Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Cancers (Basel) ; 14(19)2022 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-36230673

RESUMO

The survival of patients with solid tumors, such as prostate cancer (PCa), has been limited and fleeting with anti-angiogenic therapies. It was previously thought that the mechanism by which the vasculature regulates tumor growth was driven by a passive movement of oxygen and nutrients to the tumor tissue. However, previous evidence suggests that endothelial cells have an alternative role in changing the behavior of tumor cells and contributing to cancer progression. Determining the impact of molecular signals/growth factors released by endothelial cells (ECs) on established PCa cell lines in vitro and in vivo could help to explain the mechanism by which ECs regulate tumor growth. Using cell-conditioned media collected from HUVEC (HUVEC-CM), our data show the stimulated proliferation of all the PCa cell lines tested. However, in more aggressive PCa cell lines, HUVEC-CM selectively promoted migration and invasion in vitro and in vivo. Using a PCa-cell-line-derived xenograft model co-injected with HUVEC or preincubated with HUVEC-CM, our results are consistent with the in vitro data, showing enhanced tumor growth, increased tumor microvasculature and promoted metastasis. Gene set enrichment analyses from RNA-Seq gene expression profiles showed that HUVEC-CM induced a differential effect on gene expression when comparing low versus highly aggressive PCa cell lines, demonstrating epigenetic and migratory pathway enrichments in highly aggressive PCa cells. In summary, paracrine stimulation by HUVEC increased PCa cell proliferation and tumor growth and selectively promoted migration and metastatic potential in more aggressive PCa cell lines.

2.
Development ; 149(7)2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35297968

RESUMO

Vascular networks comprise endothelial cells and mural cells, which include pericytes and smooth muscle cells. To elucidate the mechanisms controlling mural cell recruitment during development and tissue regeneration, we studied zebrafish caudal fin arteries. Mural cells colonizing arteries proximal to the body wrapped around them, whereas those in more distal regions extended protrusions along the proximo-distal vascular axis. Both cell populations expressed platelet-derived growth factor receptor ß (pdgfrb) and the smooth muscle cell marker myosin heavy chain 11a (myh11a). Most wrapping cells in proximal locations additionally expressed actin alpha2, smooth muscle (acta2). Loss of Pdgfrb signalling specifically decreased mural cell numbers at the vascular front. Using lineage tracing, we demonstrate that precursor cells located in periarterial regions and expressing Pgdfrb can give rise to mural cells. Studying tissue regeneration, we did not find evidence that newly formed mural cells were derived from pre-existing cells. Together, our findings reveal conserved roles for Pdgfrb signalling in development and regeneration, and suggest a limited capacity of mural cells to self-renew or contribute to other cell types during tissue regeneration.


Assuntos
Miócitos de Músculo Liso , Pericitos , Receptor beta de Fator de Crescimento Derivado de Plaquetas , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Células Endoteliais/metabolismo , Miócitos de Músculo Liso/metabolismo , Pericitos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
3.
Int J Mol Sci ; 21(24)2020 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-33322837

RESUMO

Reprimo-like (RPRML) is an uncharacterized member of the Reprimo gene family. Here, we evaluated the role of RPRML and whether its regulation by DNA methylation is a potential non-invasive biomarker of gastric cancer. RPRML expression was evaluated by immunohistochemistry in 90 patients with gastric cancer and associated with clinicopathologic characteristics and outcomes. The role of RPRML in cancer biology was investigated in vitro, through RPRML ectopic overexpression. Functional experiments included colony formation, soft agar, MTS, and Ki67 immunofluorescence assays. DNA methylation-mediated silencing was evaluated by the 5-azacytidine assay and direct bisulfite sequencing. Non-invasive detection of circulating methylated RPRML DNA was assessed in 25 gastric cancer cases and 25 age- and sex-balanced cancer-free controls by the MethyLight assay. Downregulation of RPRML protein expression was associated with poor overall survival in advanced gastric cancer. RPRML overexpression significantly inhibited clonogenic capacity, anchorage-independent growth, and proliferation in vitro. Circulating methylated RPRML DNA distinguished patients with gastric cancer from controls with an area under the curve of 0.726. The in vitro overexpression results and the poor patient survival associated with lower RPRML levels suggest that RPRML plays a tumor-suppressive role in the stomach. Circulating methylated RPRML DNA may serve as a biomarker for the non-invasive detection of gastric cancer.


Assuntos
Biomarcadores Tumorais/genética , Ácidos Nucleicos Livres/sangue , Metilação de DNA , Genes Supressores de Tumor , Proteínas de Membrana/sangue , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/metabolismo , Idoso , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Epigênese Genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Inativação Gênica , Humanos , Imuno-Histoquímica , Masculino , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Estudos Retrospectivos , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidade , Regulação para Cima
4.
Sci Rep ; 10(1): 4068, 2020 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-32111873

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Sci Rep ; 9(1): 7131, 2019 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-31073223

RESUMO

The Reprimo gene family comprises a group of single-exon genes for which their physiological function remains poorly understood. Heretofore, mammalian Reprimo (RPRM) has been described as a putative p53-dependent tumor suppressor gene that functions at the G2/M cell cycle checkpoint. Another family member, Reprimo-like (RPRML), has not yet an established role in physiology or pathology. Importantly, RPRML expression pattern is conserved between zebrafish and human species. Here, using CRISPR-Cas9 and antisense morpholino oligonucleotides, we disrupt the expression of rprml in zebrafish and demonstrate that its loss leads to impaired definitive hematopoiesis. The formation of hemangioblasts and the primitive wave of hematopoiesis occur normally in absence of rprml. Later in development there is a significant reduction in erythroid-myeloid precursors (EMP) at the posterior blood island (PBI) and a significant decline of definitive hematopoietic stem/progenitor cells (HSPCs). Furthermore, loss of rprml also increases the activity of caspase-3 in endothelial cells within the caudal hematopoietic tissue (CHT), the first perivascular niche where HSPCs reside during zebrafish embryonic development. Herein, we report an essential role for rprml during hematovascular development in zebrafish embryos, specifically during the definitive waves of hematopoiesis, indicating for the first time a physiological role for the rprml gene.


Assuntos
Hemangioblastos/metabolismo , Proteínas de Membrana/genética , Peixe-Zebra/embriologia , Animais , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/genética , Desenvolvimento Embrionário , Hematopoese , Morfolinos/farmacologia , Família Multigênica , Peixe-Zebra/sangue , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
6.
Int J Mol Sci ; 19(7)2018 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-29941787

RESUMO

The reprimo (RPRM) gene family is a group of single exon genes present exclusively within the vertebrate lineage. Two out of three members of this family are present in humans: RPRM and RPRM-Like (RPRML). RPRM induces cell cycle arrest at G2/M in response to p53 expression. Loss-of-expression of RPRM is related to increased cell proliferation and growth in gastric cancer. This evidence suggests that RPRM has tumor suppressive properties. However, the molecular mechanisms and signaling partners by which RPRM exerts its functions remain unknown. Moreover, scarce studies have attempted to characterize RPRML, and its functionality is unclear. Herein, we highlight the role of the RPRM gene family in gastric carcinogenesis, as well as its potential applications in clinical settings. In addition, we summarize the current knowledge on the phylogeny and expression patterns of this family of genes in embryonic zebrafish and adult humans. Strikingly, in both species, RPRM is expressed primarily in the digestive tract, blood vessels and central nervous system, supporting the use of zebrafish for further functional characterization of RPRM. Finally, drawing on embryonic and adult expression patterns, we address the potential relevance of RPRM and RPRML in cancer. Active investigation or analytical research in the coming years should contribute to novel translational applications of this poorly understood gene family as potential biomarkers and development of novel cancer therapies.


Assuntos
Proteínas de Ciclo Celular/genética , Metilação de DNA/genética , Glicoproteínas/genética , Proteínas de Membrana/genética , Neoplasias Gástricas/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , Regiões Promotoras Genéticas , Neoplasias Gástricas/patologia
7.
Front Neuroanat ; 12: 23, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29636669

RESUMO

The Reprimo (RPRM) family is composed of highly conserved single-exon genes. The expression pattern of this gene family has been recently described during zebrafish (Danio rerio) embryogenesis, and primarily locates in the nervous system. Its most characterized member, RPRM, which duplicated to give rise rprma and rprmb in the fish lineage, is known to act as a tumor-suppressor gene in mammalian models. Here, we describe in detail the spatiotemporal expression of three rprm genes (rprma, rprmb, and rprml) within distinct anatomical structures in the developing peripheral and central nervous system. In the zebrafish, rprma mRNA is expressed in the olfactory placodes (OP) and olfactory epithelium (OE), rprmb is observed in the tectum opticum (TeO) and trigeminal ganglion (Tg), whereas rprml is found primarily in the telencephalon (Tel). At protein level, RPRM is present in a subset of cells in the OP, and neurons in the OE, TeO, hindbrain and sensory peripheral structures. Most importantly, the expression of RPRM has been conserved between teleosts and mammals. Thus, we provide a reference dataset describing the expression patterns of RPRM gene products during zebrafish and mouse development as a first step to approach the physiological role of the RPRM gene family.

8.
PLoS One ; 12(5): e0178274, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28562620

RESUMO

Reprimo (RPRM), a member of the RPRM gene family, is a tumor-suppressor gene involved in the regulation of the p53-mediated cell cycle arrest at G2/M. RPRM has been associated with malignant tumor progression and proposed as a potential biomarker for early cancer detection. However, the expression and role of RPRM, as well as its family, are poorly understood and their physiology is as yet unstudied. In this scenario, a model system like the zebrafish could serve to dissect the role of the RPRM family members in vivo. Phylogenetic analysis reveals that RPRM and RPRML have been differentially retained by most species throughout vertebrate evolution, yet RPRM3 has been retained only in a small group of distantly related species, including zebrafish. Herein, we characterized the spatiotemporal expression of RPRM (present in zebrafish as an infraclass duplication rprma/rprmb), RPRML and RPRM3 in the zebrafish. By whole-mount in situ hybridization (WISH) and fluorescent in situ hybridization (FISH), we demonstrate that rprm (rprma/rprmb) and rprml show a similar spatiotemporal expression profile during zebrafish development. At early developmental stages rprmb is expressed in somites. After one day post-fertilization, rprm (rprma/rprmb) and rprml are expressed in the notochord, brain, blood vessels and digestive tube. On the other hand, rprm3 shows the most unique expression profile, being expressed only in the central nervous system (CNS). We assessed the expression patterns of RPRM gene transcripts in adult zebrafish and human RPRM protein product in tissue samples by RT-qPCR and immunohistochemistry (IHC) staining, respectively. Strikingly, tissue-specific expression patterns of the RPRM transcripts and protein are conserved between zebrafish and humans. We propose the zebrafish as a powerful tool to elucidate the both physiological and pathological roles of the RPRM gene family.


Assuntos
Proteínas de Ciclo Celular/genética , Regulação da Expressão Gênica , Glicoproteínas/genética , Sequência de Aminoácidos , Animais , Sequência Conservada , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hibridização In Situ , Hibridização in Situ Fluorescente , Homologia de Sequência de Aminoácidos , Peixe-Zebra/embriologia
9.
Gene ; 591(1): 245-254, 2016 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-27432065

RESUMO

Genes related to human diseases should be natural targets for evolutionary studies, since they could provide clues regarding the genetic bases of pathologies and potential treatments. Here we studied the evolution of the reprimo gene family, a group of tumor-suppressor genes that are implicated in p53-mediated cell cycle arrest. These genes, especially the reprimo duplicate located on human chromosome 2, have been associated with epigenetic modifications correlated with transcriptional silencing and cancer progression. We demonstrate the presence of a third reprimo lineage that, together with the reprimo and reprimo-like genes, appears to have been differentially retained during the evolutionary history of vertebrates. We present evidence that these reprimo lineages originated early in vertebrate evolution and expanded as a result of the two rounds of whole genome duplications that occurred in the last common ancestor of vertebrates. The reprimo gene has been lost in birds, and the third reprimo gene lineage has been retained in only a few distantly related species, such as coelacanth and gar. Expression analyses revealed that the reprimo paralogs are mainly expressed in the nervous system. Different vertebrate lineages have retained different reprimo paralogs, and even in species that have retained multiple copies, only one of them is heavily expressed.


Assuntos
Proteínas de Ciclo Celular/genética , Evolução Molecular , Genes Supressores de Tumor , Família Multigênica , Filogenia , Vertebrados/genética , Sequência de Aminoácidos , Animais , Duplicação Gênica , Humanos , Funções Verossimilhança , Alinhamento de Sequência , Sintenia/genética , Transcrição Gênica , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/genética
10.
Mol Cell Biol ; 31(7): 1344-56, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21248200

RESUMO

Mitoferrin 1 (Mfrn1; Slc25a37) and mitoferrin 2 (Mfrn2; Slc25a28) function as essential mitochondrial iron importers for heme and Fe/S cluster biogenesis. A genetic deficiency of Mfrn1 results in a profound hypochromic anemia in vertebrate species. To map the cis-regulatory modules (CRMs) that control expression of the Mfrn genes, we utilized genome-wide chromatin immunoprecipitation (ChIP) datasets for the major erythroid transcription factor GATA-1. We identified the CRMs that faithfully drive the expression of Mfrn1 during blood and heart development and Mfrn2 ubiquitously. Through in vivo analyses of the Mfrn-CRMs in zebrafish and mouse, we demonstrate their functional and evolutionary conservation. Using knockdowns with morpholinos and cell sorting analysis in transgenic zebrafish embryos, we show that GATA-1 directly regulates the expression of Mfrn1. Mutagenesis of individual GATA-1 binding cis elements (GBE) demonstrated that at least two of the three GBE within this CRM are functionally required for GATA-mediated transcription of Mfrn1. Furthermore, ChIP assays demonstrate switching from GATA-2 to GATA-1 at these elements during erythroid maturation. Our results provide new insights into the genetic regulation of mitochondrial function and iron homeostasis and, more generally, illustrate the utility of genome-wide ChIP analysis combined with zebrafish transgenesis for identifying long-range transcriptional enhancers that regulate tissue development.


Assuntos
Técnicas de Transferência de Genes , Loci Gênicos/genética , Proteínas de Membrana Transportadoras/genética , Regiões Promotoras Genéticas/genética , Peixe-Zebra/genética , Animais , Pareamento de Bases/genética , Sequência de Bases , Sítios de Ligação , Proteínas de Transporte de Cátions , Imunoprecipitação da Cromatina , Elementos Facilitadores Genéticos/genética , Eritropoese/genética , Fator de Transcrição GATA1/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Genoma/genética , Proteínas de Fluorescência Verde/metabolismo , Coração/embriologia , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteínas de Peixe-Zebra/metabolismo
11.
Genes Dev ; 24(15): 1620-33, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20679398

RESUMO

The bicistronic microRNA (miRNA) locus miR-144/451 is highly expressed during erythrocyte development, although its physiological roles are poorly understood. We show that miR-144/451 ablation in mice causes mild erythrocyte instability and increased susceptibility to damage after exposure to oxidant drugs. This phenotype is deeply conserved, as miR-451 depletion synergizes with oxidant stress to cause profound anemia in zebrafish embryos. At least some protective activities of miR-451 stem from its ability to directly suppress production of 14-3-3zeta, a phospho-serine/threonine-binding protein that inhibits nuclear accumulation of transcription factor FoxO3, a positive regulator of erythroid anti-oxidant genes. Thus, in miR-144/451(-/-) erythroblasts, 14-3-3zeta accumulates, causing partial relocalization of FoxO3 from nucleus to cytoplasm with dampening of its transcriptional program, including anti-oxidant-encoding genes Cat and Gpx1. Supporting this mechanism, overexpression of 14-3-3zeta in erythroid cells and fibroblasts inhibits nuclear localization and activity of FoxO3. Moreover, shRNA suppression of 14-3-3zeta protects miR-144/451(-/-) erythrocytes against peroxide-induced destruction, and restores catalase activity. Our findings define a novel miRNA-regulated pathway that protects erythrocytes against oxidant stress, and, more generally, illustrate how a miRNA can influence gene expression by altering the activity of a key transcription factor.


Assuntos
Proteínas 14-3-3/metabolismo , Células Eritroides/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/metabolismo , Estresse Oxidativo , Proteínas 14-3-3/genética , Transporte Ativo do Núcleo Celular , Animais , Sequência de Bases , Catalase/metabolismo , Células Eritroides/enzimologia , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Knockout , MicroRNAs/genética , Alinhamento de Sequência , Deleção de Sequência/genética , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
12.
Blood ; 114(21): 4654-63, 2009 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-19729519

RESUMO

The nuclear protein FOG-1 binds transcription factor GATA-1 to facilitate erythroid and megakaryocytic maturation. However, little is known about the function of FOG-1 during myeloid and lymphoid development or how FOG-1 expression is regulated in any tissue. We used in situ hybridization, gain- and loss-of-function studies in zebrafish to address these problems. Zebrafish FOG-1 is expressed in early hematopoietic cells, as well as heart, viscera, and paraspinal neurons, suggesting that it has multifaceted functions in organogenesis. We found that FOG-1 is dispensable for endoderm specification but is required for endoderm patterning affecting the expression of late-stage T-cell markers, independent of GATA-1. The suppression of FOG-1, in the presence of normal GATA-1 levels, induces severe anemia and thrombocytopenia and expands myeloid-progenitor cells, indicating that FOG-1 is required during erythroid/myeloid commitment. To functionally interrogate whether GATA-1 regulates FOG-1 in vivo, we used bioinformatics combined with transgenic assays. Thus, we identified 2 cis-regulatory elements that control the tissue-specific gene expression of FOG-1. One of these enhancers contains functional GATA-binding sites, indicating the potential for a regulatory loop in which GATA factors control the expression of their partner protein FOG-1.


Assuntos
Desenvolvimento Embrionário/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Nucleares , Proteínas de Peixe-Zebra , Peixe-Zebra/embriologia , Animais , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Hematopoese/fisiologia , Hibridização In Situ , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Elementos Reguladores de Transcrição/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
13.
FASEB J ; 22(10): 3617-27, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18591368

RESUMO

Niemann-Pick type C (NPC) disease is a fatal autosomal recessive disorder characterized by the accumulation of free cholesterol and glycosphingolipids in the endosomal-lysosomal system. Patients with NPC disease have markedly progressive neuronal loss, mainly of cerebellar Purkinje neurons. There is strong evidence indicating that cholesterol accumulation and trafficking defects activate apoptosis in NPC brains. The purpose of this study was to analyze the relevance of apoptosis and particularly the proapoptotic c-Abl/p73 system in cerebellar neuron degeneration in NPC disease. We used the NPC1 mouse model to evaluate c-Abl/p73 expression and activation in the cerebellum and the effect of therapy with the c-Abl-specific inhibitor imatinib. The proapoptotic c-Abl/p73 system and the p73 target genes are expressed in the cerebellums of NPC mice. Furthermore, inhibition of c-Abl with imatinib preserved Purkinje neurons and reduced general cell apoptosis in the cerebellum, improved neurological symptoms, and increased the survival of NPC mice. Moreover, this prosurvival effect correlated with reduced mRNA levels of p73 proapoptotic target genes. Our results suggest that the c-Abl/p73 pathway is involved in NPC neurodegeneration and show that treatment with c-Abl inhibitors is useful in delaying progressive neurodegeneration, supporting the use of imatinib for clinical treatment of patients with NPC disease.


Assuntos
Apoptose/efeitos dos fármacos , Córtex Cerebelar/efeitos dos fármacos , Doença de Niemann-Pick Tipo C/tratamento farmacológico , Piperazinas/uso terapêutico , Pirimidinas/uso terapêutico , Animais , Benzamidas , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebelar/metabolismo , Córtex Cerebelar/patologia , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Mesilato de Imatinib , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Proteína C1 de Niemann-Pick , Doença de Niemann-Pick Tipo C/metabolismo , Doença de Niemann-Pick Tipo C/patologia , Proteínas Nucleares/metabolismo , Proteínas/genética , Proteínas Proto-Oncogênicas c-abl/metabolismo , Células de Purkinje/efeitos dos fármacos , Células de Purkinje/patologia , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , Proteína Tumoral p73 , Proteínas Supressoras de Tumor/metabolismo
14.
Nature ; 453(7198): 1127-31, 2008 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-18418376

RESUMO

Haems are metalloporphyrins that serve as prosthetic groups for various biological processes including respiration, gas sensing, xenobiotic detoxification, cell differentiation, circadian clock control, metabolic reprogramming and microRNA processing. With a few exceptions, haem is synthesized by a multistep biosynthetic pathway comprising defined intermediates that are highly conserved throughout evolution. Despite our extensive knowledge of haem biosynthesis and degradation, the cellular pathways and molecules that mediate intracellular haem trafficking are unknown. The experimental setback in identifying haem trafficking pathways has been the inability to dissociate the highly regulated cellular synthesis and degradation of haem from intracellular trafficking events. Caenorhabditis elegans and related helminths are natural haem auxotrophs that acquire environmental haem for incorporation into haemoproteins, which have vertebrate orthologues. Here we show, by exploiting this auxotrophy to identify HRG-1 proteins in C. elegans, that these proteins are essential for haem homeostasis and normal development in worms and vertebrates. Depletion of hrg-1, or its paralogue hrg-4, in worms results in the disruption of organismal haem sensing and an abnormal response to haem analogues. HRG-1 and HRG-4 are previously unknown transmembrane proteins, which reside in distinct intracellular compartments. Transient knockdown of hrg-1 in zebrafish leads to hydrocephalus, yolk tube malformations and, most strikingly, profound defects in erythropoiesis-phenotypes that are fully rescued by worm HRG-1. Human and worm proteins localize together, and bind and transport haem, thus establishing an evolutionarily conserved function for HRG-1. These findings reveal conserved pathways for cellular haem trafficking in animals that define the model for eukaryotic haem transport. Thus, uncovering the mechanisms of haem transport in C. elegans may provide insights into human disorders of haem metabolism and reveal new drug targets for developing anthelminthics to combat worm infestations.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Heme/metabolismo , Hemeproteínas/metabolismo , Homeostase , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Linhagem Celular , Eritropoese , Heme/farmacologia , Hemeproteínas/genética , Humanos , Metaloporfirinas/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
15.
Proc Natl Acad Sci U S A ; 105(9): 3333-8, 2008 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-18303114

RESUMO

MicroRNAs (miRNAs) control tissue development, but their mechanism of regulation is not well understood. We used a gene complementation strategy combined with microarray screening to identify miRNAs involved in the formation of erythroid (red blood) cells. Two conserved miRNAs, miR 144 and miR 451, emerged as direct targets of the critical hematopoietic transcription factor GATA-1. In vivo, GATA-1 binds a distal upstream regulatory element to activate RNA polymerase II-mediated transcription of a single common precursor RNA (pri-miRNA) encoding both mature miRNAs. Zebrafish embryos depleted of miR 451 by using antisense morpholinos form erythroid precursors, but their development into mature circulating red blood cells is strongly and specifically impaired. These results reveal a miRNA locus that is required for erythropoiesis and uncover a new regulatory axis through which GATA-1 controls this process.


Assuntos
Células Precursoras Eritroides/citologia , Eritropoese/genética , Fator de Transcrição GATA1/fisiologia , MicroRNAs/fisiologia , Animais , Linhagem Celular Tumoral , Fatores de Ligação de DNA Eritroide Específicos , Hibridização In Situ , Camundongos , MicroRNAs/análise , Análise em Microsséries , Peixe-Zebra
16.
Am J Physiol Regul Integr Comp Physiol ; 294(3): R1081-91, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18046018

RESUMO

The genome of zebrafish (Danio rerio) encodes two unlinked genes equally closely related to the SLC4A2/AE2 anion exchanger genes of mammals. One of these is the recently reported zebrafish ae2 gene (Shmukler BE, Kurschat CE, Ackermann GE, Jiang L, Zhou Y, Barut B, Stuart-Tilley AK, Zhao J, Zon LI, Drummond IA, Vandorpe DH, Paw BH, Alper SL. Am J Physiol Renal Physiol Renal Physiol 289: F835-F849, 2005), now called ae2.1. We now report the structural and functional characterization of Ae2.2, the product of the second zebrafish Ae2 gene, ae2.2. The ae2.2 gene of zebrafish linkage group 24 encodes a polypeptide of 1,232 aa in length, sharing 70% amino acid identity with zebrafish Ae2.1 and 67% identity with mouse AE2a. Zebrafish Ae2.2 expressed in Xenopus oocytes encodes a 135-kDa polypeptide that mediates bidirectional, DIDS-sensitive Cl(-)/Cl(-) exchange and Cl(-)/HCO3(-) exchange. Ae2.2-mediated Cl(-)/Cl(-) exchange is cation independent, voltage insensitive, and electroneutral. Acute regulation of anion exchange mediated by Ae2.2 includes activation by NH4+ and independent inhibition by acidic intracellular pH and by acidic extracellular pH. In situ hybridization reveals low-level expression of Ae2.2 mRNA in zebrafish embryo, most notably in posterior tectum, eye, pharynx, epidermal cells, and axial vascular structures, without notable expression in the Ae2.1-expressing pronephric duct. Knockdown of Ae2.2 mRNA, of Ae2.1 mRNA, or of both with nontoxic or minimally toxic levels of N-morpholino oligomers produced no grossly detectable morphological phenotype, and preserved normal structure of the head and the pronephric duct at 24 h postfertilization.


Assuntos
Proteínas de Transporte de Ânions/genética , Antiporters/genética , Peixe-Zebra/fisiologia , Sequência de Aminoácidos , Animais , Antiportadores de Cloreto-Bicarbonato , Cloretos/metabolismo , Clonagem Molecular , DNA Complementar/biossíntese , DNA Complementar/genética , Embrião não Mamífero , Imunofluorescência , Potenciais da Membrana/efeitos dos fármacos , Dados de Sequência Molecular , Oócitos/metabolismo , Técnicas de Patch-Clamp , Proteínas SLC4A , Xenopus laevis
17.
Dev Dyn ; 236(11): 3077-87, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17948311

RESUMO

The recent establishment of recombination-based cloning systems has greatly facilitated the analysis of gene function by allowing rapid and high-efficiency generation of plasmid constructs. However, the use of such an approach in zebrafish requires the availability of recombination-compatible plasmids that are appropriate for functional studies in zebrafish embryos. In this work, we describe the construction and validation of Gateway compatible vectors based on commonly used zebrafish plasmids. We have generated pCS-based plasmids that allow rapid generation of both N-terminal and C-terminal fusion proteins, and we demonstrate that mRNA synthesized from these plasmids encodes functional native or fusion proteins in injected zebrafish embryos. In parallel, we have established similar Gateway plasmids containing Tol2 cis elements that promote efficient integration into the zebrafish genome and allow expression of native or fusion proteins in a tissue-specific manner in the zebrafish embryo. Finally, we demonstrate the use of this system to rapidly identify tissue-specific cis elements to aid the establishment of blood vessel-specific transgenic constructs. Taken together, this work provides an important platform for the rapid functional analyses of open reading frames in zebrafish embryos.


Assuntos
Clonagem Molecular/métodos , Técnicas de Transferência de Genes , Vetores Genéticos , Plasmídeos/genética , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Embrião não Mamífero , Fases de Leitura Aberta/genética , Recombinação Genética/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
18.
Proc Natl Acad Sci U S A ; 104(35): 13996-4001, 2007 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-17715297

RESUMO

The zebrafish is a powerful model for studying vascular development, demonstrating remarkable conservation of this process with mammals. Here, we identify a zebrafish mutant, redhead (rhd(mi149)), that exhibits embryonic CNS hemorrhage with intact gross development of the vasculature and normal hemostatic function. We show that the rhd phenotype is caused by a hypomorphic mutation in p21-activated kinase 2a (pak2a). PAK2 is a kinase that acts downstream of the Rho-family GTPases CDC42 and RAC and has been implicated in angiogenesis, regulation of cytoskeletal structure, and endothelial cell migration and contractility among other functions. Correction of the Pak2a-deficient phenotype by Pak2a overexpression depends on kinase activity, implicating Pak2 signaling in the maintenance of vascular integrity. Rescue by an endothelial-specific transgene further suggests that the hemorrhage seen in Pak2a deficiency is the result of an autonomous endothelial cell defect. Reduced expression of another PAK2 ortholog, pak2b, in Pak2a-deficient embryos results in a more severe hemorrhagic phenotype, consistent with partially overlapping functions for these two orthologs. These data provide in vivo evidence for a critical function of Pak2 in vascular integrity and demonstrate a severe disease phenotype resulting from loss of Pak2 function.


Assuntos
Hemorragia Cerebral/genética , Mutação , Proteínas Serina-Treonina Quinases/genética , Peixe-Zebra/genética , Processamento Alternativo , Animais , Hemorragia Cerebral/embriologia , Circulação Cerebrovascular/genética , Mapeamento Cromossômico , Embrião não Mamífero , Genes Recessivos , Variação Genética , Polimorfismo de Fragmento de Restrição , Proteínas Serina-Treonina Quinases/deficiência , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Proteínas de Peixe-Zebra/genética , Quinases Ativadas por p21
19.
Dev Biol ; 299(2): 551-62, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16999953

RESUMO

In this study, we utilize fluorescent activated cell sorting (FACS) of cells from transgenic zebrafish coupled with microarray analysis to globally analyze expression of cell type specific genes. We find that it is possible to isolate cell populations from Tg(fli1:egfp)(y1) zebrafish embryos that are enriched in vascular, hematopoietic and pharyngeal arch cell types. Microarray analysis of GFP+ versus GFP- cells isolated from Tg(fli1:egfp)(y1) embryos identifies genes expressed in hematopoietic, vascular and pharyngeal arch tissue, consistent with the expression of the fli1:egfp transgene in these cell types. Comparison of expression profiles from GFP+ cells isolated from embryos at two different time points reveals that genes expressed in different fli1+ cell types display distinct temporal expression profiles. We also demonstrate the utility of this approach for gene discovery by identifying numerous previously uncharacterized genes that we find are expressed in fli1:egfp-positive cells, including new markers of blood, endothelial and pharyngeal arch cell types. In parallel, we have developed a database to allow easy access to both our microarray and in situ results. Our results demonstrate that this is a robust approach for identification of cell type specific genes as well as for global analysis of cell type specific gene expression in zebrafish embryos.


Assuntos
Endotélio Vascular/metabolismo , Sistema Hematopoético/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Região Branquial/embriologia , Região Branquial/metabolismo , Separação Celular , Endotélio Vascular/citologia , Endotélio Vascular/embriologia , Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Sistema Hematopoético/embriologia , Análise de Sequência com Séries de Oligonucleotídeos , Peixe-Zebra/embriologia
20.
Rev. cuba. med ; 24(9): 916-27, sept. 1985. ilus
Artigo em Espanhol | LILACS | ID: lil-28030

RESUMO

Se realiza una revisión de diversos procederes diagnósticos utilizados en el estudio de la hipertensión arterial debido a la elevaba morbimortalidad que se observa en la actualidad por esta afección. Un mejor conocimiento de su utilidad relativa permitirá la búsqueda de casos secundarios de hipertensión potencialmente curables. Se señalan las principales alteraciones informadas en el pielograma minutado, el renograma isotópico y la arteriografía renal. Se hacen algunas consideraciones sobre otros métodos de estudio más novedosos, como la digital substraction angiography, (DSA) y las técnicas angiográficas de angulación combinada


Assuntos
Humanos , Masculino , Feminino , Angiografia/métodos , Hipertensão Renal/diagnóstico , Hipertensão Renovascular/diagnóstico , Hipertensão/diagnóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...