Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 42(5): 112528, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37209097

RESUMO

Altered hematopoietic stem cell (HSC) fate underlies primary blood disorders but microenvironmental factors controlling this are poorly understood. Genetically barcoded genome editing of synthetic target arrays for lineage tracing (GESTALT) zebrafish were used to screen for factors expressed by the sinusoidal vascular niche that alter the phylogenetic distribution of the HSC pool under native conditions. Dysregulated expression of protein kinase C delta (PKC-δ, encoded by prkcda) increases the number of HSC clones by up to 80% and expands polyclonal populations of immature neutrophil and erythroid precursors. PKC agonists such as cxcl8 augment HSC competition for residency within the niche and expand defined niche populations. CXCL8 induces association of PKC-δ with the focal adhesion complex, activating extracellular signal-regulated kinase (ERK) signaling and expression of niche factors in human endothelial cells. Our findings demonstrate the existence of reserve capacity within the niche that is controlled by CXCL8 and PKC and has significant impact on HSC phylogenetic and phenotypic fate.


Assuntos
Células Endoteliais , Peixe-Zebra , Animais , Humanos , Células Endoteliais/metabolismo , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Filogenia , Proteína Quinase C-delta/metabolismo , Nicho de Células-Tronco , Interleucina-8/metabolismo
2.
Dev Cell ; 58(12): 1037-1051.e4, 2023 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-37119815

RESUMO

The hematopoietic niche is a supportive microenvironment composed of distinct cell types, including specialized vascular endothelial cells that directly interact with hematopoietic stem and progenitor cells (HSPCs). The molecular factors that specify niche endothelial cells and orchestrate HSPC homeostasis remain largely unknown. Using multi-dimensional gene expression and chromatin accessibility analyses in zebrafish, we define a conserved gene expression signature and cis-regulatory landscape that are unique to sinusoidal endothelial cells in the HSPC niche. Using enhancer mutagenesis and transcription factor overexpression, we elucidate a transcriptional code that involves members of the Ets, Sox, and nuclear hormone receptor families and is sufficient to induce ectopic niche endothelial cells that associate with mesenchymal stromal cells and support the recruitment, maintenance, and division of HSPCs in vivo. These studies set forth an approach for generating synthetic HSPC niches, in vitro or in vivo, and for effective therapies to modulate the endogenous niche.


Assuntos
Nicho de Células-Tronco , Fatores de Transcrição , Animais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Células Endoteliais/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Regulação da Expressão Gênica
3.
Stem Cell Reports ; 18(2): 417-419, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36638789

RESUMO

The rapidly evolving stem cell field puts much stress on developing educational resources. The ISSCR Education Committee has created a flexible stem cell syllabus rooted in core concepts to facilitate stem cell literacy. The free syllabus will be updated regularly to maintain accuracy and relevance.


Assuntos
Currículo , Alfabetização , Células-Tronco
4.
Science ; 372(6543): 716-721, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33986176

RESUMO

Transcription and metabolism both influence cell function, but dedicated transcriptional control of metabolic pathways that regulate cell fate has rarely been defined. We discovered, using a chemical suppressor screen, that inhibition of the pyrimidine biosynthesis enzyme dihydroorotate dehydrogenase (DHODH) rescues erythroid differentiation in bloodless zebrafish moonshine (mon) mutant embryos defective for transcriptional intermediary factor 1 gamma (tif1γ). This rescue depends on the functional link of DHODH to mitochondrial respiration. The transcription elongation factor TIF1γ directly controls coenzyme Q (CoQ) synthesis gene expression. Upon tif1γ loss, CoQ levels are reduced, and a high succinate/α-ketoglutarate ratio leads to increased histone methylation. A CoQ analog rescues mon's bloodless phenotype. These results demonstrate that mitochondrial metabolism is a key output of a lineage transcription factor that drives cell fate decisions in the early blood lineage.


Assuntos
Eritropoese , Mitocôndrias/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteínas de Peixe-Zebra/metabolismo , Animais , Ciclo do Ácido Cítrico , Metilação de DNA , Di-Hidro-Orotato Desidrogenase , Transporte de Elétrons , Embrião não Mamífero/metabolismo , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica , Histonas/metabolismo , Leflunomida/farmacologia , Redes e Vias Metabólicas , Metilação , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/antagonistas & inibidores , Consumo de Oxigênio , Fatores de Transcrição/genética , Ubiquinona/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
5.
ACS Nano ; 13(12): 13853-13865, 2019 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-31479239

RESUMO

The restrictive nature of the blood-brain barrier (BBB) creates a major challenge for brain drug delivery with current nanomedicines lacking the ability to cross the BBB. Extracellular vesicles (EVs) have been shown to contribute to the progression of a variety of brain diseases including metastatic brain cancer and have been suggested as promising therapeutics and drug delivery vehicles. However, the ability of native tumor-derived EVs to breach the BBB and the mechanism(s) involved in this process remain unknown. Here, we demonstrate that tumor-derived EVs can breach the intact BBB in vivo, and by using state-of-the-art in vitro and in vivo models of the BBB, we have identified transcytosis as the mechanism underlying this process. Moreover, high spatiotemporal resolution microscopy demonstrated that the endothelial recycling endocytic pathway is involved in this transcellular transport. We further identify and characterize the mechanism by which tumor-derived EVs circumvent the low physiologic rate of transcytosis in the BBB by decreasing the brain endothelial expression of rab7 and increasing the efficiency of their transport. These findings identify previously unknown mechanisms by which tumor-derived EVs breach an intact BBB during the course of brain metastasis and can be leveraged to guide and inform the development of drug delivery approaches to deliver therapeutic cargoes across the BBB for treatment of a variety of brain diseases including, but not limited to, brain malignancies.


Assuntos
Barreira Hematoencefálica/metabolismo , Neoplasias da Mama/metabolismo , Vesículas Extracelulares/metabolismo , Transcitose , Animais , Neoplasias Encefálicas/secundário , Caveolinas/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Endossomos/metabolismo , Endotélio/metabolismo , Vesículas Extracelulares/ultraestrutura , Feminino , Humanos , Camundongos Nus , Transporte Proteico , Proteínas SNARE/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
6.
Proc Natl Acad Sci U S A ; 115(37): 9252-9257, 2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30139917

RESUMO

Epoxyeicosatrienoic acids (EETs) are lipid-derived signaling molecules with cardioprotective and vasodilatory actions. We recently showed that 11,12-EET enhances hematopoietic induction and engraftment in mice and zebrafish. EETs are known to signal via G protein-coupled receptors, with evidence supporting the existence of a specific high-affinity receptor. Identification of a hematopoietic-specific EET receptor would enable genetic interrogation of EET signaling pathways, and perhaps clinical use of this molecule. We developed a bioinformatic approach to identify an EET receptor based on the expression of G protein-coupled receptors in cell lines with differential responses to EETs. We found 10 candidate EET receptors that are expressed in three EET-responsive cell lines, but not expressed in an EET-unresponsive line. Of these, only recombinant GPR132 showed EET-responsiveness in vitro, using a luminescence-based ß-arrestin recruitment assay. Knockdown of zebrafish gpr132b prevented EET-induced hematopoiesis, and marrow from GPR132 knockout mice showed decreased long-term engraftment capability. In contrast to high-affinity EET receptors, GPR132 is reported to respond to additional hydroxy-fatty acids in vitro, and we found that these same hydroxy-fatty acids enhance hematopoiesis in the zebrafish. We conducted structure-activity relationship analyses using both cell culture and zebrafish assays on diverse medium-chain fatty acids. Certain oxygenated, unsaturated free fatty acids showed high activation of GPR132, whereas unoxygenated or saturated fatty acids had lower activity. Absence of the carbon-1 position carboxylic acid prevented activity, suggesting that this moiety is required for receptor activation. GPR132 responds to a select panel of oxygenated polyunsaturated fatty acids to enhance both embryonic and adult hematopoiesis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Hematopoese/efeitos dos fármacos , Oxilipinas , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas de Peixe-Zebra/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Células Cultivadas , Hematopoese/genética , Camundongos , Camundongos Knockout , Oxilipinas/química , Oxilipinas/farmacologia , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/genética , Relação Estrutura-Atividade , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
7.
Nature ; 558(7710): 445-448, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29899448

RESUMO

Haematopoietic stem and progenitor cells (HSPCs) require a specific microenvironment, the haematopoietic niche, which regulates HSPC behaviour1,2. The location of this niche varies across species, but the evolutionary pressures that drive HSPCs to different microenvironments remain unknown. The niche is located in the bone marrow in adult mammals, whereas it is found in other locations in non-mammalian vertebrates, for example, in the kidney marrow in teleost fish. Here we show that a melanocyte umbrella above the kidney marrow protects HSPCs against ultraviolet light in zebrafish. Because mutants that lack melanocytes have normal steady-state haematopoiesis under standard laboratory conditions, we hypothesized that melanocytes above the stem cell niche protect HSPCs against ultraviolet-light-induced DNA damage. Indeed, after ultraviolet-light irradiation, unpigmented larvae show higher levels of DNA damage in HSPCs, as indicated by staining of cyclobutane pyrimidine dimers and have reduced numbers of HSPCs, as shown by cmyb (also known as myb) expression. The umbrella of melanocytes associated with the haematopoietic niche is highly evolutionarily conserved in aquatic animals, including the sea lamprey, a basal vertebrate. During the transition from an aquatic to a terrestrial environment, HSPCs relocated into the bone marrow, which is protected from ultraviolet light by the cortical bone around the marrow. Our studies reveal that melanocytes above the haematopoietic niche protect HSPCs from ultraviolet-light-induced DNA damage in aquatic vertebrates and suggest that during the transition to terrestrial life, ultraviolet light was an evolutionary pressure affecting the location of the haematopoietic niche.


Assuntos
Evolução Biológica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos da radiação , Melanócitos/citologia , Melanócitos/efeitos da radiação , Nicho de Células-Tronco/efeitos da radiação , Raios Ultravioleta/efeitos adversos , Animais , Organismos Aquáticos/classificação , Citoproteção/efeitos da radiação , Dano ao DNA/efeitos da radiação , Rim , Mutação , Petromyzon/classificação , Filogenia , Dímeros de Pirimidina/efeitos da radiação , Nicho de Células-Tronco/fisiologia , Peixe-Zebra/classificação , Peixe-Zebra/genética
8.
J Exp Med ; 214(10): 2817-2827, 2017 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-28830909

RESUMO

Hematopoietic stem cell transplantation (HSCT) is an important therapy for patients with a variety of hematological malignancies. HSCT would be greatly improved if patient-specific hematopoietic stem cells (HSCs) could be generated from induced pluripotent stem cells in vitro. There is an incomplete understanding of the genes and signals involved in HSC induction, migration, maintenance, and niche engraftment. Recent studies in zebrafish have revealed novel genes that are required for HSC induction and niche regulation of HSC homeostasis. Manipulation of these signaling pathways and cell types may improve HSC bioengineering, which could significantly advance critical, lifesaving HSCT therapies.


Assuntos
Hematopoese/fisiologia , Transplante de Células-Tronco Hematopoéticas , Peixe-Zebra/fisiologia , Animais , Células-Tronco/fisiologia
9.
J Mol Med (Berl) ; 95(8): 809-819, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28702683

RESUMO

Cells of the hematopoietic system undergo rapid turnover. Each day, humans require the production of about one hundred billion new blood cells for proper function. Hematopoietic stem cells (HSCs) are rare cells that reside in specialized niches and are required throughout life to produce specific progenitor cells that will replenish all blood lineages. There is, however, an incomplete understanding of the molecular and physical properties that regulate HSC migration, homing, engraftment, and maintenance in the niche. Endothelial cells (ECs) are intimately associated with HSCs throughout the life of the stem cell, from the specialized endothelial cells that give rise to HSCs, to the perivascular niche endothelial cells that regulate HSC homeostasis. Recent studies have dissected the unique molecular and physical properties of the endothelial cells in the HSC vascular niche and their role in HSC biology, which may be manipulated to enhance hematopoietic stem cell transplantation therapies.


Assuntos
Células Endoteliais/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Comunicação Celular , Quimiocinas/fisiologia , Transplante de Células-Tronco Hematopoéticas , Humanos
10.
Stem Cell Reports ; 8(5): 1226-1241, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28416284

RESUMO

Hematopoietic stem/progenitor cells (HSPCs) are formed during ontogeny from hemogenic endothelium in the ventral wall of the dorsal aorta (VDA). Critically, the cellular mechanism(s) allowing HSPC egress and migration to secondary niches are incompletely understood. Matrix metalloproteinases (MMPs) are inflammation-responsive proteins that regulate extracellular matrix (ECM) remodeling, cellular interactions, and signaling. Here, inhibition of vascular-associated Mmp2 function caused accumulation of fibronectin-rich ECM, retention of runx1/cmyb+ HSPCs in the VDA, and delayed caudal hematopoietic tissue (CHT) colonization; these defects were absent in fibronectin mutants, indicating that Mmp2 facilitates endothelial-to-hematopoietic transition via ECM remodeling. In contrast, Mmp9 was dispensable for HSPC budding, being instead required for proper colonization of secondary niches. Significantly, these migration defects were mimicked by overexpression and blocked by knockdown of C-X-C motif chemokine-12 (cxcl12), suggesting that Mmp9 controls CHT homeostasis through chemokine regulation. Our findings indicate Mmp2 and Mmp9 play distinct but complementary roles in developmental HSPC production and migration.


Assuntos
Movimento Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Hematopoéticas/citologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Nicho de Células-Tronco , Animais , Proliferação de Células , Quimiocina CXCL12/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/fisiologia , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
11.
Development ; 138(21): 4639-48, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21965611

RESUMO

During peripheral nerve development, each segment of a myelinated axon is matched with a single Schwann cell. Tight regulation of Schwann cell movement, proliferation and differentiation is essential to ensure that these glial cells properly associate with axons. ErbB receptors are required for Schwann cell migration, but the operative ligand and its mechanism of action have remained unknown. We demonstrate that zebrafish Neuregulin 1 (Nrg1) type III, which signals through ErbB receptors, controls Schwann cell migration in addition to its previously known roles in proliferation and myelination. Chimera analyses indicate that ErbB receptors are required in all migrating Schwann cells, and that Nrg1 type III is required in neurons for migration. Surprisingly, expression of the ligand in a few axons is sufficient to induce migration along a chimeric nerve constituted largely of nrg1 type III mutant axons. These studies also reveal a mechanism that allows Schwann cells to fasciculate axons regardless of nrg1 type III expression. Time-lapse imaging of transgenic embryos demonstrated that misexpression of human NRG1 type III results in ectopic Schwann cell migration, allowing them to aberrantly enter the central nervous system. These results demonstrate that Nrg1 type III is an essential signal that controls Schwann cell migration to ensure that these glia are present in the correct numbers and positions in developing nerves.


Assuntos
Movimento Celular/fisiologia , Neuregulina-1/metabolismo , Isoformas de Proteínas/metabolismo , Células de Schwann/fisiologia , Peixe-Zebra/anatomia & histologia , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Biomarcadores/metabolismo , Diferenciação Celular/fisiologia , Proliferação de Células , Humanos , Dados de Sequência Molecular , Neuregulina-1/genética , Neurônios/citologia , Neurônios/metabolismo , Isoformas de Proteínas/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Células de Schwann/citologia , Alinhamento de Sequência , Quimeras de Transplante , Peixe-Zebra/embriologia
12.
Development ; 137(21): 3643-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20876648

RESUMO

Although much is known about the initial construction of the peripheral nervous system (PNS), less well understood are the processes that maintain the position and connections of nerves during postembryonic growth. Here, we show that the posterior lateral line nerve in zebrafish initially grows in the epidermis and then rapidly transitions across the epidermal basement membrane into the subepidermal space. Our experiments indicate that Schwann cells, which myelinate axons in the PNS, are required to reposition the nerve. In mutants lacking Schwann cells, the nerve is mislocalized and the axons remain in the epidermis. Transplanting wild-type Schwann cells into these mutants rescues the position of the nerve. Analysis of chimeric embryos suggests that the process of nerve relocalization involves two discrete steps - the degradation and recreation of the epidermal basement membrane. Although the outgrowth of axons is normal in mutants lacking Schwann cells, the nerve becomes severely disorganized at later stages. In wild-type embryos, exclusion of the nerve from the epidermis isolates axons from migration of their targets (sensory neuromasts) within the epidermis. Without Schwann cells, axons remain within the epidermis and are dragged along with the migrating neuromasts. Our analysis of the posterior lateral line system defines a new process in which Schwann cells relocate a nerve beneath the epidermal basement membrane to insulate axons from the postembryonic remodeling of their targets.


Assuntos
Movimento Celular/fisiologia , Epiderme/crescimento & desenvolvimento , Epiderme/inervação , Nervos Periféricos/fisiologia , Células de Schwann/fisiologia , Animais , Animais Geneticamente Modificados , Membrana Basal/inervação , Membrana Basal/fisiologia , Embrião não Mamífero , Modelos Biológicos , Regeneração Nervosa/fisiologia , Peixe-Zebra/embriologia , Peixe-Zebra/fisiologia
13.
Science ; 325(5946): 1402-5, 2009 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-19745155

RESUMO

The myelin sheath allows axons to conduct action potentials rapidly in the vertebrate nervous system. Axonal signals activate expression of specific transcription factors, including Oct6 and Krox20, that initiate myelination in Schwann cells. Elevation of cyclic adenosine monophosphate (cAMP) can mimic axonal contact in vitro, but the mechanisms that regulate cAMP levels in vivo are unknown. Using mutational analysis in zebrafish, we found that the G protein-coupled receptor Gpr126 is required autonomously in Schwann cells for myelination. In gpr126 mutants, Schwann cells failed to express oct6 and krox20 and were arrested at the promyelinating stage. Elevation of cAMP in gpr126 mutants, but not krox20 mutants, could restore myelination. We propose that Gpr126 drives the differentiation of promyelinating Schwann cells by elevating cAMP levels, thereby triggering Oct6 expression and myelination.


Assuntos
Bainha de Mielina/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Células de Schwann/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Axônios/fisiologia , Axônios/ultraestrutura , Diferenciação Celular , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteína 2 de Resposta de Crescimento Precoce/genética , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Sistema da Linha Lateral/inervação , Dados de Sequência Molecular , Mutação , Proteína Básica da Mielina/metabolismo , Neuregulina-1/metabolismo , Fator 6 de Transcrição de Octâmero/genética , Fator 6 de Transcrição de Octâmero/metabolismo , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo , Receptores Acoplados a Proteínas G/genética , Células de Schwann/citologia , Transdução de Sinais , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/genética
14.
J Cell Biol ; 178(5): 721-3, 2007 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-17724116

RESUMO

Interactions between Schwann cells and axons are critical for the development and function of myelinated axons. Two recent studies (see Maurel et al. on p. 861 of this issue; Spiegel et al., 2007) report that the nectin-like (Necl) proteins Necl-1 and -4 are internodal adhesion molecules that are critical for myelination. These studies suggest that Necl proteins mediate a specific interaction between Schwann cells and axons that allows proper communication of the signals that trigger myelination.


Assuntos
Axônios/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Adesão Celular/fisiologia , Células de Schwann/metabolismo , Animais , Axônios/ultraestrutura , Moléculas de Adesão Celular Neuronais/genética , Bainha de Mielina/metabolismo , Nós Neurofibrosos/metabolismo , Nós Neurofibrosos/ultraestrutura , Células de Schwann/citologia
15.
Sci STKE ; 2007(400): pe45, 2007 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-17712137

RESUMO

The chemokine SDF1 (stromal cell-derived factor 1) directs cell migration in many different contexts, ranging from embryogenesis to inflammation. SDF1a is the guidance cue for the zebrafish lateral line primordium, a tissue that moves along the flank of the embryo and deposits cells that form mechanosensory organs. The SDF1a receptor CXCR4b acts in cells at the leading edge of the primordium to direct its migration. Two new studies show that a second SDF1 receptor, CXCR7, is required only in the trailing cells of the primordium, and they explore how these two receptors orchestrate migration of the primordium. CXCR4b and CXCR7 are expressed in complementary domains, possibly through mutual repression in which each receptor inhibits expression of the other. These studies illustrate how the entire primordium can respond to a single signal, yet generate cell type-specific responses by using different receptors.


Assuntos
Organogênese/fisiologia , Receptores de Quimiocinas/metabolismo , Transdução de Sinais , Peixe-Zebra/metabolismo , Animais , Modelos Biológicos , Receptores CXCR4/metabolismo , Receptores CXCR4/fisiologia , Receptores de Quimiocinas/fisiologia , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia
16.
Science ; 315(5811): 518-21, 2007 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-17255513

RESUMO

Adult stem cells often divide asymmetrically to produce one self-renewed stem cell and one differentiating cell, thus maintaining both populations. The asymmetric outcome of stem cell divisions can be specified by an oriented spindle and local self-renewal signals from the stem cell niche. Here we show that developmentally programmed asymmetric behavior and inheritance of mother and daughter centrosomes underlies the stereotyped spindle orientation and asymmetric outcome of stem cell divisions in the Drosophila male germ line. The mother centrosome remains anchored near the niche while the daughter centrosome migrates to the opposite side of the cell before spindle formation.


Assuntos
Divisão Celular , Centrossomo/fisiologia , Células Germinativas/citologia , Células-Tronco/citologia , Junções Aderentes/ultraestrutura , Animais , Proteínas de Ligação a Calmodulina , Diferenciação Celular , Centríolos/fisiologia , Centrossomo/ultraestrutura , Proteínas de Drosophila/análise , Proteínas de Drosophila/genética , Drosophila melanogaster , Células Germinativas/fisiologia , Interfase , Masculino , Microtúbulos/fisiologia , Microtúbulos/ultraestrutura , Proteínas Recombinantes de Fusão/análise , Fuso Acromático/fisiologia , Células-Tronco/fisiologia
17.
Science ; 306(5694): 279-83, 2004 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-15345777

RESUMO

Methylation of arginine (Arg) and lysine residues in histones has been correlated with epigenetic forms of gene regulation. Although histone methyltransferases are known, enzymes that demethylate histones have not been identified. Here, we demonstrate that human peptidylarginine deiminase 4 (PAD4) regulates histone Arg methylation by converting methyl-Arg to citrulline and releasing methylamine. PAD4 targets multiple sites in histones H3 and H4, including those sites methylated by coactivators CARM1 (H3 Arg17) and PRMT1 (H4 Arg3). A decrease of histone Arg methylation, with a concomitant increase of citrullination, requires PAD4 activity in human HL-60 granulocytes. Moreover, PAD4 activity is linked with the transcriptional regulation of estrogen-responsive genes in MCF-7 cells. These data suggest that PAD4 mediates gene expression by regulating Arg methylation and citrullination in histones.


Assuntos
Arginina/metabolismo , Histonas/metabolismo , Hidrolases/metabolismo , Sequência de Aminoácidos , Western Blotting , Calcimicina/farmacologia , Linhagem Celular Tumoral , Citrulina/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Células HL-60 , Humanos , Ionóforos/farmacologia , Proteínas de Membrana/genética , Metilaminas/metabolismo , Metilação , Dados de Sequência Molecular , Presenilina-2 , Regiões Promotoras Genéticas , Proteína-Arginina Desiminase do Tipo 4 , Desiminases de Arginina em Proteínas , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...