Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-33922263

ABSTRACT

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.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Public Health Surveillance , Universities , Wastewater
2.
medRxiv ; 2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33564791

ABSTRACT

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.

3.
Crit Rev Oncol Hematol ; 126: 186-200, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29759560

ABSTRACT

Cardiotoxic effects of chemotherapy and targeted drugs are ubiquitous and challenging in the field of oncology therapeutics. The broad spectrum of toxicities ranging from ischemic, hypertensive, cardiomyopathic, and arrhythmic complications can present as a significant challenge for clinicians treating cancer patients. If early diagnosis and intervention of cardiotoxic complications is missed, this can lead to delay or abrogation of planned treatment, which can potentially culminate to significant morbidity due to not only the cardiotoxic complications but also the progression of cancer. Hence, full knowledge of cardiovascular complications of chemotherapeutic agents, essential diagnostics tests to order, and appropriate management is paramount to oncologist, oncology pharmacists, and scientific clinical investigators. The aforementioned is particularly true in the current oncology era of plenteous early clinical trials studying several pathway/molecular-targeting agents with an increased cardiotoxic potential and the rapid expedited approval of those drugs by the FDA. Herein, we present a review discussing cardiotoxic effects of drugs and guidelines for management of the toxicities to assist the medical field in general managing patients with cancer.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiotoxicity/epidemiology , Cardiovascular System/drug effects , Molecular Targeted Therapy/adverse effects , Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Cardiotoxicity/etiology , Drug Delivery Systems , Heart Diseases/chemically induced , Heart Diseases/epidemiology , Humans , Medical Oncology/methods , Medical Oncology/trends , Molecular Targeted Therapy/statistics & numerical data , Neoplasms/epidemiology
4.
Toxicol Sci ; 153(2): 271-81, 2016 10.
Article in English | MEDLINE | ID: mdl-27413109

ABSTRACT

Synucleinopathies, including Parkinson's disease (PD), are neurodegenerative diseases characterized by accumulation of α-synuclein (SYN), a small neuronal protein with prion like properties that plays a central role in PD pathogenesis. SYN can misfold and generate toxic oligomers/aggregates, which can be cytotoxic. Environmental arsenic (As)-containing pesticide use correlates with increased incidence of PD. Moreover, because As exposure can lead to inhibition of autophagic flux we hypothesize that As can facilitate the accumulation of toxic SYN oligomers/aggregates and subsequent increases in markers of autophagy. We therefore examined the role of As in the oligomerization of SYN, and the consequences thereof. Chronic exposure of SH-SY5Y cells overexpressing SYN to As caused a dose-dependent oligomerization of SYN, with concomitant increases in protein ubiquitination and expression of other stress markers (protein glutathione binding, γ-GCS, light chain 3 (LC3)-I/II, P62, and NAD(P)H dehydrogenase quinone 1), indicative of an increased proteotoxic stress. Immunocytochemical analyses revealed an accumulation of SYN, and it's colocalization with LC3, a major autophagic protein. Mice exposed to As (100 ppb) for 1 month, exhibited elevated SYN accumulation in the cortex and striatum, and elevations in protein ubiquitination and LC3-I and II levels. However, tyrosine hydroxylase (TH), an indicator of dopaminergic cell density, was upregulated in the As exposed animals. Because SYN can inhibit TH function, and As can decrease monoamine levels, As exposure possibly leads to compensatory mechanisms leading to an increase in TH expression. Our findings suggest that susceptible individuals may be at higher risk of developing synucleinopathies and/or neurodegeneration due to environmental As exposure.


Subject(s)
Arsenic/pharmacology , Neurodegenerative Diseases/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Animals , Cell Line , Female , Mice
5.
J Biochem Mol Toxicol ; 30(7): 321-30, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26890134

ABSTRACT

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.


Subject(s)
Arsenites/toxicity , Diet, High-Fat/adverse effects , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Liver/drug effects , Non-alcoholic Fatty Liver Disease/metabolism , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Biological Transport/drug effects , Embryo, Mammalian , Female , Fetus , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/metabolism , Liver/pathology , Liver-Specific Organic Anion Transporter 1 , Male , Mice , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Pregnancy , Primary Cell Culture , Signal Transduction
6.
Drug Chem Toxicol ; 39(3): 279-83, 2016.
Article in English | MEDLINE | ID: mdl-26446802

ABSTRACT

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.


Subject(s)
Arsenites/metabolism , Heart , Liver/metabolism , Lung/metabolism , Myocardium/metabolism , Sodium Compounds/metabolism , Animals , Arsenites/toxicity , Biotransformation , Female , Gene Expression/drug effects , Heart/drug effects , Heart/embryology , Liver/drug effects , Liver/embryology , Lung/drug effects , Lung/embryology , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Organ Culture Techniques , Sodium Compounds/toxicity
7.
Cell Adh Migr ; 10(3): 259-68, 2016 05 03.
Article in English | MEDLINE | ID: mdl-26645362

ABSTRACT

During embryogenesis, the epicardium undergoes proliferation, migration, and differentiation into several cardiac cell types which contribute to the coronary vessels. The type III transforming growth factor-ß receptor (TGFßR3) is required for epicardial cell invasion and development of coronary vasculature in vivo. Bone Morphogenic Protein-2 (BMP2) is a driver of epicardial cell migration. Utilizing a primary epicardial cell line derived from Tgfbr3(+/+) and Tgfbr3(-/-) mouse embryos, we show that Tgfbr3(-/-) epicardial cells are deficient in BMP2 mRNA expression. Tgfbr3(-/-) epicardial cells are deficient in 2-dimensional migration relative to Tgfbr3(+/+) cells; BMP2 induces cellular migration to Tgfbr3(+/+) levels without affecting proliferation. We further demonstrate that Src kinase activity is required for BMP2 driven Tgfbr3(-/-) migration. BMP2 also requires Src for filamentous actin polymerization in Tgfbr3(-/-) epicardial cells. Taken together, our data identifies a novel pathway in epicardial cell migration required for development of the coronary vessels.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Cell Movement/drug effects , Pericardium/cytology , Proteoglycans/deficiency , Receptors, Transforming Growth Factor beta/deficiency , src-Family Kinases/metabolism , Actins/metabolism , Animals , Cell Movement/genetics , Cell Proliferation/drug effects , Gene Expression Profiling , Humans , Mice , Polymerization , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism
8.
Environ Health Perspect ; 124(2): 201-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26151952

ABSTRACT

BACKGROUND: Chronic exposure to arsenicals at various life stages and across a range of exposures has been implicated in cardiometabolic and liver disease, but disease predisposition from developmental exposures remains unclear. OBJECTIVES: In utero and post-weaning exposure to trivalent arsenic (AsIII) was examined on the background of a Western-style diet to determine whether AsIII exposure affects metabolic disease. METHODS: Male Swiss Webster mice were exposed to 100 ppb AsIII in utero, after weaning, or both. Ad libitum access to a Western-style diet was provided after weaning, and the plasma metabolome, liver histopathology, liver enzyme activity, and gene expression were analyzed. RESULTS: Hepatic lipid composition and histopathology revealed that developmental AsIII exposure exacerbated Western-style diet-induced fatty liver disease. Continuous AsIII exposure increased cardiometabolic risk factors including increased body weight, insulin resistance, hyperglycemia, and plasma triglycerides. AsIII exposure produced a decrease in the intermediates of glycolysis and the TCA cycle while increasing ketones. Hepatic isocitrate dehydrogenase activity was also decreased, which confirmed disruption of the TCA cycle. Developmental AsIII exposure increased the expression of genes involved in fatty acid synthesis, lipogenesis, inflammation, and packaging of triglycerides, suggesting an increased acetyl coenzyme A (acetyl-CoA) load. CONCLUSIONS: In utero and continuous early-life exposure to AsIII disrupted normal metabolism and elevated the risk for fatty liver disease in mice maintained on a high-fat diet. Our findings suggest that individuals exposed to AsIII during key developmental periods and who remain exposed to AsIII on the background of a Western-style diet may be at increased risk for metabolic disease later in life.


Subject(s)
Arsenites/toxicity , Diet, Western/adverse effects , Disease Susceptibility/epidemiology , Energy Metabolism , Fatty Liver/epidemiology , Metabolic Diseases/epidemiology , Prenatal Exposure Delayed Effects/metabolism , Animals , Disease Susceptibility/etiology , Fatty Liver/etiology , Female , Male , Metabolic Diseases/etiology , Mice , Pregnancy
9.
Toxicol Sci ; 148(2): 409-20, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26354774

ABSTRACT

TGFß2 (transforming growth factor-ß2) is a key growth factor regulating epithelial to mesenchymal transition (EMT). TGFß2 triggers cardiac progenitor cells to differentiate into mesenchymal cells and give rise to the cellular components of coronary vessels as well as cells of aortic and pulmonary valves. TGFß signaling is dependent on a dynamic on and off switch in Smad activity. Arsenite exposure of 1.34 µM for 24-48 h has been reported to disrupt Smad phosphorylation leading to deficits in TGFß2-mediated cardiac precursor differentiation and transformation. In this study, the molecular mechanism of acute arsenite toxicity on TGFß2-induced Smad2/3 nuclear shuttling and TGFß2-mediated cardiac EMT was investigated. A 4-h exposure to 5 µM arsenite blocks nuclear accumulation of Smad2/3 in response to TGFß2 without disrupting Smad phosphorylation or nuclear importation. The depletion of nuclear Smad is restored by knocking-down Smad-specific exportins, suggesting that arsenite augments Smad2/3 nuclear exportation. The blockage in TGFß2-Smad signaling is likely due to the loss of Zn(2+) cofactor in Smad proteins, as Zn(2+) supplementation reverses the disruption in Smad2/3 nuclear translocation and transcriptional activity by arsenite. This coincides with Zn(2+) supplementation rescuing arsenite-mediated deficits in cardiac EMT. Thus, zinc partially protects cardiac EMT from developmental toxicity by arsenite.


Subject(s)
Arsenites/toxicity , Cell Differentiation/drug effects , Myocytes, Cardiac/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Stem Cells/drug effects , Transforming Growth Factor beta2/pharmacology , Zinc/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Epithelial-Mesenchymal Transition/drug effects , HEK293 Cells , Humans , Karyopherins/genetics , Karyopherins/metabolism , Mice, Transgenic , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation , RNA Interference , Signal Transduction/drug effects , Stem Cells/metabolism , Stem Cells/pathology , Time Factors , Transcription, Genetic/drug effects , Transfection
10.
Cell Signal ; 27(3): 453-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25499979

ABSTRACT

During embryogenesis, the epicardium undergoes proliferation, migration, and differentiation into several cardiac cell types which contribute to the coronary vessels. This process requires epithelial to mesenchymal transition (EMT) and directed cellular invasion. The Type III Transforming Growth Factor-beta Receptor (TGFßR3) is required for epicardial cell invasion and coronary vessel development. Using primary epicardial cells derived from Tgfbr3(+/+) and Tgfbr3(-/-) mouse embryos, high-molecular weight hyaluronan (HMWHA) stimulated cellular invasion and filamentous (f-actin) polymerization are detected in Tgfbr3(+/+) cells, but not in Tgfbr3(-/-) cells. Furthermore, HMWHA-stimulated cellular invasion and f-actin polymerization in Tgfbr3(+/+) epicardial cells are dependent on Src kinase. Src activation in HMWHA-stimulated Tgfbr3(-/-) epicardial cells is not detected in response to HMWHA. RhoA and Rac1 also fail to activate in response to HMWHA in Tgfbr3(-/-) cells. These events coincide with defective f-actin formation and deficient cellular invasion. Finally, a T841A activating substitution in TGFßR3 drives ligand-independent Src activation. Collectively, these data define a TGFßR3-Src-RhoA/Rac1 pathway that is essential for hyaluronan-directed cell invasion in epicardial cells.


Subject(s)
Hyaluronic Acid/pharmacology , Pericardium/drug effects , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , src-Family Kinases/metabolism , Actin Cytoskeleton/drug effects , Amino Acid Substitution , Animals , Arrestin/chemistry , Arrestin/metabolism , Cell Movement/drug effects , Cells, Cultured , Epithelial-Mesenchymal Transition , Mice , Neuropeptides/metabolism , Pericardium/cytology , Pericardium/metabolism , Protein Binding , Proteoglycans/antagonists & inhibitors , Proteoglycans/genetics , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/genetics , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
11.
Toxicol Sci ; 142(1): 225-38, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25145660

ABSTRACT

Arsenic exposure during embryonic development can cause ischemic heart pathologies later in adulthood which may originate from impairment in proper blood vessel formation. The arsenic-associated detrimental effects are mediated by arsenite (iAs(III)) and its most toxic metabolite, monomethylarsonous acid [MMA (III)]. The impact of MMA (III) on coronary artery development has not yet been studied. The key cellular process that regulates coronary vessel development is the epithelial-mesenchymal transition (EMT). During cardiac EMT, activated epicardial progenitor cells transform to mesenchymal cells to form the cellular components of coronary vessels. Smad2/3 mediated TGFß2 signaling, the key regulator of cardiac EMT, is disrupted by arsenite exposure. In this study, we compared the cardiac toxicity of MMA (III) with arsenite. Epicardial progenitor cells are 15 times more sensitive to MMA (III) cytotoxicity when compared with arsenite. MMA (III) caused a significant blockage in epicardial cellular transformation and invasion at doses 10 times lower than arsenite. Key EMT genes including TGFß ligands, TßRIII, Has2, CD44, Snail1, TBX18, and MMP2 were down regulated by MMA (III) exposure. MMA (III) disrupted Smad2/3 activation at a dose 20 times lower than arsenite. Both arsenite and MMA (III) significantly inhibited Erk1/2 and Erk5 phosphorylation. Nuclear translocation of Smad2/3 and Erk5 was also blocked by arsenical exposure. However, p38 activation, as well as smooth muscle differentiation, was refractory to the inhibition by the arsenicals. Collectively, these findings revealed that MMA (III) is a selective disruptor of cardiac EMT and as such may predispose to arsenic-associated cardiovascular disorders.


Subject(s)
Arsenites/toxicity , Coronary Vessels/drug effects , Epithelial-Mesenchymal Transition/drug effects , Organometallic Compounds/toxicity , Pericardium/drug effects , Animals , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Coronary Vessels/embryology , Coronary Vessels/metabolism , Coronary Vessels/pathology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Developmental/drug effects , Mice, Transgenic , Organogenesis/drug effects , Organogenesis/genetics , Pericardium/embryology , Pericardium/metabolism , Pericardium/pathology , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/pathology
12.
Cell Signal ; 26(1): 70-82, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24036211

ABSTRACT

Human MAP3K4 (MTK1) functions upstream of mitogen activated protein kinases (MAPKs). In this study we show MTK1 is required for human epidermal growth factor receptor 2/3 (HER2/HER3)-heregulin beta1 (HRG) induced cell migration in MCF-7 breast cancer cells. We demonstrate that HRG stimulation leads to association of MTK1 with activated HER3 in MCF-7 and T-47D breast cancer cells. Activated HER3 association with MTK1 is dependent on HER2 activation and is decreased by pre-treatment with the HER2 inhibitor, lapatinib. Moreover, we also identify the actin interacting region (AIR) on MTK1. Disruption of actin cytoskeletal polymerization with cytochalasin D inhibited HRG induced MTK1/HER3 association. Additionally, HRG stimulation leads to extracellular acidification that is independent of cellular proliferation. HRG induced extracellular acidification is significantly inhibited when MTK1 is knocked down in MCF-7 cells. Similarly, pre-treatment with lapatinib significantly decreased HRG induced extracellular acidification. Extracellular acidification is linked with cancer cell migration. We performed scratch assays that show HRG induced cell migration in MCF-7 cells. Knockdown of MTK1 significantly inhibited HRG induced cell migration. Furthermore, pre-treatment with lapatinib also significantly decreased cell migration. Cell migration is required for cancer cell metastasis, which is the major cause of cancer patient mortality. We identify MTK1 in the HER2/HER3-HRG mediated extracellular acidification and cell migration pathway in breast cancer cells.


Subject(s)
Acids/metabolism , Cell Movement , Extracellular Space/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actins/metabolism , Amino Acid Sequence , Cell Movement/drug effects , Extracellular Space/drug effects , Female , Gene Knockdown Techniques , Humans , Immunoprecipitation , MAP Kinase Kinase Kinase 4/chemistry , MAP Kinase Kinase Kinase 4/metabolism , MCF-7 Cells , Molecular Sequence Data , Molecular Weight , Neuregulin-1/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Binding/drug effects , Protein Structure, Tertiary
13.
Toxicol Appl Pharmacol ; 272(1): 147-53, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23732083

ABSTRACT

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.


Subject(s)
Arsenites/toxicity , Coronary Vessels/cytology , Epithelial-Mesenchymal Transition/drug effects , Signal Transduction/drug effects , Stem Cells/drug effects , Transforming Growth Factor beta/physiology , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Coronary Vessels/drug effects , Fluorescent Antibody Technique , Indicators and Reagents , Mesenchymal Stem Cells/drug effects , Mice , Microfilament Proteins/genetics , Muscle Proteins/genetics , Myocytes, Smooth Muscle/drug effects , Smad Proteins/metabolism
14.
Toxicol Appl Pharmacol ; 261(3): 263-70, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22521605

ABSTRACT

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.


Subject(s)
Arsenites/pharmacology , C-Reactive Protein/physiology , NF-kappa B/metabolism , Amides/pharmacology , Animals , Biomarkers/analysis , Biotransformation/drug effects , Blotting, Western , C-Reactive Protein/analysis , C-Reactive Protein/biosynthesis , Cell Line , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , L-Lactate Dehydrogenase/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Luciferases/metabolism , Mice , NF-kappa B/genetics , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Reactive Oxygen Species/metabolism , Transfection , rho-Associated Kinases/antagonists & inhibitors
15.
Toxicol Pathol ; 40(3): 504-12, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22215511

ABSTRACT

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.


Subject(s)
Arsenites/toxicity , Blood Pressure/drug effects , Hypertension/chemically induced , Hypertrophy, Left Ventricular/chemically induced , Sodium Compounds/toxicity , Administration, Oral , Analysis of Variance , Animals , Arsenites/administration & dosage , Drinking Water/standards , Echocardiography , Female , Histocytochemistry , Hypertrophy, Left Ventricular/pathology , Mice , Sodium Compounds/administration & dosage , Toxicity Tests, Chronic
16.
Cell Tissue Res ; 347(1): 203-23, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21953136

ABSTRACT

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.


Subject(s)
Cardiac Rehabilitation , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Aging/physiology , Angiotensin II/metabolism , Animals , Cardiovascular Diseases/therapy , Epithelial-Mesenchymal Transition/physiology , Gene Expression , Genetic Variation , Humans , Mitogen-Activated Protein Kinases/metabolism , Mutation , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Smad Proteins/metabolism
17.
Dev Biol ; 358(2): 331-43, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21871877

ABSTRACT

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.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Neuropeptides/chemistry , Neuropeptides/metabolism , Pericardium/cytology , Pericardium/metabolism , Proteoglycans/chemistry , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/chemistry , Receptors, Transforming Growth Factor beta/metabolism , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Coronary Vessel Anomalies/embryology , Coronary Vessel Anomalies/genetics , Coronary Vessel Anomalies/metabolism , DNA Primers/genetics , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Models, Cardiovascular , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Neuropeptides/antagonists & inhibitors , Neuropeptides/genetics , Pericardium/embryology , Pregnancy , Protein Interaction Domains and Motifs , Proteoglycans/deficiency , Proteoglycans/genetics , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Time-Lapse Imaging , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta2/pharmacology
18.
Dev Dyn ; 240(9): 2127-41, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21780244

ABSTRACT

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.


Subject(s)
Heart Valves/metabolism , Heart/embryology , Transforming Growth Factor beta2/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Extracellular Matrix/metabolism , Heart Valves/embryology , Immunohistochemistry , Mesoderm/cytology , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta2/genetics
19.
Semin Cell Dev Biol ; 21(9): 929-35, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20933094

ABSTRACT

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.


Subject(s)
ErbB Receptors/metabolism , Heart Diseases/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction , Animals , ErbB Receptors/genetics , Humans
20.
Exp Cell Res ; 316(20): 3397-405, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20633555

ABSTRACT

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.


Subject(s)
Cell Movement/physiology , Epithelial-Mesenchymal Transition/physiology , Hyaluronic Acid/metabolism , Pericardium/cytology , Stem Cells/cytology , Transforming Growth Factor beta2/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Line , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression/drug effects , Gene Expression/genetics , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Hyaluronan Receptors/immunology , Hyaluronan Synthases , Hyaluronoglucosaminidase/pharmacology , MAP Kinase Kinase Kinase 3/genetics , MAP Kinase Kinase Kinase 3/metabolism , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Phosphorylation/drug effects , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Stem Cells/drug effects , Stem Cells/metabolism , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...