Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
J Biochem Mol Toxicol ; 30(7): 321-30, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26890134

RESUMO

Non-alcoholic fatty liver disease can result in changes to drug metabolism and disposition potentiating adverse drug reactions. Furthermore, arsenite exposure during development compounds the severity of diet-induced fatty liver disease. This study examines the effects of arsenite potentiated diet-induced fatty liver disease on hepatic transport in male mice. Changes were detected for Mrp2/3/4 hepatic transporter gene expression as well as for Oatp1a4/2b1/1b2. Plasma concentrations of Mrp and Oatp substrates were increased in arsenic exposure groups compared with diet-only controls. In addition, murine embryonic hepatocytes and adult primary hepatocytes show significantly altered transporter expression after exposure to arsenite alone: a previously unreported phenomenon. These data indicate that developmental exposure to arsenite leads to changes in hepatic transport which could increase the risk for ADRs during fatty liver disease.


Assuntos
Arsenitos/toxicidade , Dieta Hiperlipídica/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Embrião de Mamíferos , Feminino , Feto , Hepatócitos/metabolismo , Hepatócitos/patologia , Fígado/metabolismo , Fígado/patologia , Transportador 1 de Ânion Orgânico Específico do Fígado , Masculino , Camundongos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Proteínas de Transporte de Cátions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Gravidez , Cultura Primária de Células , Transdução de Sinais
2.
Drug Chem Toxicol ; 39(3): 279-83, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26446802

RESUMO

Although it is generally believed that the developing fetus is principally exposed to inorganic arsenic and the methylated metabolites from the maternal metabolism of arsenic, little is known about whether the developing embryo can autonomously metabolize arsenic. This study investigates inorganic arsenic methylation by murine embryonic organ cultures of the heart, lung, and liver. mRNA for AS3mt, the gene responsible for methylation of arsenic, was detected in all embryonic tissue types studied. In addition, methylated arsenic metabolites were generated by all three tissue types. The fetal liver explants yielded the most methylated arsenic metabolites (∼7% of total arsenic/48 h incubation) while the heart, and lung preparations produced slightly greater than 2% methylated metabolites. With all tissues the methylation proceeded mostly to the dimethylated arsenic species. This has profound implications for understanding arsenic-induced fetal toxicity, particularly if the methylated metabolites are produced autonomously by embryonic tissues.


Assuntos
Arsenitos/metabolismo , Coração , Fígado/metabolismo , Pulmão/metabolismo , Miocárdio/metabolismo , Compostos de Sódio/metabolismo , Animais , Arsenitos/toxicidade , Biotransformação , Feminino , Expressão Gênica/efeitos dos fármacos , Coração/efeitos dos fármacos , Coração/embriologia , Fígado/efeitos dos fármacos , Fígado/embriologia , Pulmão/efeitos dos fármacos , Pulmão/embriologia , Metilação , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos , Técnicas de Cultura de Órgãos , Compostos de Sódio/toxicidade
3.
Toxicol Appl Pharmacol ; 272(1): 147-53, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23732083

RESUMO

Exposure to arsenic results in several types of cancers as well as heart disease. A major contributor to ischemic heart pathologies is coronary artery disease, however the influences by environmental arsenic in this disease process are not known. Similarly, the impact of toxicants on blood vessel formation and function during development has not been studied. During embryogenesis, the epicardium undergoes proliferation, migration, and differentiation into several cardiac cell types including smooth muscle cells which contribute to the coronary vessels. The TGFß family of ligands and receptors is essential for developmental cardiac epithelial to mesenchymal transition (EMT) and differentiation into coronary smooth muscle cells. In this in vitro study, 18hour exposure to 1.34µM arsenite disrupted developmental EMT programming in murine epicardial cells causing a deficit in cardiac mesenchyme. The expression of EMT genes including TGFß2, TGFß receptor-3, Snail, and Has-2 are decreased in a dose-dependent manner following exposure to arsenite. TGFß2 cell signaling is abrogated as detected by decreases in phosphorylated Smad2/3 when cells are exposed to 1.34µM arsenite. There is also loss of nuclear accumulation pSmad due to arsenite exposure. These observations coincide with a decrease in vimentin positive mesenchymal cells invading three-dimensional collagen gels. However, arsenite does not block TGFß2 mediated smooth muscle cell differentiation by epicardial cells. Overall these results show that arsenic exposure blocks developmental EMT gene programming in murine coronary progenitor cells by disrupting TGFß2 signals and Smad activation, and that smooth muscle cell differentiation is refractory to this arsenic toxicity.


Assuntos
Arsenitos/toxicidade , Vasos Coronários/citologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Fator de Crescimento Transformador beta/fisiologia , Animais , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Vasos Coronários/efeitos dos fármacos , Imunofluorescência , Indicadores e Reagentes , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas Musculares/genética , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas Smad/metabolismo
4.
Semin Cell Dev Biol ; 21(9): 929-35, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20933094

RESUMO

The ErbB family of receptor tyrosine kinases (RTKs) is a family of receptors that allow cells to interact with the extracellular environment and transduce signals to the nucleus that promote differentiation, migration and proliferation necessary for proper heart morphogenesis and function. This review focuses on the role of the ErbB family of receptor tyrosine kinases, and their importance in proper heart morphogenesis, as well as their role in maintenance and function of the adult heart. Studies from transgenic mouse models have shown the importance of ErbB receptors in heart development, and provide insight into potential future therapeutic targets to help reduce congenital heart defect (CHD) mortality rates and prevent disease in adults. Cancer therapeutics have also shed light to the ErbB receptors and signaling network, as undesired side effects have demonstrated their importance in adult cardiomyocytes and prevention of cardiomyopathies. This review will discuss ErbB receptor tyrosine kinases (RTK) in heart development and disease including valve formation and partitioning of a four-chambered heart as well as cardiotoxicity when ErbB signaling is attenuated in adults.


Assuntos
Receptores ErbB/metabolismo , Cardiopatias/metabolismo , Miócitos Cardíacos/metabolismo , Transdução de Sinais , Animais , Receptores ErbB/genética , Humanos
5.
Dev Biol ; 358(2): 331-43, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21871877

RESUMO

The epicardium is a major contributor of the cells that are required for the formation of coronary vessels. Mice lacking both copies of the gene encoding the Type III Transforming Growth Factor ß Receptor (TGFßR3) fail to form the coronary vasculature, but the molecular mechanism by which TGFßR3 signals coronary vessel formation is unknown. We used intact embryos and epicardial cells from E11.5 mouse embryos to reveal the mechanisms by which TGFßR3 signals and regulates epicardial cell behavior. Analysis of E13.5 embryos reveals a lower rate of epicardial cell proliferation and decreased epicardially derived cell invasion in Tgfbr3(-/-) hearts. Tgfbr3(-/-) epicardial cells in vitro show decreased proliferation and decreased invasion in response to TGFß1 and TGFß2. Unexpectedly, loss of TGFßR3 also decreases responsiveness to two other important regulators of epicardial cell behavior, FGF2 and HMW-HA. Restoring full length TGFßR3 in Tgfbr3(-/-) cells rescued deficits in invasion in vitro in response TGFß1 and TGFß2 as well as FGF2 and HMW-HA. Expression of TGFßR3 missing the 3 C-terminal amino acids that are required to interact with the scaffolding protein GIPC1 did not rescue any of the deficits. Overexpression of GIPC1 alone in Tgfbr3(-/-) cells did not rescue invasion whereas knockdown of GIPC1 in Tgfbr3(+/+) cells decreased invasion in response to TGFß2, FGF2, and HMW-HA. We conclude that TGFßR3 interaction with GIPC1 is critical for regulating invasion and growth factor responsiveness in epicardial cells and that dysregulation of epicardial cell proliferation and invasion contributes to failed coronary vessel development in Tgfbr3(-/-) mice.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Neuropeptídeos/química , Neuropeptídeos/metabolismo , Pericárdio/citologia , Pericárdio/metabolismo , Proteoglicanas/química , Proteoglicanas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/química , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Sequência de Bases , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Anomalias dos Vasos Coronários/embriologia , Anomalias dos Vasos Coronários/genética , Anomalias dos Vasos Coronários/metabolismo , Primers do DNA/genética , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Cardiovasculares , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/genética , Pericárdio/embriologia , Gravidez , Domínios e Motivos de Interação entre Proteínas , Proteoglicanas/deficiência , Proteoglicanas/genética , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais , Imagem com Lapso de Tempo , Fator de Crescimento Transformador beta1/farmacologia , Fator de Crescimento Transformador beta2/farmacologia
6.
Cell Tissue Res ; 347(1): 203-23, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21953136

RESUMO

The majority of children with congenital heart disease now live into adulthood due to the remarkable surgical and medical advances that have taken place over the past half century. Because of this, adults now represent the largest age group with adult cardiovascular diseases. It includes patients with heart diseases that were not detected or not treated during childhood, those whose defects were surgically corrected but now need revision due to maladaptive responses to the procedure, those with exercise problems and those with age-related degenerative diseases. Because adult cardiovascular diseases in this population are relatively new, they are not well understood. It is therefore necessary to understand the molecular and physiological pathways involved if we are to improve treatments. Since there is a developmental basis to adult cardiovascular disease, transforming growth factor beta (TGFß) signaling pathways that are essential for proper cardiovascular development may also play critical roles in the homeostatic, repair and stress response processes involved in adult cardiovascular diseases. Consequently, we have chosen to summarize the current information on a subset of TGFß ligand and receptor genes and related effector genes that, when dysregulated, are known to lead to cardiovascular diseases and adult cardiovascular deficiencies and/or pathologies. A better understanding of the TGFß signaling network in cardiovascular disease and repair will impact genetic and physiologic investigations of cardiovascular diseases in elderly patients and lead to an improvement in clinical interventions.


Assuntos
Reabilitação Cardíaca , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Envelhecimento/fisiologia , Angiotensina II/metabolismo , Animais , Doenças Cardiovasculares/terapia , Transição Epitelial-Mesenquimal/fisiologia , Expressão Gênica , Variação Genética , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Smad/metabolismo
7.
Toxicol Appl Pharmacol ; 261(3): 263-70, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22521605

RESUMO

C-reactive protein (CRP) is an acute phase protein in humans. Elevated levels of CRP are produced in response to inflammatory cytokines and are associated with atherosclerosis, hypertension, cardiovascular disease and insulin resistance. Exposure to inorganic arsenic, a common environmental toxicant, also produces cardiovascular disorders, namely atherosclerosis and is associated with insulin-resistance. Inorganic arsenic has been shown to contribute to cardiac toxicities through production of reactive oxygen species (ROS) that result in the activation of NFκB. In this study we show that exposure of the hepatic cell line, HepG2, to environmentally relevant levels of arsenite (0.13 to 2 µM) results in elevated CRP expression and secretion. ROS analysis of the samples showed that a minimal amount of ROS are produced by HepG2 cells in response to these concentrations of arsenic. In addition, treatment of FvB mice with 100 ppb sodium arsenite in the drinking water for 6 months starting at weaning age resulted in dramatically higher levels of CRP in both the liver and inner medullary region of the kidney. Further, mouse Inner Medullary Collecting Duct cells (mIMCD-4), a mouse kidney cell line, were stimulated with 10 ng/ml CRP which resulted in activation of NFκB. Pretreatment with 10 nM Y27632, a known Rho-kinase inhibitor, prior to CRP exposure attenuated NFκB activation. These data suggest that arsenic causes the expression and secretion of CRP and that CRP activates NFκB through activation of the Rho-kinase pathway, thereby providing a novel pathway by which arsenic can contribute to metabolic syndrome and cardiovascular disease.


Assuntos
Arsenitos/farmacologia , Proteína C-Reativa/fisiologia , NF-kappa B/metabolismo , Amidas/farmacologia , Animais , Biomarcadores/análise , Biotransformação/efeitos dos fármacos , Western Blotting , Proteína C-Reativa/análise , Proteína C-Reativa/biossíntese , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , L-Lactato Desidrogenase/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Luciferases/metabolismo , Camundongos , NF-kappa B/genética , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transfecção , Quinases Associadas a rho/antagonistas & inibidores
8.
Toxicol Pathol ; 40(3): 504-12, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22215511

RESUMO

Cardiovascular disease is the leading cause of death in the United States and worldwide. High incidence of cardiovascular diseases has been linked to populations with elevated arsenic content in their drinking water. Although this correlation has been established in many epidemiological studies, a lack of experimental models to study mechanisms of arsenic-related cardiovascular pathogenesis has limited our understanding of how arsenic exposure predisposes for development of hypertension and increased cardiovascular mortality. Our studies show that mice chronically exposed to drinking water containing 100 parts per billion (ppb) sodium arsenite for 22 weeks show an increase in both systolic and diastolic blood pressure. Echocardiographic analyses as well as histological assessment show concentric left ventricular hypertrophy, a primary cardiac manifestation of chronic hypertension. Live imaging by echocardiography shows a 43% increase in left ventricular mass in arsenic-treated animals. Relative wall thickness (RWT) was calculated showing that all the arsenic-exposed animals show an RWT greater than 0.45, indicating concentric hypertrophy. Importantly, left ventricular hypertrophy, although often associated with chronic hypertension, is an independent risk factor for cardiovascular-related mortalities. These results suggest that chronic low-level arsenite exposure promotes the development of hypertension and the comorbidity of concentric hypertrophy.


Assuntos
Arsenitos/toxicidade , Pressão Sanguínea/efeitos dos fármacos , Hipertensão/induzido quimicamente , Hipertrofia Ventricular Esquerda/induzido quimicamente , Compostos de Sódio/toxicidade , Administração Oral , Análise de Variância , Animais , Arsenitos/administração & dosagem , Água Potável/normas , Ecocardiografia , Feminino , Histocitoquímica , Hipertrofia Ventricular Esquerda/patologia , Camundongos , Compostos de Sódio/administração & dosagem , Testes de Toxicidade Crônica
9.
Dev Dyn ; 240(9): 2127-41, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21780244

RESUMO

Although the function of transforming growth factor beta2 (TGFß2) in epithelial mesenchymal transition (EMT) is well studied, its role in valve remodeling remains to be fully explored. Here, we used histological, morphometric, immunohistochemical and molecular approaches and showed that significant dysregulation of major extracellular matrix (ECM) components contributed to valve remodeling defects in Tgfb2(-/-) embryos. The data indicated that cushion mesenchymal cell differentiation was impaired in Tgfb2(-/-) embryos. Hyaluronan and cartilage link protein-1 (CRTL1) were increased in hyperplastic valves of Tgfb2(-/-) embryos, indicating increased expansion and diversification of cushion mesenchyme into the cartilage cell lineage during heart development. Finally, Western blot and immunohistochemistry analyses indicate that the activation of SMAD2/3 was decreased in Tgfb2(-/-) embryos during valve remodeling. Collectively, the data indicate that TGFß2 promotes valve remodeling and differentiation by inducing matrix organization and suppressing cushion mesenchyme differentiation into cartilage cell lineage during heart development.


Assuntos
Valvas Cardíacas/metabolismo , Coração/embriologia , Fator de Crescimento Transformador beta2/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Matriz Extracelular/metabolismo , Valvas Cardíacas/embriologia , Imuno-Histoquímica , Mesoderma/citologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta2/genética
10.
Nat Med ; 8(8): 850-5, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12134143

RESUMO

Heart septation and valve malformations constitute the most common anatomical birth defects. These structures arise from the endocardial cushions within the atrioventricular canal (AVC) through dynamic interactions between cushion cells and the extracellular matrix (termed cardiac jelly). Transformation of endothelial cells to mesenchymal cells is essential for the proper development of the AVC and subsequent septation and valve formation. Atrioventricular septal defects can result from incomplete endocardial cushion morphogenesis. We show that hyaluronan-deficient AVC explants from Has2(-/-) embryos, which normally lack mesenchyme formation, are rescued by heregulin treatment, which restores phosphorylation of ErbB2 and ErbB3. These events were blocked using a soluble ErbB3 molecule, as well as with an inhibitor of ErbB2, herstatin. We show further that ErbB3 is activated during hyaluronan treatment of Has2(-/-) explants. These data provide a link between extracellular matrix-hyaluronan and ErbB receptor activation during development of early heart-valve and septal mesenchyme.


Assuntos
Valvas Cardíacas/embriologia , Valvas Cardíacas/metabolismo , Ácido Hialurônico/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Mesoderma/metabolismo , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Técnicas de Cultura , Endotélio/citologia , Endotélio/metabolismo , Cardiopatias Congênitas/fisiopatologia , Valvas Cardíacas/citologia , Ácido Hialurônico/genética , Mesoderma/citologia , Camundongos , Camundongos Knockout , Neuregulina-1/metabolismo , Fenótipo , Fosforilação , Receptor ErbB-2/genética , Receptor ErbB-3/genética
11.
Exp Cell Res ; 316(20): 3397-405, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-20633555

RESUMO

In the developing heart, the epicardium is a major source of progenitor cells that contribute to the formation of the coronary vessel system. These epicardial progenitors give rise to the different cellular components of the coronary vasculature by undergoing a number of morphological and physiological changes collectively known as epithelial to mesenchymal transformation (EMT). However, the specific signaling mechanisms that regulate epicardial EMT are yet to be delineated. In this study we investigated the role of TGFß2 and hyaluronan (HA) during epicardial EMT and how signals from these two molecules are integrated during this important process. Here we show that TGFß2 induces MEKK3 activation, which in turn promotes ERK1/2 and ERK5 phosphorylation. TGFß2 also increases Has2 expression and subsequent HA production. Nevertheless, inhibition of MEKK3 kinase activity, silencing of ERK5 or pharmacological disruption of ERK1/2 activation significantly abrogates this response. Thus, TGFß2 promotes Has2 expression and HA production through a MEKK3/ERK1/2/5-dependent cascade. Furthermore, TGFß2 is able to induce epicardial cell invasion and differentiation but not proliferation. However, inhibition of MEKK3-dependent pathways, degradation of HA by hyaluronidases or blockade of CD44, significantly impairs the biological response to TGFß2. Taken together, these findings demonstrate that TGFß2 activation of MEKK3/ERK1/2/5 signaling modulates Has2 expression and HA production leading to the induction of EMT events. This is an important and novel mechanism showing how TGFß2 and HA signals are integrated to regulate changes in epicardial cell behavior.


Assuntos
Movimento Celular/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Ácido Hialurônico/metabolismo , Pericárdio/citologia , Células-Tronco/citologia , Fator de Crescimento Transformador beta2/farmacologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Humanos , Receptores de Hialuronatos/imunologia , Hialuronan Sintases , Hialuronoglucosaminidase/farmacologia , MAP Quinase Quinase Quinase 3/genética , MAP Quinase Quinase Quinase 3/metabolismo , Camundongos , Camundongos Transgênicos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/genética , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Vimentina/metabolismo
12.
medRxiv ; 2021 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-33564791

RESUMO

Background: Wastewater surveillance for SARS-CoV-2 is an emerging approach to help identify the risk of a COVID-19 outbreak. This tool can contribute to public health surveillance at both community (wastewater treatment system) and institutional (e.g., colleges, prisons, nursing homes) scales. Objectives: This research aims to understand the successes, challenges, and lessons learned from initial wastewater surveillance efforts at colleges and university systems to inform future research, development and implementation. Methods: This paper presents the experiences of 25 college and university systems in the United States that monitored campus wastewater for SARS-CoV-2 during the fall 2020 academic period. We describe the broad range of approaches, findings, resource needs, and lessons learned from these initial efforts. These institutions range in size, social and political geographies, and include both public and private institutions. Discussion: Our analysis suggests that wastewater monitoring at colleges requires consideration of information needs, local sewage infrastructure, resources for sampling and analysis, college and community dynamics, approaches to interpretation and communication of results, and follow-up actions. Most colleges reported that a learning process of experimentation, evaluation, and adaptation was key to progress. This process requires ongoing collaboration among diverse stakeholders including decision-makers, researchers, faculty, facilities staff, students, and community members.

13.
Artigo em Inglês | MEDLINE | ID: mdl-33922263

RESUMO

Wastewater surveillance for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emerging approach to help identify the risk of a coronavirus disease (COVID-19) outbreak. This tool can contribute to public health surveillance at both community (wastewater treatment system) and institutional (e.g., colleges, prisons, and nursing homes) scales. This paper explores the successes, challenges, and lessons learned from initial wastewater surveillance efforts at colleges and university systems to inform future research, development and implementation. We present the experiences of 25 college and university systems in the United States that monitored campus wastewater for SARS-CoV-2 during the fall 2020 academic period. We describe the broad range of approaches, findings, resources, and impacts from these initial efforts. These institutions range in size, social and political geographies, and include both public and private institutions. Our analysis suggests that wastewater monitoring at colleges requires consideration of local information needs, sewage infrastructure, resources for sampling and analysis, college and community dynamics, approaches to interpretation and communication of results, and follow-up actions. Most colleges reported that a learning process of experimentation, evaluation, and adaptation was key to progress. This process requires ongoing collaboration among diverse stakeholders including decision-makers, researchers, faculty, facilities staff, students, and community members.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Vigilância em Saúde Pública , Universidades , Águas Residuárias
14.
J Exp Med ; 196(1): 135-40, 2002 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12093878

RESUMO

Mice lacking the membrane tyrosine kinase c-mer have been shown to have altered macro-phage cytokine production and defective phagocytosis of apoptotic cells despite normal phagocytosis of other particles. We show here that c-mer-deficient mice have impaired clearance of infused apoptotic cells and that they develop progressive lupus-like autoimmunity, with antibodies to chromatin, DNA, and IgG. The autoimmunity appears to be driven by endogenous antigens, with little polyclonal B cell activation. These mice should be an excellent model for studying the role of apoptotic debris as an immunogenic stimulus for systemic autoimmunity.


Assuntos
Apoptose/imunologia , Autoimunidade/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Proteínas Tirosina Quinases/deficiência , Proteínas Proto-Oncogênicas/deficiência , Receptores Proteína Tirosina Quinases , Animais , Autoanticorpos/sangue , Linfócitos B/imunologia , Cardiolipinas/imunologia , Cromatina/imunologia , DNA/imunologia , Modelos Animais de Doenças , Feminino , Corantes Fluorescentes , Mesângio Glomerular/patologia , Imunoglobulina G/imunologia , Lúpus Eritematoso Sistêmico/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fagocitose/imunologia , Proteínas Tirosina Quinases/genética , Proteinúria/complicações , Proteinúria/patologia , Proteínas Proto-Oncogênicas/genética , Fator Reumatoide/sangue , Rodaminas , c-Mer Tirosina Quinase
15.
Circ Res ; 103(12): 1430-40, 2008 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-19008476

RESUMO

Congenital heart defects occur at a rate of 5% and are the most prevalent birth defects. A better understanding of the complex signaling networks regulating heart development is necessary to improve repair strategies for congenital heart defects. The mitogen-activated protein 3 kinase (MEKK3) is important to early embryogenesis, but developmental processes affected by MEKK3 during heart morphogenesis have not been fully examined. We identify MEKK3 as a critical signaling molecule during endocardial cushion development. We report the detection of MEKK3 transcripts to embryonic hearts before, during, and after cardiac cushion cells have executed epithelial-to-mesenchymal transition (EMT). MEKK3 is observed to endocardial cells of the cardiac cushions with a diminishing gradient of expression into the cushions. These observations suggest that MEKK3 may function during production of cushion mesenchyme as required for valvular development and septation of the heart. We used a kinase inactive form of MEKK3 (MEKK3(KI)) in an in vitro assay that recapitulates in vivo EMT and show that MEKK3(KI) attenuates mesenchyme formation. Conversely, constitutively active MEKK3 (ca-MEKK3) triggers mesenchyme production in ventricular endocardium, a tissue that does not normally undergo EMT. MEKK3-driven mesenchyme production is further substantiated by increased expression of EMT-relevant genes, including TGFbeta(2), Has2, and periostin. Furthermore, we show that MEKK3 stimulates EMT via a TGFbeta(2)-dependent mechanism. Thus, the activity of MEKK3 is sufficient for developmental EMT in the heart. This knowledge provides a basis to understand how MEKK3 integrates signaling cascades activating endocardial cushion EMT.


Assuntos
Coxins Endocárdicos/embriologia , Coxins Endocárdicos/enzimologia , Células Epiteliais/citologia , Células Epiteliais/enzimologia , MAP Quinase Quinase Quinase 3/fisiologia , Mesoderma/embriologia , Morfogênese/fisiologia , Fator de Crescimento Transformador beta2/fisiologia , Animais , Diferenciação Celular/fisiologia , Coxins Endocárdicos/citologia , Coxins Endocárdicos/metabolismo , Células Epiteliais/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , MAP Quinase Quinase Quinase 3/deficiência , MAP Quinase Quinase Quinase 3/genética , MAP Quinase Quinase Quinase 3/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos
16.
Glycobiology ; 19(8): 890-8, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19451547

RESUMO

Hyaluronan (HA) induces changes in cellular behavior that are crucial during both embryonic development and cancer progression. However, the biological effects of varying sizes of HA and the signal transduction mechanisms that these polymers may activate remain unclear. In this study, we demonstrate that pulse stimulation of mouse embryonic fibroblasts with high-molecular-weight (HMW) HA, but not HA of lower molecular sizes, leads to increases in Snail2 protein which are dependent on NFkappaB activity. Involvement of CD44, the main HA receptor, in these responses was determined by use of a CD44 blocking antibody and CD44 siRNA. Both the blockade and silencing of CD44 significantly abrogate the increases in nuclear factor kappaB (NFkappaB) activity and Snail2 protein following HMW-HA stimulation. Furthermore, we show that HMW-HA induces cellular invasion and that inhibition of CD44, Snail2, or NFkappaB significantly decreases this response. These studies elucidate a novel HA/Snail2 functional connection through CD44 and NFkappaB that is important for the induction of cellular invasion and is dependent on HA size.


Assuntos
Movimento Celular/fisiologia , Ácido Hialurônico/metabolismo , Fatores de Transcrição/metabolismo , Animais , Fibroblastos/metabolismo , Receptores de Hialuronatos/metabolismo , Camundongos , NF-kappa B/metabolismo , Células NIH 3T3 , Transdução de Sinais/fisiologia , Fatores de Transcrição da Família Snail
17.
J Cell Biol ; 160(4): 605-15, 2003 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-12591918

RESUMO

Atrioventricular (AV) septal defects resulting from aberrant endocardial cushion (EC) formation are observed at increased rates in infants of diabetic mothers. EC formation occurs via an epithelial-mesenchymal transformation (EMT), involving transformation of endocardial cells into mesenchymal cells, migration, and invasion into extracellular matrix. Here, we report that elevated glucose inhibits EMT by reducing myocardial vascular endothelial growth factor A (VEGF-A). This effect is reversed with exogenous recombinant mouse VEGF-A165, whereas addition of soluble VEGF receptor-1 blocks EMT. We show that disruption of EMT is associated with persistence of platelet endothelial cell adhesion molecule-1 (PECAM-1) and decreased matrix metalloproteinase-2 (MMP-2) expression. These findings correlate with retention of a nontransformed endocardial sheet and lack of invasion. The MMP inhibitor GM6001 blocks invasion, whereas explants from PECAM-1 deficient mice exhibit MMP-2 induction and normal EMT in high glucose. PECAM-1-negative endothelial cells are highly motile and express more MMP-2 than do PECAM-1-positive endothelial cells. During EMT, loss of PECAM-1 similarly promotes single cell motility and MMP-2 expression. Our findings suggest that high glucose-induced inhibition of AV cushion morphogenesis results from decreased myocardial VEGF-A expression and is, in part, mediated by persistent endocardial cell PECAM-1 expression and failure to up-regulate MMP-2 expression.


Assuntos
Indutores da Angiogênese/metabolismo , Glucose/metabolismo , Coração/embriologia , Metaloproteinase 2 da Matriz/metabolismo , Morfogênese , Miocárdio/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Fator A de Crescimento do Endotélio Vascular , Indutores da Angiogênese/genética , Animais , Movimento Celular/fisiologia , Tamanho Celular , Células Cultivadas , Técnicas de Cultura , Dipeptídeos/metabolismo , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Comunicação Atrioventricular , Feminino , Masculino , Metaloproteinase 2 da Matriz/genética , Camundongos , Miocárdio/citologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Inibidores de Proteases/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
18.
Mini Rev Med Chem ; 8(7): 669-76, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18537722

RESUMO

The possibility to make precise modifications to the genome at high frequency holds tremendous potential for biotechnology, conventional drug development and gene therapy. Homologous recombination is a powerful method for introducing such modifications in organisms such as mice. However, in mammals and plants, the frequency of gene modification by homologous recombination is quite low, precluding the therapeutic use of this methodology. In the past few years, tremendous progress has been made in overcoming one of primary barriers to efficient recombination, namely the introduction of a targeted double-strand break near the intended recombination site. This review will discuss the advances in engineering custom zinc-finger nucleases and their application in stimulating homologous recombination in higher eukaryotic cells at efficiencies approaching 1 in 2 cells.


Assuntos
Desoxirribonucleases/química , Recombinação Genética , Dedos de Zinco , Animais , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Desoxirribonucleases/genética , Desoxirribonucleases/metabolismo , Marcação de Genes , Terapia Genética , Genoma , Humanos , Camundongos , Mutagênese , Engenharia de Proteínas , Transgenes
19.
Circ Res ; 99(6): 583-9, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16931798

RESUMO

Cardiac malformations constitute the most common birth defects, of which heart septal and valve defects are the most frequent forms diagnosed in infancy. These cardiac structures arise from the endocardial cushions through dynamic interactions between cells and the extracellular matrix (cardiac jelly). Targeted deletion of the hyaluronan synthase-2 (Has2) gene in mice results in an absence of hyaluronan (HA), cardiac jelly, and endocardial cushions, a loss of vascular integrity, and death at embryonic day 9.5. Despite the requirements for Has2 and its product, HA, in the developing heart, little is known about the normal processing and removal of HA during development. Cell culture studies show that HA obtains new bioactivity after depolymerization into small oligosaccharides. We previously showed reduction in Has2 expression and diminished presence of HA at later stages of heart development as tissue remodeling formed the leaflets of the cardiac valves. Here we show that small oligosaccharide forms of HA (o-HA) act antagonistically to developmental epithelial-to-mesenchymal transformation (EMT), which is required to generate the progenitor cells that populate the endocardial cushions. We further show that o-HA induces vascular endothelial growth factor (VEGF), which acts as a negative regulator of EMT. This is the first report illustrating a functional link between oligosaccharide HA and VEGF. Collectively, our data indicate that following endocardial cell EMT, native HA is likely processed to o-HA, which stimulates VEGF activity to attenuate cardiac developmental EMT.


Assuntos
Indução Embrionária , Células Endoteliais/citologia , Epitélio/embriologia , Coração/embriologia , Ácido Hialurônico/farmacologia , Mesoderma/citologia , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Células Cultivadas , Embrião de Galinha , Embrião de Mamíferos , Regulação da Expressão Gênica , Coração/crescimento & desenvolvimento , Ácido Hialurônico/metabolismo , Camundongos , Oligossacarídeos/farmacologia
20.
Toxicol Pathol ; 36(6): 805-17, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18812580

RESUMO

Chronic ingestion of arsenic is associated with increased incidence of respiratory and cardiovascular diseases. To investigate the role of arsenic in early events in vascular pathology, C57BL/6 mice ingested drinking water with or without 50 ppb sodium arsenite (AsIII) for four, five, or eight weeks. At five and eight weeks, RNA from the lungs of control and AsIII-exposed animals was processed for microarray. Sixty-five genes were significantly and differentially expressed. Differential expression of extracellular matrix (ECM) gene transcripts was particularly compelling, as 91% of genes in this category, including elastin and collagen, were significantly decreased. In additional experiments, real-time RT-PCR showed an AsIII-induced decrease in many of these ECM gene transcripts in the heart and NIH3T3 fibroblast cells. Histological stains for collagen and elastin show a distinct disruption in the ECM surrounding small arteries in the heart and lung of AsIII-exposed mice. Immunohistochemical detection of alpha-smooth muscle actin in blood vessel walls was decreased in the AsIII-exposed animals. These data reveal a functional link between AsIII exposure and disruption in the vascular ECM. These AsIII-induced early pathological events may predispose humans to respiratory and cardiovascular diseases linked to chronic low-dose AsIII exposure.


Assuntos
Arsenitos/toxicidade , Vasos Sanguíneos/efeitos dos fármacos , Proteínas da Matriz Extracelular/genética , Expressão Gênica/efeitos dos fármacos , Miocárdio/patologia , Compostos de Sódio/toxicidade , Animais , Arsênio/toxicidade , Arsenitos/administração & dosagem , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Colágeno/genética , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/patologia , Elastina/genética , Proteínas de Choque Térmico/genética , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Compostos de Sódio/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA