Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Am Heart Assoc ; 8(22): e012792, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31718444

RESUMO

Background The SNRK (sucrose-nonfermenting-related kinase) enzyme is critical for cardiac function. However, the underlying cause for heart failure observed in Snrk cardiac conditional knockout mouse is unknown. Methods and Results Previously, 6-month adult mice knocked out for Snrk in cardiomyocytes (CMs) displayed left ventricular dysfunction. Here, 4-month adult mice, on angiotensin II (Ang II) infusion, show rapid decline in cardiac systolic function, which leads to heart failure and death in 2 weeks. These mice showed increased expression of nuclear factor κ light chain enhancer of activated B cells (NF-κB), inflammatory signaling proteins, proinflammatory proteins in the heart, and fibrosis. Interestingly, under Ang II infusion, mice knocked out for Snrk in endothelial cells did not show significant systolic or diastolic dysfunction. Although an NF-κB inflammation signaling pathway was increased in Snrk knockout endothelial cells, this did not lead to fibrosis or mortality. In hearts of adult mice knocked out for Snrk in CMs, we also observed NF-κB pathway activation in CMs, and an increased presence of Mac2+ macrophages was observed in basal and Ang II-infused states. In vitro analysis of Snrk knockdown HL-1 CMs revealed similar upregulation of the NF-κB signaling proteins and proinflammatory proteins that was exacerbated on Ang II treatment. The Ang II-induced NF-κB pathway-mediated proinflammatory effects were mediated in part through protein kinase B or AKT, wherein AKT inhibition restored the proinflammatory signaling protein levels to baseline in Snrk knockdown HL-1 CMs. Conclusions During heart failure, SNRK acts as a cardiomyocyte-specific repressor of cardiac inflammation and fibrosis.


Assuntos
Células Endoteliais/metabolismo , Insuficiência Cardíaca/genética , Inflamação/genética , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/genética , Angiotensina II/farmacologia , Animais , Linhagem Celular , Fibrose/genética , Fibrose/metabolismo , Fibrose/patologia , Coração/efeitos dos fármacos , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Técnicas In Vitro , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , Miocárdio/patologia , Vasoconstritores/farmacologia , Disfunção Ventricular Esquerda
2.
Arterioscler Thromb Vasc Biol ; 38(2): 373-385, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29242271

RESUMO

OBJECTIVE: SNRK (sucrose nonfermenting 1-related kinase) is a novel member of the AMPK (adenosine monophosphate-activated protein kinase)-related superfamily that is activated in the process of angiogenesis. Currently, little is known about the function of SNRK in angiogenesis in the physiological and pathological conditions. APPROACH AND RESULTS: In this study, in Snrk global heterozygous knockout mice, retina angiogenesis and neovessel formation after hindlimb ischemia were suppressed. Consistently, mice with endothelial cell (EC)-specific Snrk deletion exhibited impaired retina angiogenesis, and delayed perfusion recovery and exacerbated muscle apoptosis in ischemic hindlimbs, compared with those of littermate wide-type mice. Endothelial SNRK expression was increased in the extremity vessel samples from nonischemic human. In ECs cultured in hypoxic conditions, HIF1α (hypoxia inducible factor 1α) bound to the SNRK promoter to upregulate SNRK expression. In the nuclei of hypoxic ECs, SNRK complexed with SP1 (specificity protein 1), and together, they bound to an SP1-binding motif in the ITGB1 (ß1 integrin) promoter, resulting in enhanced ITGB1 expression and promoted EC migration. Furthermore, SNRK or SP1 deficiency in ECs ameliorated hypoxia-induced ITGB1 expression and, consequently, inhibited EC migration and angiogenesis. CONCLUSIONS: Taken together, our data have revealed that SNRK/SP1-ITGB1 signaling axis promotes angiogenesis in vivo.


Assuntos
Células Endoteliais/enzimologia , Isquemia/enzimologia , Pulmão/irrigação sanguínea , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Proteínas Serina-Treonina Quinases/metabolismo , Vasos Retinianos/enzimologia , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Apoptose , Velocidade do Fluxo Sanguíneo , Caderinas/genética , Caderinas/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Regulação Enzimológica da Expressão Gênica , Membro Posterior , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Integrina beta1/genética , Integrina beta1/metabolismo , Isquemia/genética , Isquemia/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Fluxo Sanguíneo Regional , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo
3.
Cancer Invest ; 35(7): 456-462, 2017 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-28722495

RESUMO

Sucrose non-fermenting related kinase (SNRK) is a serine/threonine kinase known to regulate cellular metabolism and adipocyte inflammation. Since alterations in adipocyte metabolism play a role in ovarian cancer metastasis, we investigated the expression of SNRK in benign and malignant human ovarian tissue using immunohistochemistry and qPCR. The number of SNRK positive (+) nuclei is increased in malignant tissue compared to benign tissue (21.03% versus 14.90%, p < .0431). The most strongly stained malignant SNRK+ nuclei were stage 1 compared to stage 2-4 disease. Differential expression of SNRK in early versus late stage disease suggests specific roles for SNRK in ovarian cancer metastasis.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias Epiteliais e Glandulares/enzimologia , Neoplasias Ovarianas/enzimologia , Proteínas Serina-Treonina Quinases/análise , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/genética , Carcinoma Epitelial do Ovário , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/secundário , Neoplasias Epiteliais e Glandulares/terapia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
PLoS One ; 11(12): e0167246, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27936095

RESUMO

The mitogen-activated protein kinase (MAPK) pathway regulates many key cellular processes such as differentiation, apoptosis, and survival. The final proteins in this pathway, ERK1/2, are regulated by dual specificity phosphatase 5 (DUSP5). DUSP5 is a nuclear, inducible phosphatase with high affinity and fidelity for ERK1/2. By regulating the final step in the MAPK signaling cascade, DUSP5 exerts strong regulatory control over a central cellular pathway. Like other DUSPs, DUSP5 plays an important role in immune function. In this study, we have utilized new knockout mouse reagents to explore its function further. We demonstrate that global loss of DUSP5 does not result in any gross phenotypic changes. However, loss of DUSP5 affects memory/effector CD8+ T cell populations in response to acute viral infection. Specifically, Dusp5-/- mice have decreased proportions of short-lived effector cells (SLECs) and increased proportions of memory precursor effector cells (MPECs) in response to infection. Further, we show that this phenotype is T cell intrinsic; a bone marrow chimera model restricting loss of DUSP5 to the CD8+ T cell compartment displays a similar phenotype. Dusp5-/- T cells also display increased proliferation, increased apoptosis, and altered metabolic profiles, suggesting that DUSP5 is a pro-survival protein in T cells.


Assuntos
Apoptose/genética , Proliferação de Células/genética , Fosfatases de Especificidade Dupla/genética , Linfócitos T/metabolismo , Animais , Western Blotting , Linfócitos T CD8-Positivos/enzimologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Sobrevivência Celular/genética , Células Cultivadas , Fosfatases de Especificidade Dupla/metabolismo , Regulação Enzimológica da Expressão Gênica , Interferon gama/metabolismo , Vírus da Coriomeningite Linfocítica/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/enzimologia , Linfócitos T/virologia , Fator de Necrose Tumoral alfa/metabolismo
5.
Circ Cardiovasc Genet ; 9(6): 474-486, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27780848

RESUMO

BACKGROUND: Cardiac metabolism is critical for the functioning of the heart, and disturbance in this homeostasis is likely to influence cardiac disorders or cardiomyopathy. Our laboratory has previously shown that SNRK (sucrose nonfermenting related kinase) enzyme, which belongs to the AMPK (adenosine monophosphate-activated kinase) family, was essential for cardiac metabolism in mammals. Snrk global homozygous knockout (KO) mice die at postnatal day 0, and conditional deletion of Snrk in cardiomyocytes (Snrk cmcKO) leads to cardiac failure and death by 8 to 10 months. METHODS AND RESULTS: We performed additional cardiac functional studies using echocardiography and identified further cardiac functional deficits in Snrk cmcKO mice. Nuclear magnetic resonance-based metabolomics analysis identified key metabolic pathway deficits in SNRK knockdown cardiomyocytes in vitro. Specifically, metabolites involved in lipid metabolism and oxidative phosphorylation are altered, and perturbations in these pathways can result in cardiac function deficits and heart failure. A phosphopeptide-based proteomic screen identified ROCK (Rho-associated kinase) as a putative substrate for SNRK, and mass spec-based fragment analysis confirmed key amino acid residues on ROCK that are phosphorylated by SNRK. Western blot analysis on heart lysates from Snrk cmcKO adult mice and SNRK knockdown cardiomyocytes showed increased ROCK activity. In addition, in vivo inhibition of ROCK partially rescued the in vivo Snrk cmcKO cardiac function deficits. CONCLUSIONS: Collectively, our data suggest that SNRK in cardiomyocytes is responsible for maintaining cardiac metabolic homeostasis, which is mediated in part by ROCK, and alteration of this homeostasis influences cardiac function in the adult heart.


Assuntos
Células-Tronco Embrionárias/enzimologia , Metabolismo Energético , Insuficiência Cardíaca/enzimologia , Miócitos Cardíacos/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Quinases Associadas a rho/metabolismo , Animais , Células Cultivadas , Ecocardiografia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/patologia , Metabolismo Energético/efeitos dos fármacos , Fibrose , Predisposição Genética para Doença , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Homozigoto , Células Endoteliais da Veia Umbilical Humana/enzimologia , Metabolismo dos Lipídeos , Espectroscopia de Ressonância Magnética , Metabolômica/métodos , Camundongos Knockout , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Fosforilação Oxidativa , Fenótipo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , Transdução de Sinais , Transfecção , Quinases Associadas a rho/antagonistas & inibidores
6.
Dev Biol ; 410(2): 190-201, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26746789

RESUMO

Nogo-B receptor (NgBR) was identified as a receptor specific for Nogo-B. Our previous work has shown that Nogo-B and its receptor (NgBR) are essential for chemotaxis and morphogenesis of endothelial cells in vitro and intersomitic vessel formation via Akt pathway in zebrafish. Here, we further demonstrated the roles of NgBR in regulating vasculature development in mouse embryo and primitive blood vessel formation in embryoid body culture systems, respectively. Our results showed that NgBR homozygous knockout mice are embryonically lethal at E7.5 or earlier, and Tie2Cre-mediated endothelial cell-specific NgBR knockout (NgBR ecKO) mice die at E11.5 and have severe blood vessel assembly defects in embryo. In addition, mutant embryos exhibit dilation of cerebral blood vessel, resulting in thin-walled endothelial caverns. The similar vascular defects also were detected in Cdh5(PAC)-CreERT2 NgBR inducible ecKO mice. Murine NgBR gene-targeting embryonic stem cells (ESC) were generated by homologous recombination approaches. Homozygous knockout of NgBR in ESC results in cell apoptosis. Heterozygous knockout of NgBR does not affect ESC cell survival, but reduces the formation and branching of primitive blood vessels in embryoid body culture systems. Mechanistically, NgBR knockdown not only decreases both Nogo-B and VEGF-stimulated endothelial cell migration by abolishing Akt phosphorylation, but also decreases the expression of CCM1 and CCM2 proteins. Furthermore, we performed immunofluorescence (IF) staining of NgBR in human cerebral cavernous malformation patient tissue sections. The quantitative analysis results showed that NgBR expression levels in CD31 positive endothelial cells is significantly decreased in patient tissue sections. These results suggest that NgBR may be one of important genes coordinating the cerebral vasculature development.


Assuntos
Vasos Sanguíneos/embriologia , Circulação Cerebrovascular , Receptores de Superfície Celular/genética , Animais , Feminino , Camundongos , Camundongos Knockout , Gravidez
7.
PLoS One ; 10(12): e0144185, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26692198

RESUMO

Elevated plasma triglycerides are associated with increased susceptibility to heart disease and stroke, but the mechanisms behind this relationship are unclear. A clearer understanding of gene products which influence plasma triglycerides might help identify new therapeutic targets for these diseases. The Endothelial Cell Surface expressed Chemotaxis and apoptosis Regulator (ECSCR) was initially studied as an endothelial cell marker, but has recently been identified in white adipocytes, the primary storage cell type for triglycerides. Here we confirm ECSCR expression in white adipocytes and show that Ecscr knockout mice show elevated fasting plasma triglycerides. At a cellular level, cultured 3T3-L1 adipocytes silenced for Ecscr show a blunted Akt phosphorylation response. Additionally we show that the phosphatase and tensin homology containing (PTEN) lipid phosphatase association with ECSCR is increased by insulin stimulation. These data suggest a scenario by which ECSCR contributes to control of white adipocyte lipolysis. In this scenario, white adipocytes lacking Ecscr display elevated PTEN activity, thereby reducing AKT activation and impairing insulin-mediated suppression of lipolysis. Collectively, these results suggest that ECSCR plays a critical function in regulating lipolysis in white adipose tissue.


Assuntos
Adipócitos Brancos/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Lipólise/fisiologia , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Células 3T3-L1 , Adipócitos Brancos/citologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas de Membrana , Camundongos , Camundongos Knockout , PTEN Fosfo-Hidrolase/genética , Proteínas Proto-Oncogênicas c-akt/genética , Triglicerídeos/genética , Triglicerídeos/metabolismo
9.
J Biol Chem ; 290(9): 5449-61, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25568324

RESUMO

Sepsis-mediated endothelial Angiopoeitin-2 (Ang2) signaling may contribute to microvascular remodeling in the developing lung. The mechanisms by which bacterial cell wall components such as LPS mediate Ang2 signaling in human pulmonary microvascular endothelial cells (HPMECs) remain understudied. In HPMEC, LPS-induced Ang2, Tie2, and VEGF-A protein expression was preceded by increased superoxide formation. NADPH oxidase 2 (Nox2) inhibition, but not Nox4 or Nox1 inhibition, attenuated LPS-induced superoxide formation and Ang2, Tie2, and VEGF-A expression. Nox2 silencing, but not Nox4 or Nox1 silencing, inhibited LPS-mediated inhibitor of κ-B kinase ß (IKKß) and p38 phosphorylation and nuclear translocation of NF-κB and AP-1. In HPMECs, LPS increased the number of angiogenic tube and network formations in Matrigel by >3-fold. Conditioned media from LPS-treated cells also induced angiogenic tube and network formation in the presence of Toll-like receptor 4 blockade but not in the presence of Ang2 and VEGF blockade. Nox2 inhibition or conditioned media from Nox2-silenced cells attenuated LPS-induced tube and network formation. Ang2 and VEGF-A treatment rescued angiogenesis in Nox2-silenced cells. We propose that Nox2 regulates LPS-mediated Ang2-dependent autocrine angiogenesis in HPMECs through the IKKß/NF-κB and MAPK/AP-1 pathways.


Assuntos
Angiopoietina-2/metabolismo , Células Endoteliais/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Glicoproteínas de Membrana/metabolismo , NADPH Oxidases/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Angiopoietina-2/genética , Animais , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Expressão Gênica/efeitos dos fármacos , Humanos , Quinase I-kappa B/metabolismo , Immunoblotting , Pulmão/irrigação sanguínea , Glicoproteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Microvasos/citologia , NADPH Oxidase 2 , NADPH Oxidases/genética , NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , Interferência de RNA , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição AP-1/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Biol Open ; 4(1): 48-61, 2014 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-25505152

RESUMO

In this study, we have identified a novel member of the AMPK family, namely Sucrose non-fermenting related kinase (Snrk), that is responsible for maintaining cardiac metabolism in mammals. SNRK is expressed in the heart, and brain, and in cell types such as endothelial cells, smooth muscle cells and cardiomyocytes (CMs). Snrk knockout (KO) mice display enlarged hearts, and die at postnatal day 0. Microarray analysis of embryonic day 17.5 Snrk hearts, and blood profile of neonates display defect in lipid metabolic pathways. SNRK knockdown CMs showed altered phospho-acetyl-coA carboxylase and phospho-AMPK levels similar to global and endothelial conditional KO mouse. Finally, adult cardiac conditional KO mouse displays severe cardiac functional defects and lethality. Our results suggest that Snrk is essential for maintaining cardiac metabolic homeostasis, and shows an autonomous role for SNRK during mammalian development.

11.
Dev Biol ; 386(1): 204-15, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24380800

RESUMO

During early development, GATA factors have been shown to be important for key events of coronary vasculogenesis, including formation of the epicardium. Myocardial GATA factors are required for coronary vascular (CV) formation; however, the role of epicardial localized GATAs in this process has not been addressed. The current study was conducted to investigate the molecular mechanisms by which the epicardium controls coronary vasculogenesis, focusing on the role of epicardial GATAs in establishing the endothelial plexus during early coronary vasculogenesis. To address the role of epicardial GATAs, we ablated GATA4 and GATA6 transcription factors specifically from the mouse epicardium and found that the number of endothelial cells in the sub-epicardium was drastically reduced, and concomitant coronary vascular plexus formation was significantly compromised. Here we present evidence for a novel role for epicardial GATA factors in controlling plexus formation by recruiting endothelial cells to the sub-epicardium.


Assuntos
Vasos Coronários/metabolismo , Endotélio Vascular/metabolismo , Fator de Transcrição GATA4/fisiologia , Fator de Transcrição GATA6/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Pericárdio/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Cruzamentos Genéticos , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA6/genética , Genótipo , Coração/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocárdio/metabolismo , Transdução de Sinais , Fatores de Tempo
12.
PLoS One ; 8(12): e80933, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24324647

RESUMO

Brain metastasis is a defining component of tumor pathophysiology, and the underlying mechanisms responsible for this phenomenon are not well understood. Current dogma is that tumor cells stimulate and activate astrocytes, and this mutual relationship is critical for tumor cell sustenance in the brain. Here, we provide evidence that primary rat neonatal and adult astrocytes secrete factors that proactively induced human lung and breast tumor cell invasion and metastasis capabilities. Among which, tumor invasion factors namely matrix metalloprotease-2 (MMP-2) and MMP-9 were partly responsible for the astrocyte media-induced tumor cell invasion. Inhibiting MMPs reduced the ability of tumor cell to migrate and invade in vitro. Further, injection of astrocyte media-conditioned breast cancer cells in mice showed increased invasive activity to the brain and other distant sites. More importantly, blocking the preconditioned tumor cells with broad spectrum MMP inhibitor decreased the invasion and metastasis of the tumor cells, in particular to the brain in vivo. Collectively, our data implicate astrocyte-derived MMP-2 and MMP-9 as critical players that facilitate tumor cell migration and invasion leading to brain metastasis.


Assuntos
Astrócitos/patologia , Fatores Biológicos/farmacologia , Neoplasias da Mama/patologia , Neoplasias Pulmonares/patologia , Metaloproteinase 2 da Matriz/farmacologia , Metaloproteinase 9 da Matriz/farmacologia , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Fatores Biológicos/metabolismo , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/química , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Invasividade Neoplásica/patologia , Invasividade Neoplásica/prevenção & controle , Transplante de Neoplasias , Cultura Primária de Células , Inibidores de Proteases/farmacologia , Ratos , Ratos Sprague-Dawley , Células Tumorais Cultivadas
13.
Dev Dyn ; 240(10): 2344-53, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21932312

RESUMO

The proepicardium is a transient embryonic structure that is a source of precursors of the epicardium, coronary smooth muscle cells, and may be a source of coronary endothelial cells (EC). To better understand proepicardium development a systematic analysis of EC appearance was performed. Multiple marker analysis showed that EC are present in the mouse proepicardium at embryonic day (E) 9.0 through E9.75. Distinct populations of EC were found that were associated with the liver bud, and the sinus venosus, as well as a population that do not appear to be associated with either of these structures. There was a temporal increase in the number of EC and temporal changes in the distribution of EC within the different populations during PE development. These findings indicate that EC exist in the proepicardium before coronary vasculogenesis, and support a model in which there is a heterogeneous origin for EC in the proepicardium.


Assuntos
Células Endoteliais/citologia , Coração/embriologia , Pericárdio/citologia , Pericárdio/embriologia , Animais , Biomarcadores/metabolismo , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/metabolismo , Células Endoteliais/metabolismo , Feminino , Camundongos , Morfogênese , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo
14.
J Child Neurol ; 25(4): 429-34, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19773461

RESUMO

Autistic spectrum disorders encompass etiologically heterogeneous persons, with many genetic causes. A subgroup of these individuals has mitochondrial disease. Because a variety of metabolic disorders, including mitochondrial disease show regression with fever, a retrospective chart review was performed and identified 28 patients who met diagnostic criteria for autistic spectrum disorders and mitochondrial disease. Autistic regression occurred in 60.7% (17 of 28), a statistically significant increase over the general autistic spectrum disorder population (P < .0001). Of the 17 individuals with autistic regression, 70.6% (12 of 17) regressed with fever and 29.4% (5 of 17) regressed without identifiable linkage to fever or vaccinations. None showed regression with vaccination unless a febrile response was present. Although the study is small, a subgroup of patients with mitochondrial disease may be at risk of autistic regression with fever. Although recommended vaccinations schedules are appropriate in mitochondrial disease, fever management appears important for decreasing regression risk.


Assuntos
Transtornos Globais do Desenvolvimento Infantil/epidemiologia , Febre/epidemiologia , Doenças Mitocondriais/epidemiologia , Adolescente , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Criança , Transtornos Globais do Desenvolvimento Infantil/metabolismo , Transtornos Globais do Desenvolvimento Infantil/fisiopatologia , Pré-Escolar , Comorbidade , Metabolismo Energético/fisiologia , Feminino , Humanos , Incidência , Lactente , Masculino , Mitocôndrias/genética , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/fisiopatologia , Prevalência , Recidiva , Regressão Psicológica , Estudos Retrospectivos , Fatores de Risco , Estresse Fisiológico/fisiologia , Adulto Jovem
15.
Differentiation ; 72(5): 239-49, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15270780

RESUMO

The role of thyroid hormone in Xenopus metamorphosis is particularly well understood as it plays an essential role in that process. However, recent evidence suggests that thyroid hormone may play an earlier role in amphibian embryogenesis. We demonstrate that Xenopus thyroid hormone receptor beta (XTR beta) is expressed shortly after neural fold closure, and that its expression is localized to the developing retina. Retinoid X receptor gamma (RXR gamma), a potential dimerization partner for XTR beta, was also found to be expressed in the retina at early stages, and at later stages RXR gamma was also expressed in the liver diverticulum. Addition of either thyroid hormone or 9-cis retinoic acid, the ligands for XTR beta and RXR gamma, respectively, did not alter the expression of their receptors. However, the addition of thyroid hormone and 9-cis retinoic acid did alter rhodopsin mRNA expression. Addition of thyroid hormone generates a small expansion of the rhodopsin expression domain. When 9-cis retinoic acid or a combination of thyroid hormone and 9-cis retinoic acid was administered, there was a decrease in the expression domain of rhodopsin in the developing retina. These results provide evidence for an early role for XTR beta and RXR gamma in the developing Xenopus retina.


Assuntos
Diferenciação Celular/fisiologia , Embrião não Mamífero/metabolismo , Olho/metabolismo , Receptor X Retinoide gama/genética , Receptores beta dos Hormônios Tireóideos/genética , Animais , Olho/citologia , Olho/embriologia , Perfilação da Expressão Gênica , Ligantes , Receptor X Retinoide gama/metabolismo , Rodopsina/metabolismo , Receptores beta dos Hormônios Tireóideos/metabolismo , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA