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 153
Filtrar
2.
Medicine (Baltimore) ; 103(12): e37487, 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38518015

RESUMEN

GATA transcriptional factors are zinc finger DNA binding proteins that regulate transcription during development and cell differentiation. The 3 important GATA transcription factors GATA1, GATA2 and GATA3 play essential role in the development and maintenance of hematopoietic systems. GATA1 is required for the erythroid and Megakaryocytic commitment during hematopoiesis. GATA2 is crucial for the proliferation and survival of early hematopoietic cells, and is also involved in lineage specific transcriptional regulation as the dynamic partner of GATA1. GATA3 plays an essential role in T lymphoid cell development and immune regulation. As a result, mutations in gene encoding the GATA transcription factor or alteration in the protein expression level or their function have been linked to a variety of human haematological malignancies. This review presents a summary of recent understanding of how the disrupted biological function of GATA may contribute to hematologic diseases.


Asunto(s)
Factores de Transcripción GATA , Neoplasias Hematológicas , Humanos , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Regulación de la Expresión Génica , Diferenciación Celular , Hematopoyesis/genética , Neoplasias Hematológicas/genética
3.
J Biol Chem ; 299(12): 105419, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37923140

RESUMEN

The Bol2 homolog Fra2 and monothiol glutaredoxin Grx4 together play essential roles in regulating iron homeostasis in Schizosaccharomyces pombe. In vivo studies indicate that Grx4 and Fra2 act as coinhibitory partners that inactivate the transcriptional repressor Fep1 in response to iron deficiency. In Saccharomyces cerevisiae, Bol2 is known to form a [2Fe-2S]-bridged heterodimer with the monothiol Grxs Grx3 and Grx4, with the cluster ligands provided by conserved residues in Grx3/4 and Bol2 as well as GSH. In this study, we characterized this analogous [2Fe-2S]-bridged Grx4-Fra2 complex in S. pombe by identifying the specific residues in Fra2 that act as ligands for the Fe-S cluster and are required to regulate Fep1 activity. We present spectroscopic and biochemical evidence confirming the formation of a [2Fe-2S]-bridged Grx4-Fra2 heterodimer with His66 and Cys29 from Fra2 serving as Fe-S cluster ligands in S. pombe. In vivo transcription and growth assays confirm that both His66 and Cys29 are required to fully mediate the response of Fep1 to low iron conditions. Furthermore, we analyzed the interaction between Fep1 and Grx4-Fra2 using CD spectroscopy to monitor changes in Fe-S cluster coordination chemistry. These experiments demonstrate unidirectional [2Fe-2S] cluster transfer from Fep1 to Grx4-Fra2 in the presence of GSH, revealing the Fe-S cluster dependent mechanism of Fep1 inactivation mediated by Grx4 and Fra2 in response to iron deficiency.


Asunto(s)
Antígeno 2 Relacionado con Fos , Factores de Transcripción GATA , Glutarredoxinas , Homeostasis , Proteínas Hierro-Azufre , Proteínas de Schizosaccharomyces pombe , Schizosaccharomyces , Humanos , Antígeno 2 Relacionado con Fos/genética , Antígeno 2 Relacionado con Fos/metabolismo , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Glutarredoxinas/genética , Glutarredoxinas/metabolismo , Hierro/metabolismo , Proteínas Hierro-Azufre/metabolismo , Oxidorreductasas/metabolismo , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo
4.
Transfus Apher Sci ; 61(6): 103481, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35690555

RESUMEN

BACKGROUND: Umbilical cord blood (UCB) has improved into an attractive and alternative source of allogeneic hematopoietic stem cells (all-HSCs) in clinics and, research for three decades. Recently, it has been shown that the limited cell dose of, this valuable source can be enhanced by the ex vivo expansion of cells in many, ways. We evaluated the expression of the Gata transcription factors family and FOG-1, in expanded and differentiated cord blood-derived CD34 + hematopoietic stem cells to, megakaryocytes lineage., Methods: Separated mononuclear cells were cultured in DMEM complete medium., Harvested cells as a mesenchymal stem cell at 85 % confluency were cultured with, trypsin/EDTA and in 24-well plates. The characteristic analyses of isolated UCB- MSCs, were done by flow cytometry and adipogenic, chondrogenic, and osteogenic, differentiation assays. MACS purified UCB-CD34 + hematopoietic cells cultivated and, differentiated to megakaryocyte progenitor cells in the presence of cytokine cocktail, with UCB-MSCs. Then, the GATA1, GATA2, GATA3, and FOG-1 genes expression, after differentiation to megakaryocyte progenitor cells were performed by quantitative, real-time polymerase chain reaction (PCR)., Results: In this study, the results of real-time-PCR showed that the fold change, expression of GATA-1, FOG-1, and GATA-2 genes after co-culturing with UCB-MSCs, significantly increased to 7.3, 4.7, and 3.3-fold in comparison with control groups;respectively., Conclusion: UCB-MSCs can increase the expansion and differentiation of UCBCD34 + , to megakaryocyte progenitor cells through upregulation of GATA-1, GATA-2, and FOG-1 gene expression.


Asunto(s)
Sangre Fetal , Células Madre Mesenquimatosas , Humanos , Antígenos CD34/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Factores de Transcripción GATA/metabolismo , Células Madre Hematopoyéticas/metabolismo , Megacariocitos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Regulación hacia Arriba
5.
Front Endocrinol (Lausanne) ; 13: 902198, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35692407

RESUMEN

Defining how genes get turned on and off in a correct spatiotemporal manner is integral to our understanding of the development, differentiation, and function of different cell types in both health and disease. Testis development and subsequent male sex differentiation of the XY fetus are well-orchestrated processes that require an intricate network of cell-cell communication and hormonal signals that must be properly interpreted at the genomic level. Transcription factors are at the forefront for translating these signals into a coordinated genomic response. The GATA family of transcriptional regulators were first described as essential regulators of hematopoietic cell differentiation and heart morphogenesis but are now known to impact the development and function of a multitude of tissues and cell types. The mammalian testis is no exception where GATA factors play essential roles in directing the expression of genes crucial not only for testis differentiation but also testis function in the developing male fetus and later in adulthood. This minireview provides an overview of the current state of knowledge of GATA factors in the male gonad with a particular emphasis on their mechanisms of action in the control of testis development, gene expression in the fetal testis, testicular disease, and XY sex differentiation in humans.


Asunto(s)
Diferenciación Sexual , Testículo , Adulto , Animales , Feto/metabolismo , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Expresión Génica , Humanos , Masculino , Mamíferos/genética , Diferenciación Sexual/genética , Testículo/metabolismo , Factores de Transcripción/metabolismo
6.
J Exp Bot ; 73(3): 835-847, 2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-34545936

RESUMEN

BRAHMA (BRM) is the ATPase of the SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodelling complex, which is indispensable for transcriptional inhibition and activation, associated with vegetative and reproductive development in Arabidopsis thaliana. Here, we show that BRM directly binds to the chromatin of SUPPRESSOR OF OVEREXPRESSION OF CONSTANS 1 (SOC1), which integrates multiple flowering signals to regulate floral transition, leading to flowering. In addition, genetic and molecular analysis showed that BRM interacts with GNC (GATA, NITRATE-INDUCIBLE, CARBON METABOLISM INVOLVED), a GATA transcription factor that represses flowering by directly repressing SOC1 expression. Furthermore, BRM is recruited by GNC to directly bind to the chromatin of SOC1. The transcript level of SOC1 is elevated in brm-3, gnc, and brm-3/gnc mutants, which is associated with increased histone H3 lysine 4 tri-methylation (H3K4Me3) but decreased DNA methylation. Taken together, our results indicate that BRM associates with GNC to regulate SOC1 expression and flowering time.


Asunto(s)
Adenosina Trifosfatasas , Proteínas de Arabidopsis , Arabidopsis , Ensamble y Desensamble de Cromatina , Factores de Transcripción , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Arabidopsis/crecimiento & desarrollo , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Regulación de la Expresión Génica de las Plantas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
J Microbiol ; 59(12): 1075-1082, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34705258

RESUMEN

Aconitase, a highly conserved protein across all domains of life, functions in converting citrate to isocitrate in the tricarboxylic acid cycle. Cytosolic aconitase is also known to act as an iron regulatory protein in mammals, binding to the RNA hairpin structures known as iron-responsive elements within the untranslated regions of specific RNAs. Aconitase-2 (Aco2) in fission yeast is a fusion protein consisting of an aconitase and a mitochondrial ribosomal protein, bL21, residing not only in mitochondria but also in cytosol and the nucleus. To investigate the role of Aco2 in the nucleus and cytoplasm of fission yeast, we analyzed the transcriptome of aco2ΔN mutant that is deleted of nuclear localization signal (NLS). RNA sequencing revealed that the aco2ΔN mutation caused increase in mRNAs encoding iron uptake transporters, such as Str1, Str3, and Shu1. The half-lives of mRNAs for these genes were found to be significantly longer in the aco2ΔN mutant than the wild-type strain, suggesting the role of Aco2 in mRNA turnover. The three conserved cysteines required for the catalytic activity of aconitase were not necessary for this role. The UV cross-linking RNA immunoprecipitation analysis revealed that Aco2 directly bound to the mRNAs of iron uptake transporters. Aco2-mediated degradation of iron-uptake mRNAs appears to utilize exoribonuclease pathway that involves Rrp6 as evidenced by genetic interactions. These results reveal a novel role of non-mitochondrial aconitase protein in the mRNA turnover in fission yeast to fine-tune iron homeostasis, independent of regulation by transcriptional repressor Fep1.


Asunto(s)
Aconitato Hidratasa/metabolismo , Proteínas de Transporte de Catión/genética , Regulación Fúngica de la Expresión Génica , Hierro/metabolismo , ARN de Hongos/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/genética , Proteínas de Transporte de Catión/metabolismo , Núcleo Celular/enzimología , Citoplasma/enzimología , Exorribonucleasas/genética , Exorribonucleasas/metabolismo , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Genes Fúngicos , Proteínas Reguladoras del Hierro/genética , Proteínas Reguladoras del Hierro/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Estabilidad del ARN , ARN Mensajero/metabolismo , Regulón , Ribonucleasas/genética , Ribonucleasas/metabolismo , Schizosaccharomyces/enzimología , Proteínas de Schizosaccharomyces pombe/genética
8.
J Cell Biol ; 220(8)2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34047771

RESUMEN

Mesenchymal-to-epithelial transition (MET) converts cells from migratory mesenchymal to polarized epithelial states. Despite its importance for both normal and pathological processes, very little is known about the regulation of MET in vivo. Here we exploit midgut morphogenesis in Drosophila melanogaster to investigate the mechanisms underlying MET. We show that down-regulation of the EMT transcription factor Serpent is required for MET, but not sufficient, as interactions with the surrounding mesoderm are also essential. We find that midgut MET relies on the secretion of specific laminins via the CopII secretory pathway from both mesoderm and midgut cells. We show that secretion of the laminin trimer containing the Wingblister α-subunit from the mesoderm is an upstream cue for midgut MET, leading to basal polarization of αPS1 integrin in midgut cells. Polarized αPS1 is required for the formation of a monolayered columnar epithelium and for the apical polarization of αPS3, Baz, and E-Cad. Secretion of a distinct LamininA-containing trimer from midgut cells is required to reinforce the localization of αPS1 basally, and αPS3 apically, for robust repolarization. Our data suggest that targeting these MET pathways, in conjunction with therapies preventing EMT, may present a two-pronged strategy toward blocking metastasis in cancer.


Asunto(s)
Sistema Digestivo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Laminina/metabolismo , Animales , Animales Modificados Genéticamente , Vesículas Cubiertas por Proteínas de Revestimiento/genética , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Movimiento Celular , Polaridad Celular , Sistema Digestivo/embriología , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Regulación del Desarrollo de la Expresión Génica , Cadenas alfa de Integrinas/genética , Cadenas alfa de Integrinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Laminina/genética , Microscopía Confocal , Microscopía Fluorescente , Transducción de Señal , Factores de Tiempo , Imagen de Lapso de Tiempo
9.
J Microbiol ; 59(5): 449-459, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33877578

RESUMEN

The genome is highly organized hierarchically by the function of structural maintenance of chromosomes (SMC) complex proteins such as condensin and cohesin from bacteria to humans. Although the roles of SMC complex proteins have been well characterized, their specialized roles in nuclear processes remain unclear. Condensin and cohesin have distinct binding sites and mediate long-range and short-range genomic associations, respectively, to form cell cycle-specific genome organization. Condensin can be recruited to highly expressed genes as well as dispersed repeat genetic elements, such as Pol III-transcribed genes, LTR retrotransposon, and rDNA repeat. In particular, mitotic transcription factors Ace2 and Ams2 recruit condensin to their target genes, forming centromeric clustering during mitosis. Condensin is potentially involved in various chromosomal processes such as the mobility of chromosomes, chromosome territories, DNA reannealing, and transcription factories. The current knowledge of condensin in fission yeast summarized in this review can help us understand how condensin mediates genome organization and participates in chromosomal processes in other organisms.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Cromosomas , Proteínas de Unión al ADN/metabolismo , Complejos Multiproteicos/metabolismo , Schizosaccharomyces/fisiología , Proteínas de Ciclo Celular , Proteínas Cromosómicas no Histona , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Humanos , Mitosis , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Factores de Transcripción/genética , Cohesinas
10.
Am J Chin Med ; 49(2): 461-485, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33641653

RESUMEN

Traditional Chinese Medicine (TCM) is a practical medicine based on thousands of years of medical practice in China. Arsenic dispensing powder (ADP) has been used as a treatment for MDS patients with a superior efficacy on anemia at Xiyuan Hospital of China Academy of Chinese Medical Sciences. In this study, we retrospectively analyzed MDS patients that received ADP treatment in the past 9 years and confirmed that ADP improves patients' anemia and prolongs overall survival in intermediate-risk MDS patients. Then, we used the MDS transgenic mice model and cell line to explore the drug mechanism. In normal and MDS cells, ADP does not show cellular toxicity but promotes differentiation. In mouse MDS models, we observed that ADP showed significant efficacy on promoting erythropoiesis. In the BFU-E and CFU-E assays, ADP could promote erythropoiesis not only in normal clones but also in MDS clones. Mechanistically, we found that ADP could downregulate HIF1A in MDS clones through upregulation of VHL, P53 and MDM2, which is involved in two parallel pathways to downregulate HIF1A. We also confirmed that ADP upregulates GATA factors in normal clones. Thus, our clinical and experimental studies indicate that ADP is a promising drug to promote erythropoiesis in both MDS and normal clones with a superior outcome than current regular therapies. ADP promotes erythropoiesis in myelodysplastic syndromes via downregulation of HIF1A and upregulation of GATA factors.


Asunto(s)
Arsenicales/farmacología , Medicamentos Herbarios Chinos/farmacología , Eritropoyesis/efectos de los fármacos , Factores de Transcripción GATA/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Síndromes Mielodisplásicos/tratamiento farmacológico , Animales , Línea Celular , Regulación hacia Abajo , Humanos , Ratones , Polvos , Estudios Retrospectivos , Regulación hacia Arriba
11.
FEBS J ; 288(10): 3231-3245, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33283408

RESUMEN

The multi-subunit nucleosome remodeling and deacetylase (NuRD) complex consists of seven subunits, each of which comprises two or three paralogs in vertebrates. These paralogs define mutually exclusive and functionally distinct complexes. In addition, several proteins in the complex are multimeric, which complicates structural studies. Attempts to purify sufficient amounts of endogenous complex or recombinantly reconstitute the complex for structural studies have proven quite challenging. Until now, only substructures of individual domains or proteins and low-resolution densities of (partial) complexes have been reported. In this study, we comprehensively investigated the relative orientation of different subunits within the NuRD complex using multiple cross-link IP mass spectrometry (xIP-MS) experiments. Our results confirm that the core of the complex is formed by MTA, RBBP, and HDAC proteins. Assembly of a copy of MBD and GATAD2 onto this core enables binding of the peripheral CHD and CDK2AP proteins. Furthermore, our experiments reveal that not only CDK2AP1 but also CDK2AP2 interacts with the NuRD complex. This interaction requires the C terminus of CHD proteins. Our data provide a more detailed understanding of the topology of the peripheral NuRD subunits relative to the core complex. DATABASE: Proteomics data are available in the PRIDE database under the accession numbers PXD017244 and PXD017378.


Asunto(s)
Quinasas Ciclina-Dependientes/química , Factores de Transcripción GATA/química , Histona Desacetilasas/química , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/química , Nucleosomas/ultraestructura , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular Tumoral , Reactivos de Enlaces Cruzados/química , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Células HeLa , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Espectrometría de Masas/métodos , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Modelos Moleculares , Nucleosomas/genética , Nucleosomas/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido
12.
PLoS Genet ; 16(12): e1009286, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33370779

RESUMEN

Developmental-regulatory networks often include large gene families encoding mechanistically-related proteins like G-protein-coupled receptors, zinc finger transcription factors and solute carrier (SLC) transporters. In principle, a common mechanism may confer expression of multiple members integral to a developmental process, or diverse mechanisms may be deployed. Using genetic complementation and enhancer-mutant systems, we analyzed the 456 member SLC family that establishes the small molecule constitution of cells. This analysis identified SLC gene cohorts regulated by GATA1 and/or GATA2 during erythroid differentiation. As >50 SLC genes shared GATA factor regulation, a common mechanism established multiple members of this family. These genes included Slc29a1 encoding an equilibrative nucleoside transporter (Slc29a1/ENT1) that utilizes adenosine as a preferred substrate. Slc29a1 promoted erythroblast survival and differentiation ex vivo. Targeted ablation of murine Slc29a1 in erythroblasts attenuated erythropoiesis and erythrocyte regeneration in response to acute anemia. Our results reveal a GATA factor-regulated SLC ensemble, with a nucleoside transporter component that promotes erythropoiesis and prevents anemia, and establish a mechanistic link between GATA factor and adenosine mechanisms. We propose that integration of the GATA factor-adenosine circuit with other components of the GATA factor-regulated SLC ensemble establishes the small molecule repertoire required for progenitor cells to efficiently generate erythrocytes.


Asunto(s)
Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Eritropoyesis , Factores de Transcripción GATA/metabolismo , Adenosina/metabolismo , Animales , Células Cultivadas , Tranportador Equilibrativo 1 de Nucleósido/genética , Ratones , Ratones Endogámicos C57BL
13.
PLoS Genet ; 16(6): e1008881, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32525871

RESUMEN

Iron is an essential nutrient required as a cofactor for many biological processes. As a fungal commensal-pathogen of humans, Candida albicans encounters a range of bioavailable iron levels in the human host and maintains homeostasis with a conserved regulatory circuit. How C. albicans senses and responds to iron availability is unknown. In model yeasts, regulation of the iron homeostasis circuit requires monothiol glutaredoxins (Grxs), but their functions beyond the regulatory circuit are unclear. Here, we show Grx3 is required for virulence and growth on low iron for C. albicans. To explore the global roles of Grx3, we applied a proteomic approach and performed in vivo cross-linked tandem affinity purification coupled with mass spectrometry. We identified a large number of Grx3 interacting proteins that function in diverse biological processes. This included Fra1 and Bol2/Fra2, which function with Grxs in intracellular iron trafficking in other organisms. Grx3 interacts with and regulates the activity of Sfu1 and Hap43, components of the C. albicans iron regulatory circuit. Unlike the regulatory circuit, which determines expression or repression of target genes in response to iron availability, Grx3 amplifies levels of gene expression or repression. Consistent with the proteomic data, the grx3 mutant is sensitive to heat shock, oxidative, nitrosative, and genotoxic stresses, and shows growth dependence on histidine, leucine, and tryptophan. We suggest Grx3 is a conserved global regulator of iron-dependent processes occurring within the cell.


Asunto(s)
Candida albicans/fisiología , Candidiasis Invasiva/microbiología , Proteínas Fúngicas/metabolismo , Glutarredoxinas/metabolismo , Hierro/metabolismo , Animales , Candida albicans/patogenicidad , Modelos Animales de Enfermedad , Proteínas Fúngicas/genética , Proteínas Fúngicas/aislamiento & purificación , Factores de Transcripción GATA/metabolismo , Regulación Fúngica de la Expresión Génica , Glutarredoxinas/genética , Glutarredoxinas/aislamiento & purificación , Homeostasis , Humanos , Hifa , Masculino , Ratones , Mutación , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas/genética , Proteómica , Virulencia/genética
14.
J Integr Plant Biol ; 62(8): 1112-1131, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32022417

RESUMEN

Self-incompatibility (SI) is a genetic mechanism that rejects self-pollen and thus prevents inbreeding in some hermaphroditic angiosperms. In the Brassicaceae, SI involves a pollen-stigma recognition system controlled by a single locus known as the S locus, which consists of two highly polymorphic genes that encode S-locus cysteine-rich protein (SCR) and S-receptor kinase (SRK). When self-pollen lands on the stigma, the S-haplotype-specific interaction between SCR and SRK triggers SI. Here, we show that the GATA transcription factor BnA5.ZML1 suppresses SI responses in Brassica napus and is induced after compatible pollination. The loss-of-function mutant bna5.zml1 displays reduced self-compatibility. In contrast, overexpression of BnA5.ZML1 in self-incompatible stigmas leads to a partial breakdown of SI responses, suggesting that BnA5.ZML1 is a stigmatic compatibility factor. Furthermore, the expression levels of SRK and ARC1 are up-regulated in bna5.zml1 mutants, and they are down-regulated in BnA5.ZML1 overexpressing lines. SRK affects the cellular localization of BnA5.ZML1 through direct protein-protein interaction. Overall, our findings highlight the fundamental role of BnA5.ZML1 in SI responses in B. napus, establishing a direct interaction between BnA5.ZML1 and SRK in this process.


Asunto(s)
Brassica napus/metabolismo , Flores/metabolismo , Factores de Transcripción GATA/metabolismo , Proteínas de Plantas/metabolismo , Brassica napus/genética , Flores/genética , Factores de Transcripción GATA/genética , Regulación de la Expresión Génica de las Plantas , Genotipo , Modelos Biológicos , Mutación/genética , Proteínas Nucleares/metabolismo , Filogenia , Proteínas de Plantas/genética , Polinización , Unión Proteica , Autoincompatibilidad en las Plantas con Flores/genética , Activación Transcripcional/genética
16.
Development ; 147(1)2020 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-31806659

RESUMEN

The GATA and PAX-SIX-EYA-DACH transcriptional networks (PSEDNs) are essential for proper development across taxa. Here, we demonstrate novel PSEDN roles in vivo in Drosophila hematopoiesis and in human erythropoiesis in vitro Using Drosophila genetics, we show that PSEDN members function with GATA to block lamellocyte differentiation and maintain the prohemocyte pool. Overexpression of human SIX1 stimulated erythroid differentiation of human erythroleukemia TF1 cells and primary hematopoietic stem-progenitor cells. Conversely, SIX1 knockout impaired erythropoiesis in both cell types. SIX1 stimulation of erythropoiesis required GATA1, as SIX1 overexpression failed to drive erythroid phenotypes and gene expression patterns in GATA1 knockout cells. SIX1 can associate with GATA1 and stimulate GATA1-mediated gene transcription, suggesting that SIX1-GATA1 physical interactions contribute to the observed functional interactions. In addition, both fly and human SIX proteins regulated GATA protein levels. Collectively, our findings demonstrate that SIX proteins enhance GATA function at multiple levels, and reveal evolutionarily conserved cooperation between the GATA and PSEDN networks that may regulate developmental processes beyond hematopoiesis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Eritropoyesis/genética , Redes Reguladoras de Genes , Hematopoyesis/genética , Animales , Línea Celular Tumoral , Drosophila , Factores de Transcripción GATA/metabolismo , Técnicas de Inactivación de Genes , Proteínas de Homeodominio/metabolismo , Humanos , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción Paired Box/metabolismo
17.
IUBMB Life ; 72(1): 27-38, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31769130

RESUMEN

Members of the GATA family of transcription factors play key roles in the differentiation of specific cell lineages by regulating the expression of target genes. Three GATA factors play distinct roles in hematopoietic differentiation. In order to better understand how these GATA factors function to regulate genes throughout the genome, we are studying the epigenomic and transcriptional landscapes of hematopoietic cells in a model-driven, integrative fashion. We have formed the collaborative multi-lab VISION project to conduct ValIdated Systematic IntegratiON of epigenomic data in mouse and human hematopoiesis. The epigenomic data included nuclease accessibility in chromatin, CTCF occupancy, and histone H3 modifications for 20 cell types covering hematopoietic stem cells, multilineage progenitor cells, and mature cells across the blood cell lineages of mouse. The analysis used the Integrative and Discriminative Epigenome Annotation System (IDEAS), which learns all common combinations of features (epigenetic states) simultaneously in two dimensions-along chromosomes and across cell types. The result is a segmentation that effectively paints the regulatory landscape in readily interpretable views, revealing constitutively active or silent loci as well as the loci specifically induced or repressed in each stage and lineage. Nuclease accessible DNA segments in active chromatin states were designated candidate cis-regulatory elements in each cell type, providing one of the most comprehensive registries of candidate hematopoietic regulatory elements to date. Applications of VISION resources are illustrated for the regulation of genes encoding GATA1, GATA2, GATA3, and Ikaros. VISION resources are freely available from our website http://usevision.org.


Asunto(s)
Cromatina/metabolismo , Epigenoma , Factores de Transcripción GATA/metabolismo , Regulación de la Expresión Génica , Hematopoyesis , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Diferenciación Celular , Cromatina/genética , Factores de Transcripción GATA/genética , Humanos
18.
IUBMB Life ; 72(1): 53-67, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31520462

RESUMEN

Cardiac development is governed by a complex network of transcription factors (TFs) that regulate cell fates in a spatiotemporal manner. Among these, the GATA family of zinc finger TFs plays prominent roles in regulating the development of the myocardium, endocardium, and outflow tract. This family comprises six members three of which, GATA4, 5, and 6, are predominantly expressed in cardiac cells where they activate specific downstream gene targets via interactions with one another and with other TFs and signaling molecules. Their critical function in heart formation is evidenced by the phenotypes of animal models lacking these factors and by the broad spectrum of human congenital heart diseases associated with mutations in their genes. Similarly, in the postnatal heart, these proteins play significant and nonredundant roles in cardiac function, regulating adaptive stress responses including cardiomyocyte hypertrophy and survival, as well as endothelial homeostasis and angiogenesis. As such, decreased expression of either GATA4, 5, or 6 results in impaired cardiovascular homeostasis and increased risk of premature and serious cardiovascular events such as hypertension, arrhythmia, aortopathy, and heart failure. Although a great deal of progress has been made in understanding GATA-dependent regulatory processes in the heart, the molecular mechanisms underlying the specificity of GATA factors and their upstream regulation remain incompletely understood. The knowledge and tools developed since their discovery 25 years ago should accelerate progress toward further elucidation of their mechanisms of action in health and disease. This in turn will greatly improve diagnosis and care for the millions of individuals affected by congenital and acquired cardiac disease worldwide.


Asunto(s)
Desarrollo Embrionario , Factores de Transcripción GATA/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Organogénesis , Animales , Factores de Transcripción GATA/genética , Humanos , Transducción de Señal
19.
IUBMB Life ; 72(1): 10-26, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31574210

RESUMEN

The members of the GATA family of transcription factors have homologous zinc fingers and bind to similar sequence motifs. Recent advances in genome-wide technologies and the integration of bioinformatics data have led to a better understanding of how GATA factors regulate gene expression; GATA-factor-induced transcriptional and epigenetic changes have now been analyzed at unprecedented levels of detail. Here, we review the results of genome-wide studies of GATA factor occupancy in human and murine cell lines and primary cells (as determined by chromatin immunoprecipitation sequencing), and then discuss the molecular mechanisms underlying the mediation of transcriptional and epigenetic regulation by GATA factors.


Asunto(s)
Epigénesis Genética , Factores de Transcripción GATA/metabolismo , Regulación Neoplásica de la Expresión Génica , Genoma , Neoplasias/patología , Animales , Biología Computacional , Factores de Transcripción GATA/genética , Humanos , Neoplasias/genética , Neoplasias/metabolismo
20.
IUBMB Life ; 72(1): 80-88, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31580534

RESUMEN

There is an urgent need for the development of novel therapeutics options for diabetic patients given the high prevalence of diabetes worldwide and that, currently, there is no cure for this disease. The transplantation of pancreatic islets that contain insulin-producing cells is a promising therapeutic alternative, particularly for type 1 diabetes. However, the shortage of organ donors constitutes a major limitation for this approach; thus, developing alternative sources of insulin-producing cells is of critical importance. In the last decade, our knowledge of the molecular mechanisms controlling embryonic pancreas development has significantly advanced. More importantly, this knowledge has provided the basis for the in vitro generation of insulin-producing cells from stem cells. Recent studies have revealed that GATA transcription factors are involved in various stages of pancreas formation and in the adult ß cell function. Here, we review the fundamental role of GATA transcription factors in pancreas morphogenesis and their association with congenital diseases associated with pancreas.


Asunto(s)
Factores de Transcripción GATA/metabolismo , Regulación del Desarrollo de la Expresión Génica , Páncreas/embriología , Enfermedades Pancreáticas/patología , Animales , Factores de Transcripción GATA/genética , Humanos , Páncreas/metabolismo , Enfermedades Pancreáticas/genética , Enfermedades Pancreáticas/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA