Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Mol Cancer Res ; 20(3): 373-386, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34753803

ABSTRACT

MALT1 is the effector protein of the CARMA/Bcl10/MALT1 (CBM) signalosome, a multiprotein complex that drives pro-inflammatory signaling pathways downstream of a diverse set of receptors. Although CBM activity is best known for its role in immune cells, emerging evidence suggests that it plays a key role in the pathogenesis of solid tumors, where it can be activated by selected G protein-coupled receptors (GPCR). Here, we demonstrated that overexpression of GPCRs implicated in breast cancer pathogenesis, specifically the receptors for Angiotensin II and thrombin (AT1R and PAR1), drove a strong epithelial-to-mesenchymal transition (EMT) program in breast cancer cells that is characteristic of claudin-low, triple-negative breast cancer (TNBC). In concert, MALT1 was activated in these cells and contributed to the dramatic EMT phenotypic changes through regulation of master EMT transcription factors including Snail and ZEB1. Importantly, blocking MALT1 signaling, through either siRNA-mediated depletion of MALT1 protein or pharmacologic inhibition of its activity, was effective at partially reversing the molecular and phenotypic indicators of EMT. Treatment of mice with mepazine, a pharmacologic MALT1 inhibitor, reduced growth of PAR1+, MDA-MB-231 xenografts and had an even more dramatic effect in reducing the burden of metastatic disease. These findings highlight MALT1 as an attractive therapeutic target for claudin-low TNBCs harboring overexpression of one or more selected GPCRs. IMPLICATIONS: This study nominates a GPCR/MALT1 signaling axis as a pathway that can be pharmaceutically targeted to abrogate EMT and metastatic progression in TNBC, an aggressive form of breast cancer that currently lacks targeted therapies.


Subject(s)
Triple Negative Breast Neoplasms , Animals , Cell Line, Tumor , Cell Movement , Claudins/pharmacology , Claudins/therapeutic use , Epithelial-Mesenchymal Transition , Humans , Mice , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Receptor, PAR-1/therapeutic use , Triple Negative Breast Neoplasms/metabolism
2.
J Immunol ; 204(9): 2337-2348, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32213560

ABSTRACT

The signaling protein MALT1 plays a key role in promoting NF-κB activation in Ag-stimulated lymphocytes. In this capacity, MALT1 has two functions, acting as a scaffolding protein and as a substrate-specific protease. MALT1 is also required for NF-κB-dependent induction of proinflammatory cytokines after FcεR1 stimulation in mast cells, implicating a role in allergy. Because MALT1 remains understudied in this context, we sought to investigate how MALT1 proteolytic activity contributes to the overall allergic response. We compared bone marrow-derived mast cells from MALT1 knockout (MALT1-/-) and MALT1 protease-deficient (MALTPD/PD) mice to wild-type cells. We found that MALT1-/- and MALT1PD/PD mast cells are equally impaired in cytokine production following FcεRI stimulation, indicating that MALT1 scaffolding activity is insufficient to drive the cytokine response and that MALT1 protease activity is essential. In addition to cytokine production, acute mast cell degranulation is a critical component of allergic response. Intriguingly, whereas degranulation is MALT1-independent, MALT1PD/PD mice are protected from vascular edema induced by either passive cutaneous anaphylaxis or direct challenge with histamine, a major granule component. This suggests a role for MALT1 protease activity in endothelial cells targeted by mast cell-derived vasoactive substances. Indeed, we find that in human endothelial cells, MALT1 protease is activated following histamine treatment and is required for histamine-induced permeability. We thus propose a dual role for MALT1 protease in allergic response, mediating 1) IgE-dependent mast cell cytokine production, and 2) histamine-induced endothelial permeability. This dual role indicates that therapeutic inhibitors of MALT1 protease could work synergistically to control IgE-mediated allergic disease.


Subject(s)
Endothelial Cells/metabolism , Hypersensitivity/metabolism , Mast Cells/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Line , Cytokines/immunology , Cytokines/metabolism , Endothelial Cells/immunology , Female , Histamine/immunology , Humans , Hypersensitivity/immunology , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Inflammation/immunology , Inflammation/metabolism , Lymphocyte Activation/immunology , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/immunology , NF-kappa B/immunology , NF-kappa B/metabolism , Receptors, IgE/immunology , Receptors, IgE/metabolism
3.
J Clin Invest ; 130(2): 1036-1051, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31961340

ABSTRACT

Antigen receptor-dependent (AgR-dependent) stimulation of the NF-κB transcription factor in lymphocytes is a required event during adaptive immune response, but dysregulated activation of this signaling pathway can lead to lymphoma. AgR stimulation promotes assembly of the CARMA1-BCL10-MALT1 complex, wherein MALT1 acts as (a) a scaffold to recruit components of the canonical NF-κB machinery and (b) a protease to cleave and inactivate specific substrates, including negative regulators of NF-κB. In multiple lymphoma subtypes, malignant B cells hijack AgR signaling pathways to promote their own growth and survival, and inhibiting MALT1 reduces the viability and growth of these tumors. As such, MALT1 has emerged as a potential pharmaceutical target. Here, we identified G protein-coupled receptor kinase 2 (GRK2) as a new MALT1-interacting protein. We demonstrated that GRK2 binds the death domain of MALT1 and inhibits MALT1 scaffolding and proteolytic activities. We found that lower GRK2 levels in activated B cell-type diffuse large B cell lymphoma (ABC-DLBCL) are associated with reduced survival, and that GRK2 knockdown enhances ABC-DLBCL tumor growth in vitro and in vivo. Together, our findings suggest that GRK2 can function as a tumor suppressor by inhibiting MALT1 and provide a roadmap for developing new strategies to inhibit MALT1-dependent lymphomagenesis.


Subject(s)
Carcinogenesis/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Oncogene Proteins/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Female , G-Protein-Coupled Receptor Kinase 2/genetics , Humans , Jurkat Cells , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Inbred NOD , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Oncogene Proteins/genetics
4.
Oncogene ; 38(49): 7384-7398, 2019 12.
Article in English | MEDLINE | ID: mdl-31420608

ABSTRACT

Protease-activated receptor 1 (PAR1), a thrombin-responsive G protein-coupled receptor (GPCR), is implicated in promoting metastasis in multiple tumor types, including both sarcomas and carcinomas, but the molecular mechanisms responsible remain largely unknown. We previously discovered that PAR1 stimulation in endothelial cells leads to activation of NF-κB, mediated by a protein complex comprised of CARMA3, Bcl10, and the MALT1 effector protein (CBM complex). Given the strong association between NF-κB and metastasis, we hypothesized that this CBM complex could play a critical role in the PAR1-driven metastatic progression of specific solid tumors. In support of our hypothesis, we demonstrate that PAR1 stimulation results in NF-κB activation in both osteosarcoma and breast cancer, which is suppressed by siRNA-mediated MALT1 knockdown, suggesting that an intact CBM complex is required for the response in both tumor cell types. We identify several metastasis-associated genes that are upregulated in a MALT1-dependent manner after PAR1 stimulation in cancer cells, including those encoding the matrix remodeling protein, MMP9, and the cytokines, IL-1ß and IL-8. Further, exogenous expression of PAR1 in MCF7 breast cancer cells confers highly invasive and metastatic behavior which can be blocked by CRISPR/Cas9-mediated MALT1 knockout. Importantly, we find that PAR1 stimulation induces MALT1 protease activity in both osteosarcoma and breast cancer cells, an activity that is mechanistically linked to NF-κB activation and potentially other responses associated with aggressive phenotype. Several small molecule MALT1 protease inhibitors have recently been described that could therefore represent promising new therapeutics for the prevention and/or treatment of PAR1-driven tumor metastasis.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , NF-kappa B/metabolism , Osteosarcoma/pathology , Receptor, PAR-1/metabolism , Animals , Apoptosis , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Movement , Cell Proliferation , Female , Humans , Mice , Mice, Nude , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , NF-kappa B/genetics , Osteosarcoma/genetics , Osteosarcoma/metabolism , Receptor, PAR-1/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Cancer Res ; 78(5): 1225-1240, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29259013

ABSTRACT

The angiotensin II receptor AGTR1, which mediates vasoconstrictive and inflammatory signaling in vascular disease, is overexpressed aberrantly in some breast cancers. In this study, we established the significance of an AGTR1-responsive NFκB signaling pathway in this breast cancer subset. We documented that AGTR1 overexpression occurred in the luminal A and B subtypes of breast cancer, was mutually exclusive of HER2 expression, and correlated with aggressive features that include increased lymph node metastasis, reduced responsiveness to neoadjuvant therapy, and reduced overall survival. Mechanistically, AGTR1 overexpression directed both ligand-independent and ligand-dependent activation of NFκB, mediated by a signaling pathway that requires the triad of CARMA3, Bcl10, and MALT1 (CBM signalosome). Activation of this pathway drove cancer cell-intrinsic responses that include proliferation, migration, and invasion. In addition, CBM-dependent activation of NFκB elicited cancer cell-extrinsic effects, impacting endothelial cells of the tumor microenvironment to promote tumor angiogenesis. CBM/NFκB signaling in AGTR1+ breast cancer therefore conspires to promote aggressive behavior through pleiotropic effects. Overall, our results point to the prognostic and therapeutic value of identifying AGTR1 overexpression in a subset of HER2-negative breast cancers, and they provide a mechanistic rationale to explore the repurposing of drugs that target angiotensin II-dependent NFκB signaling pathways to improve the treatment of this breast cancer subset.Significance: These findings offer a mechanistic rationale to explore the repurposing of drugs that target angiotensin action to improve the treatment of AGTR1-expressing breast cancers. Cancer Res; 78(5); 1225-40. ©2017 AACR.


Subject(s)
B-Cell CLL-Lymphoma 10 Protein/metabolism , Breast Neoplasms/pathology , CARD Signaling Adaptor Proteins/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , NF-kappa B/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, Angiotensin/metabolism , Animals , Apoptosis , B-Cell CLL-Lymphoma 10 Protein/antagonists & inhibitors , B-Cell CLL-Lymphoma 10 Protein/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CARD Signaling Adaptor Proteins/antagonists & inhibitors , CARD Signaling Adaptor Proteins/genetics , Cell Movement , Cell Proliferation , Chick Embryo , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , NF-kappa B/genetics , Neovascularization, Pathologic , Prognosis , RNA, Small Interfering/genetics , Receptor, Angiotensin, Type 1/genetics , Receptors, Angiotensin/chemistry , Receptors, Angiotensin/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Toxicol Lett ; 265: 147-155, 2017 Jan 04.
Article in English | MEDLINE | ID: mdl-27932253

ABSTRACT

Low-dose chronic exposure to arsenic in drinking water represents a global public health concern with established risks for metabolic and cardiovascular disease, as well as cancer. While the linkage between arsenic and disease is strong, further understanding of the molecular mechanisms of its pathogenicity is required. Previous reports demonstrated the ability of arsenic to interfere with adipogenesis, which may mediate its effects in promoting metabolic disease. We hypothesized that microRNA are important regulators of most if not all mesenchymal stem cell processes that are dysregulated by arsenic exposure to impair lipogenesis. Arsenic increased the expression of miR-29b in white adipose tissue, as well as human mesenchymal stem cells (hMSCs) isolated from adipose tissue. Exposing hMSCs to arsenic increased abundance of miR-29b and cyclin D1 to promote proliferation over differentiation. Paradoxically, inhibition of miR-29b enhanced the inhibitory effect of arsenic on differentiation. This paradox was attributed to a requirement for miR-29 in regulating cyclin D1 expression as stable inhibition of miR-29b eliminated the cyclic pattern of cyclin D1 expression. Temporal regulation of cyclin D1 is critical for adipogenic differentiation, and the data suggest a paradigm where arsenic disruption of miR-29b regulatory pathways impairs adipogenic differentiation and ultimately adipose metabolic homeostasis.


Subject(s)
Adipogenesis/drug effects , Arsenites/toxicity , Cyclin D1/genetics , Gene Expression Regulation/drug effects , MicroRNAs/genetics , Water Pollutants, Chemical/toxicity , Adipogenesis/genetics , Animals , Cell Culture Techniques , Cells, Cultured , Humans , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL
7.
Cell Rep ; 17(1): 221-232, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27681433

ABSTRACT

Microvascular endothelial cells maintain a tight barrier to prevent passage of plasma and circulating immune cells into the extravascular tissue compartment, yet endothelial cells respond rapidly to vasoactive substances, including thrombin, allowing transient paracellular permeability. This response is a cornerstone of acute inflammation, but the mechanisms responsible are still incompletely understood. Here, we demonstrate that thrombin triggers MALT1 to proteolytically cleave cylindromatosis (CYLD). Fragmentation of CYLD results in microtubule disruption and a cascade of events leading to endothelial cell retraction and an acute permeability response. This finding reveals an unexpected role for the MALT1 protease, which previously has been viewed mostly as a driver of pro-inflammatory NF-κB signaling in lymphocytes. Thus, MALT1 not only promotes immune cell activation but also acutely regulates endothelial cell biology, actions that together facilitate tissue inflammation. Pharmacologic inhibition of MALT1 may therefore have synergistic impact by targeting multiple disparate steps in the overall inflammatory response.


Subject(s)
Caspases/immunology , Cysteine Endopeptidases/immunology , Endothelial Cells/drug effects , Microtubules/drug effects , Neoplasm Proteins/immunology , Thrombin/pharmacology , Animals , Biological Transport , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/immunology , Caspases/genetics , Cell Line , Cysteine Endopeptidases/genetics , Deubiquitinating Enzyme CYLD , Endothelial Cells/cytology , Endothelial Cells/immunology , Gene Expression Regulation , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Mice , Mice, Transgenic , Microtubules/ultrastructure , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , NF-kappa B/genetics , NF-kappa B/immunology , Neoplasm Proteins/genetics , Permeability/drug effects , Primary Cell Culture , Receptor, PAR-1/genetics , Receptor, PAR-1/immunology , Signal Transduction , Thrombin/metabolism
8.
Toxicol Sci ; 134(2): 335-44, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23650128

ABSTRACT

Arsenic in drinking water promotes a number of diseases that may stem from dysfunctional adipose lipid and glucose metabolism. Arsenic inhibits adipocyte differentiation and promotes insulin resistance; however, little is known of the impacts of and mechanisms for arsenic effects on adipose lipid storage and lipolysis. Based on our earlier studies of arsenic-signaling mechanisms for vascular remodeling and inhibition of adipogenesis, we investigated the hypothesis that arsenic acts through specific adipocyte G-protein-coupled receptors (GPCRs) to promote lipolysis and decrease lipid storage. We first demonstrated that 5-week exposure of mice to 100 µg/l of arsenic in drinking water stimulated epididymal adipocyte hypertrophy, reduced the adipose tissue expression of perilipin (PLIN1, a lipid droplet coat protein), and increased perivascular ectopic fat deposition in skeletal muscle. Incubating adipocytes, differentiated from adipose-derived human mesenchymal stem cell, with arsenic stimulated lipolysis and decreased both Nile Red positive lipid droplets and PLIN1 expression. Arsenic-stimulated lipolysis was not associated with increased cAMP levels. However, preincubation of adipocytes with the Gi inhibitor, Pertussis toxin, attenuated As(III)-stimulated lipolysis and lipid droplet loss. Antagonizing Gi-coupled endothelin-1 type A and B receptors (EDNRA/EDNRB) also attenuated arsenic effects, but antagonizing other adipose Gi-coupled receptors that regulate fat metabolism was ineffective. The endothelin receptors have different roles in arsenic responses because only EDNRA inhibition prevented arsenic-stimulated lipolysis, but antagonists to either receptor protected lipid droplets and PLIN1 expression. These data support a role for specific GPCRs in arsenic signaling for aberrant lipid storage and metabolism that may contribute to the pathogenesis of metabolic disease caused by environmental arsenic exposures.


Subject(s)
Adipose Tissue/drug effects , Arsenic/toxicity , Lipolysis/drug effects , Receptors, G-Protein-Coupled/physiology , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL , Receptor, Endothelin A/drug effects , Reverse Transcriptase Polymerase Chain Reaction
9.
Toxicol Sci ; 131(2): 512-20, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23152186

ABSTRACT

Dysfunctional lipid and glucose metabolism contribute to metabolic syndrome-a major public health concern that enhances cardiovascular disease risk. Arsenic (As(III)) exposure may increase metabolic syndrome and cardiovascular disease risk by impairing adipose tissue differentiation, function, and insulin sensitivity through pathogenic mechanisms that remain unclear. We hypothesized that As(III) signals through the Pertussis toxin (Ptx) sensitive, Gi protein-coupled receptor (GPCR) to impair adipogenesis, as previously demonstrated for its stimulation of vascular oxidant generation, angiogenesis, and remodeling. Because both As(III) and GPCR ligands inhibit progenitor cell differentiation into adipocytes, we investigated the hypothesis in a model of low-passage human mesenchymal stem cells (hMSC). As(III) (0.1-1.0 µM) suppressed dexamethasone/insulin-induced hMSC adipogenesis, as indicated by decreased transcriptional promoters of differentiation, decreased fat droplet formation, and decreased expression of differentiated adipocyte markers, such as adiponectin and perilipin. Preincubating hMSC with Ptx prevented 90% of the suppressive effect of As(III). Selective competitive antagonists of Gi-coupled endothelin-1 type A and B receptors were ~60% effective in blocking As(III) inhibition and combination of antagonists to both receptors were 85% effective. In contrast, antagonists to the sphingosine-1-phosphate type 1 receptor (previously shown to mediate As(III) vascular effects) or the angiotensin II type 1 receptor were ineffective in blocking As(III) effects. These studies suggest a majority of arsenic-inhibited adipocyte differentiation, and metabolism requires endothelin-1 GPCRs and that As(III) effects on GPCR signaling are tissue and context specific. This may represent a significant mechanism for the contribution of arsenic exposure to increased metabolic and cardiovascular diseases.


Subject(s)
Adipogenesis , Arsenic/pharmacology , Cell Differentiation , Endothelin-1/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Stem Cells/cytology , Animals , Blotting, Western , Cells, Cultured , Humans , Mice , Polymerase Chain Reaction , Protein Binding
10.
Am J Pathol ; 174(5): 1949-58, 2009 May.
Article in English | MEDLINE | ID: mdl-19349368

ABSTRACT

Arsenic in drinking water is a major public health concern as it increases risk and incidence of cardiovascular disease and cancer. Arsenic exposure affects multiple vascular beds, promoting liver sinusoidal capillarization and portal hypertension, ischemic heart disease, peripheral vascular disease, and tumor angiogenesis. While Rac1-GTPase and NADPH oxidase activities are essential for arsenic-stimulated endothelial cell signaling for angiogenesis or liver sinusoid capillarization, the mechanism for initiating these effects is unknown. We found that arsenic-stimulated cell signaling and angiogenic gene expression in human microvascular endothelial cells were Pertussis toxin sensitive, indicating a G-protein coupled signaling pathway. Incubating human microvascular endothelial cells with the sphingosine-1-phosphate type 1 receptor (S1P(1)) inhibitor VPC23019 or performing small interfering RNA knockdown of S1P(1) blocked arsenic-stimulated HMVEC angiogenic gene expression and tube formation, but did not affect induction of either HMOX1 or IL8. Liver sinusoidal endothelial cells (LSECs) defenestrate and capillarize in response to aging and environmental oxidant stresses. We found that S1P(1) was enriched on LSECs in vivo and in primary cell culture and that VPC23019 inhibited both sphingosine-1-phosphate-stimulated and arsenic-stimulated LSEC oxidant generation and defenestration. These studies identified novel roles for S1P(1) in mediating arsenic stimulation of both angiogenesis and pathogenic LSEC capillarization, as well as demonstrating a role for S1P(1) in mediating environmental responses in the liver vasculature, providing possible mechanistic insight into arsenic-induced vascular pathogenesis and disease.


Subject(s)
Arsenic/toxicity , Endothelium, Vascular/physiology , Heme Oxygenase-1/metabolism , Interleukin-8/metabolism , Liver/blood supply , Liver/drug effects , Neovascularization, Physiologic/genetics , Receptors, Lysosphingolipid/metabolism , Animals , Cells, Cultured , Heme Oxygenase-1/genetics , Interleukin-8/genetics , Liver/pathology , Lysophospholipids/metabolism , Male , Mice , Mice, Inbred C57BL , Pertussis Toxin/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Superoxides/metabolism , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/metabolism
11.
Toxicol Sci ; 102(2): 319-27, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18182400

ABSTRACT

Arsenic in the drinking water may promote vascular diseases in millions of people worldwide through unresolved mechanisms. In addition, little is known of the effects of coexposures to arsenic and other common vasculature toxicants, such as alcohol. To investigate signaling interactions between arsenic and alcohols, primary human microvascular endothelial (HMVEC) cells were exposed to noncytotoxic concentrations of arsenite (1-5 microM) in the presence or absence of 0.1% ethanol (EtOH). Coexposure, but not exposure to either agent alone, rapidly increased active Fyn tyrosine kinase, tyrosine phosphorylation of a 109-kDa protein and serine phosphorylation of protein kinase C (PKC)delta. The 109-kDa protein was identified as PYK2, a regulator of vascular integrin signaling and an upstream activator of PKCdelta. Membrane localization of phospholipase Cgamma1 was increased by coexposure within 15 min, but not by either agent alone. In contrast, both agents equally increased membrane localization of Rac1-GTPase. Coexposure, but not exposure to either agent alone, induced transcript levels for the angiogenic genes, vascular endothelial cell growth factor (Vegfa) and insulin-like growth factor-1 (Igf1). However, EtOH inhibited arsenic-induced, nuclear factor-kappaB-driven interleukin-8 and collagen-1 expression. Differential effects of selective PKC inhibitors on induced gene expression combined with a lack of interaction for induction of hemeoxygenase-1 further demonstrated that arsenic-responsive signaling pathways differ in sensitivity to EtOH interactions. Finally, coexposure enhanced endothelial tube formation in in vitro angiogenesis assays. These data indicate that complex interactions occur between arsenic and EtOH exposures that functionally affect endothelial signaling for gene induction and remodeling stimuli.


Subject(s)
Arsenites/adverse effects , Cell Survival/drug effects , Endothelium, Vascular/drug effects , Ethanol/toxicity , Neovascularization, Pathologic/chemically induced , Water Pollutants, Chemical/toxicity , Blotting, Western , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Gene Expression Regulation/drug effects , Humans , Membrane Proteins/metabolism , Microcirculation/drug effects , Microcirculation/metabolism , Microcirculation/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , RNA, Messenger/metabolism , Signal Transduction/drug effects , Tetrazolium Salts , Transcriptional Activation
12.
Biochem J ; 402(2): 261-9, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17078813

ABSTRACT

Chronic inhalation of low amounts of Cr(VI) promotes pulmonary diseases and cancers through poorly defined mechanisms. SFKs (Src family kinases) in pulmonary airway cells may mediate Cr(VI) signalling for lung injury, although the downstream effectors of Cr(VI)-stimulated SFKs and how they relate to pathogenic gene induction are unknown. Therefore SFK-dependent activation of transcription factors by non-cytotoxic exposure of human bronchial epithelial cells to Cr(VI) was determined. Protein-DNA binding arrays demonstrated that exposing BEAS 2B cells to 5 microM Cr(VI) for 4 and 24 h resulted in increased protein binding to 25 and 43 cis-elements respectively, while binding to 12 and 16 cis-elements decreased. Of note, Cr(VI) increased protein binding to several STAT (signal transducer and activator of transcription) cis-elements. Cr(VI) stimulated acute tyrosine phosphorylation and nuclear translocation of STAT1 over a 4 h period and a prolonged activation of STAT3 that reached a peak between 48 and 72 h. This prolonged activation was observed for both STAT3alpha and STAT3beta. Immunofluorescent confocal microscopy confirmed that Cr(VI) increased nuclear localization of phosphorylated STAT3 for more than 72 h in both primary and BEAS 2B human airway cells. Cr(VI) induced transactivation of both a STAT3-driven luciferase reporter construct and the endogenous inflammatory gene IL-6 (interleukin-6). Inhibition with siRNA (small interfering RNA) targeting the SFK Lck, but not dominant-negative JAK (Janus kinase), prevented Cr(VI)-stimulated phosphorylation of both STAT3 isoforms and induction of IL-6. The results suggest that Cr(VI) activates epithelial cell Lck to signal for prolonged STAT3 activation and transactivation of IL-6, an important immunomodulator of lung disease progression.


Subject(s)
Bronchi/metabolism , Cell Nucleus/metabolism , Chromium/pharmacology , Epithelial Cells/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Phosphotyrosine/metabolism , STAT3 Transcription Factor/metabolism , Active Transport, Cell Nucleus , Bronchi/cytology , Bronchi/drug effects , Cell Line , Cell Survival/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Humans , Interleukin-6/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Protein Binding , RNA, Messenger/genetics , Time Factors , Transcriptional Activation
13.
Toxicol Appl Pharmacol ; 222(3): 327-36, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17123562

ABSTRACT

The vascular effects of arsenic in drinking water are global health concerns contributing to human disease worldwide. Arsenic targets the endothelial cells lining blood vessels, and endothelial cell activation or dysfunction may underlie the pathogenesis of both arsenic-induced vascular diseases and arsenic-enhanced tumorigenesis. The purpose of the current studies was to demonstrate that exposing mice to drinking water containing environmentally relevant levels of arsenic promoted endothelial cell dysfunction and pathologic vascular remodeling. Increased angiogenesis, neovascularization, and inflammatory cell infiltration were observed in Matrigel plugs implanted in C57BL/6 mice following 5-week exposures to 5-500 ppb arsenic [Soucy, N.V., Mayka, D., Klei, L.R., Nemec, A.A., Bauer, J.A., Barchowsky, A., 2005. Neovascularization and angiogenic gene expression following chronic arsenic exposure in mice. Cardiovasc.Toxicol 5, 29-42]. Therefore, functional in vivo effects of arsenic on endothelial cell function and vessel remodeling in an endogenous vascular bed were investigated in the liver. Liver sinusoidal endothelial cells (LSEC) became progressively defenestrated and underwent capillarization to decrease vessel porosity following exposure to 250 ppb arsenic for 2 weeks. Sinusoidal expression of PECAM-1 and laminin-1 proteins, a hallmark of capillarization, was also increased by 2 weeks of exposure. LSEC caveolin-1 protein and caveolae expression were induced after 2 weeks of exposure indicating a compensatory change. Likewise, CD45/CD68-positive inflammatory cells did not accumulate in the livers until after LSEC porosity was decreased, indicating that inflammation is a consequence and not a cause of the arsenic-induced LSEC phenotype. The data demonstrate that the liver vasculature is an early target of pathogenic arsenic effects and that the mouse liver vasculature is a sensitive model for investigating vascular health effects of arsenic.


Subject(s)
Arsenic/toxicity , Inflammation/chemically induced , Liver Circulation/drug effects , Neovascularization, Pathologic/chemically induced , Poisons/toxicity , Animals , Collagen , Dose-Response Relationship, Drug , Drug Combinations , Fluorescent Antibody Technique , Inflammation/pathology , Laminin/metabolism , Leukocytes/pathology , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Microvilli/pathology , Neovascularization, Pathologic/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Proteoglycans
14.
Hepatology ; 45(1): 205-12, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17187425

ABSTRACT

UNLABELLED: Trivalent arsenic [As(III)] is a well-known environmental toxicant that causes a wide range of organ-specific diseases and cancers. In the human liver, As(III) promotes vascular remodeling, portal fibrosis, and hypertension, but the pathogenesis of these As(III)-induced vascular changes is unknown. To investigate the hypothesis that As(III) targets the hepatic endothelium to initiate pathogenic change, mice were exposed to 0 or 250 parts per billion (ppb) of As(III) in their drinking water for 5 weeks. Arsenic(III) exposure did not affect the overall health of the animals, the general structure of the liver, or hepatocyte morphology. There was no change in the total tissue arsenic levels, indicating that arsenic does not accumulate in the liver at this level of exposure. However, there was significant vascular remodeling with increased sinusoidal endothelial cell (SEC) capillarization, vascularization of the peribiliary vascular plexus (PBVP), and constriction of hepatic arterioles in As(III)-exposed mice. In addition to ultrastructural demonstration of SEC defenestration and capillarization, quantitative immunofluorescence analysis revealed increased sinusoidal PECAM-1 and laminin-1 protein expression, suggesting gain of adherens junctions and a basement membrane. Conversion of SECs to a capillarized, dedifferentiated endothelium was confirmed at the cellular level with demonstration of increased caveolin-1 expression and SEC caveolae, as well as increased membrane-bound Rac1-GTPase. CONCLUSION: These data demonstrate that exposure to As(III) causes functional changes in SEC signaling for sinusoidal capillarization that may be initial events in pathogenic changes in the liver.


Subject(s)
Arsenic/toxicity , Blood Vessels/drug effects , Capillaries/drug effects , Endothelial Cells/drug effects , Liver/blood supply , Liver/drug effects , Animals , Blood Vessels/pathology , Capillaries/pathology , Caveolin 1/genetics , Caveolin 1/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Endothelium/blood supply , Gene Expression Regulation/drug effects , Laminin/genetics , Laminin/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic , Neuropeptides/genetics , Neuropeptides/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Vasoconstriction/drug effects , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
15.
J Cell Physiol ; 209(1): 113-21, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16775837

ABSTRACT

Inhaled hexavalent chromium (Cr(VI)) promotes lung injury and pulmonary diseases through poorly defined mechanisms. One hypothesis for this lung pathogenesis is that Cr(VI) silences induction of cytoprotective genes, such as heme oxygenase-1 (HO-1), whose total lung mRNA levels were reduced 21 days after nasal instillation of potassium dichromate in C57BL/6 mice. To investigate the mechanisms for this inhibition, Cr(VI) effects on basal and arsenic (As(III))-induced HO-1 expression were examined in cultured human bronchial epithelial (BEAS-2B) cells. An effect of Cr(VI) on the low basal HO-1 mRNA and protein levels in BEAS-2B cells was not detectible. In contrast, Cr(VI) added to the cells before As(III), but not simultaneously with As(III), attenuated As(III)-induced HO-1 expression. Transient transfection with luciferase reporter gene constructs controlled by the full length ho-1 promoter or deletion mutants demonstrated that this inhibition occurred in the E1 enhancer region containing critical antioxidant response elements (ARE). Cr(VI) pretreatment inhibited As(III)-induced activity of a transiently expressed reporter construct regulated by three ARE tandem repeats. The mechanism for this Cr(VI)-attenuated transactivation appeared to be Cr(VI) reduction of the nuclear levels of the transcription factor Nrf2 and As(III)-stimulated Nrf2 transcriptional complex binding to the ARE cis element. Finally, exposing cells to Cr(VI) prior to co-exposure with As(III) synergized for apoptosis and loss of membrane integrity. These data suggest that Cr(VI) silences induction of ARE-driven genes required for protection from secondary insults. The data also have important implications for understanding the toxic mechanisms of low level, mixed metal exposures in the lung.


Subject(s)
Arsenic/toxicity , Chromium/pharmacology , Heme Oxygenase-1/metabolism , RNA, Messenger/metabolism , Response Elements , Trachea/cytology , Animals , Apoptosis , Cell Line , Drug Synergism , Enhancer Elements, Genetic , Epithelial Cells , Heme Oxygenase-1/genetics , Humans , Mice , Mice, Inbred C57BL , Transcriptional Activation , Transfection
16.
Cardiovasc Toxicol ; 5(1): 29-41, 2005.
Article in English | MEDLINE | ID: mdl-15738583

ABSTRACT

Exposure to arsenic in drinking water increases incidence of cardiovascular diseases. However, the basic mechanisms and genetic changes that promote these diseases are unknown. This study investigated the effects of chronic arsenic exposure on vessel growth and expression of angiogenic and tissue remodeling genes in cardiac tissues. Male mice were exposed to low to moderately high levels of arsenite (AsIII) for 5, 10, or 20 wk in their drinking water. Vessel growth in Matrigel implants was tested during the last 2 wk of each exposure period. Implant vascularization increased in mice exposed to 5-500 ppb AsIII for 5 wk. Similar increases were seen following exposure to 50-250 ppb of AsIII over 20 wk, but the response to 500 ppb decreased with time. RT-PCR analysis of cardiac mRNA revealed differential expression of angiogenic or tissue remodeling genes, such as vascular endothelial cell growth factor (VEGF), VEGF receptors, plasminogen activator inhibitor-1, endothelin-1, and matrix metalloproteinase-9, which varied with time or amount of exposure. VEGF receptor mRNA and cardiac microvessel density were reduced by exposure to 500 ppb AsIII for 20 wk. These data demonstrate differential concentration and time-dependent effects of chronic arsenic exposure on cardiovascular phenotype and vascular remodeling that may explain the etiology for AsIII-induced disease.


Subject(s)
Angiogenic Proteins/biosynthesis , Arsenic/toxicity , Neovascularization, Pathologic/chemically induced , Angiogenic Proteins/genetics , Animals , Arsenic/administration & dosage , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology
17.
Chem Res Toxicol ; 17(4): 555-63, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15089098

ABSTRACT

Chronic arsenic exposure is associated with an increased risk for cancer, cardiovascular disease (including ischemic heart disease and hypertension), peripheral vascular disease, and diabetes. Arsenic causes blood vessel growth and remodeling in vivo and cell specific, dose-dependent induction vascular endothelial growth factor-A (VEGF), which is essential for both processes. The current study examined the hypothesis that low, environmentally relevant levels of trivalent arsenic (AsIII) activate discrete signaling pathways in vascular smooth muscle cells (SMC) to induce expression of VEGF. AsIII caused a progressive increase in VEGF mRNA levels over a 48 h period in primary porcine SMC with a threshold of 1-2.5 microM. VEGF protein levels increased with a similar concentration dependence and time course. Hypoxia inducible factor-1alpha (HIF-1alpha) protein and mRNA levels also increased in response to AsIII. However, unlike the response to an iron chelator, AsIII-induced VEGF was not inhibited by siRNA directed toward HIF-1alpha. Instead, a novel protein kinase C, PKCdelta, was activated by AsIII to induce VEGF and stabilize HIF-1alpha. Consistent with this activation, AsIII caused coordinate increases in the levels of the intracellular second messenger diacyglycerol (DAG). These data suggest that AsIII induced divergent signaling pathways in SMCs that lead to independent increases in VEGF expression and HIF-1alpha signaling. However, these pathways both require initial increases in DAG levels and PKC activity.


Subject(s)
Arsenic/toxicity , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Arsenic/chemistry , Chelating Agents , Dose-Response Relationship, Drug , Hypoxia-Inducible Factor 1, alpha Subunit , Muscle, Smooth, Vascular/cytology , RNA, Messenger/analysis , Risk Factors , Second Messenger Systems , Signal Transduction , Swine , Transcription Factors/biosynthesis
18.
Toxicol Sci ; 76(2): 271-9, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12970581

ABSTRACT

Trivalent inorganic arsenic (arsenite, arsenic trioxide, As[III]) is currently being used to treat hematologic tumors and is being investigated for treating solid tumors. However, low concentrations of As(III) stimulate vascular cell proliferation in cell culture, although this has not been confirmed in vivo. Therefore, the hypothesis that As(III) enhances blood vessel growth (angiogenesis) and tumorigenesis was tested in two in vivo models of angiogenesis and a model of tumor growth. In the first, arsenite caused a dose-dependent increase in vessel density in a chicken chorioallantoic-membrane (CAM) assay. The threshold As(III) concentration for this response was 0.033 microM and inhibition of vessel growth was observed at concentrations greater than 1 microM. Mouse Matrigel implants were used to test the angiogenic effects of As(III) in an adult mammalian system. Mice were injected with 0.8-80 microg/kg As(III)/day over a three-week period. During the last two weeks, Matrigel plugs were placed on the abdominal wall. Low and high doses of As(III) were synergistic with fibroblast growth factor-2 (FGF-2) in increasing vessel density in the Matrigel assay, while a middle dose had no effect. To test the effects of As(III) on tumor growth, GFP-labeled B16-F10 mouse melanoma cells were implanted in nude mice, which subsequently received biweekly injections of 0.5-5.0 mg/kg As(III). Significant tumor growth and lung metastasis was seen in all animals, with the largest tumors occurring in animals treated with lower doses of As(III). These studies support the hypothesis and indicate that induction of angiogenesis, enhanced tumor growth, and metastasis are potential dose-dependent toxic side effects of arsenic therapies.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Arsenites/pharmacology , Blood Vessels/drug effects , Melanoma, Experimental/blood supply , Neovascularization, Pathologic , Skin Neoplasms/blood supply , Allantois/blood supply , Allantois/drug effects , Allantois/pathology , Animals , Blood Vessels/growth & development , Blood Vessels/pathology , Chick Embryo , Chorion/blood supply , Chorion/drug effects , Chorion/pathology , Dose-Response Relationship, Drug , Drug Synergism , Fibroblast Growth Factor 2/pharmacology , Lung Neoplasms/blood supply , Lung Neoplasms/secondary , Male , Melanoma, Experimental/secondary , Mice , Mice, Nude , Skin Neoplasms/pathology
19.
Toxicol Appl Pharmacol ; 190(3): 214-23, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12902192

ABSTRACT

Inhaled hexavalent chromium (Cr(VI)) promotes pulmonary disease and lung cancer through poorly defined mechanisms. These mechanisms were studied in A549 lung epithelial cells to investigate the hypothesis that nontoxic Cr(VI) exposures selectively activate cell signaling that shifts the balance of gene transcription. These studies demonstrated that nontoxic doses of Cr(VI) (10 microM) increased reactive oxygen species and selectively activated c-Jun N-terminal kinase (JNK), relative to ERK or p38 MAP kinase. In contrast, only toxic, nonselective levels of exogenous oxidants stimulated JNK. However, JNK activation in response to Cr(VI) and exogenous H(2)O(2) (1 mM) shared requirements for intracellular thiol oxidation, activation of Src family kinases, and p130(cas) (Cas). Cr(VI) did not mimic H(2)O(2)-mediated stimulation of JNK in fibroblasts containing only Src and did not activate Src or Yes in A549 cells. Instead, Fyn and Lck were activated in A549 cells, indicating activation of specific Src family kinases in response to Cr(VI). Finally, Cr(VI) was demonstrated to directly activate purified Fyn in vitro and the majority of this activation did not require oxidant generation. These data suggest that nontoxic levels of Cr(VI), which can shift patterns of gene transcription, are selective in their activation of cell signaling and that Cr(VI) can directly activate Src family kinases independently of reactive oxygen species generation.


Subject(s)
Chromium/toxicity , Epithelial Cells/drug effects , Mitogen-Activated Protein Kinases/biosynthesis , Neoplasm Proteins , Proteins , src-Family Kinases/biosynthesis , Cells, Cultured , Crk-Associated Substrate Protein , Dose-Response Relationship, Drug , Epithelial Cells/enzymology , Fibroblasts/drug effects , Fibroblasts/enzymology , Humans , Hydrogen Peroxide/pharmacology , JNK Mitogen-Activated Protein Kinases , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/biosynthesis , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Protein-Tyrosine Kinases/biosynthesis , Reactive Oxygen Species/metabolism , Retinoblastoma-Like Protein p130 , Signal Transduction , Transfection , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...