Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Exp Med Biol ; 961: 125-35, 2013.
Article in English | MEDLINE | ID: mdl-23224875

ABSTRACT

Changes in cardiac gene expression contribute to the progression of heart failure by affecting cardiomyocyte growth, function, and survival. The Na(+)-Ca(2+) exchanger gene (Ncx1) is upregulated in hypertrophy and is often found elevated in end-stage heart failure. Studies have shown that the change in its expression contributes to contractile dysfunction. Several transcriptional pathways mediate Ncx1 expression in pathological cardiac remodeling. Both α-adrenergic receptor (α-AR) and ß-adrenergic receptor (ß-AR) signaling can play a role in the regulation of calcium homeostasis in the cardiomyocyte, but chronic activation in periods of cardiac stress contributes to heart failure by mechanisms which include Ncx1 upregulation. Our studies have even demonstrated that NCX1 can directly act as a regulator of "activity-dependent signal transduction" mediating changes in its own expression. Finally, we present evidence that histone deacetylases (HDACs) and histone acetyltransferases (HATs) act as master regulators of Ncx1 expression. We show that many of the transcription factors regulating Ncx1 expression are important in cardiac development and also in the regulation of many other genes in the so-called fetal gene program, which are activated by pathological stimuli. Importantly, studies have revealed that the transcriptional network regulating Ncx1 expression is also mediating many of the other changes in genetic remodeling contributing to the development of cardiac dysfunction and revealed potential therapeutic targets for the treatment of hypertrophy and failure.


Subject(s)
Cardiomegaly/metabolism , Gene Expression Regulation , Heart Failure/metabolism , Muscle Proteins/biosynthesis , Sodium-Calcium Exchanger/biosynthesis , Transcription, Genetic , Animals , Calcium/metabolism , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/therapy , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/therapy , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Muscle Proteins/genetics , Myocardial Contraction/genetics , Receptors, Adrenergic, alpha/genetics , Receptors, Adrenergic, alpha/metabolism , Receptors, Adrenergic, beta/genetics , Receptors, Adrenergic, beta/metabolism , Sodium-Calcium Exchanger/genetics
2.
Am J Physiol Gastrointest Liver Physiol ; 297(2): G259-68, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19520742

ABSTRACT

Endothelial activation and surface expression of cell adhesion molecules (CAMs) is critical for binding and recruitment of circulating leukocytes in tissues during the inflammatory response. Endothelial CAM expression plays a critical role in the intestinal microvasculature in inflammatory bowel disease (IBD), as blockade of leukocyte alpha4-integrin binding by gut endothelial CAM ligands has therapeutic benefit in IBD. Mechanisms underlying expression of vascular cell adhesion molecule (VCAM)-1, a ligand for alpha4-integrin in primary cultures of human intestinal microvascular endothelial cells (HIMEC) has not been defined. We investigated the effect of curcumin, phosphatidylinositol 3-kinase (PI 3-kinase)/protein kinase B (Akt), and mitogen-activated protein kinase (MAPK) inhibitors on VCAM-1 expression and function in HIMEC. CAM expression was assessed and HIMEC-leukocyte adhesion was visualized under static and flow conditions. Western blotting and in vitro kinase assays were used to assess Akt and MAPK activation. Nuclear factor-kappaB (NF-kappaB) activation and nuclear translocation of its p65 subunit were determined. Tumor necrosis factor (TNF)-alpha/lipopolysaccharide (LPS)-induced VCAM-1 expression in HIMEC was suppressed by Akt small-interfering RNA, curcumin, and inhibitors of NF-kappaB (SN-50), p38 MAPK (SB-203580) and PI 3-kinase/Akt (LY-294002). VCAM-1 induction was partially suppressed by p44/42 MAPK (PD-098059) but unaffected by c-Jun NH2-terminal kinase (SP-600125) inhibition. Curcumin inhibited Akt/MAPK/NF-kappaB activity and prevented nuclear translocation of the p65 NF-kappaB subunit following TNF-alpha/LPS. At physiological shear stress, curcumin attenuated leukocyte adhesion to TNF-alpha/LPS-activated HIMEC monolayers. In conclusion, curcumin inhibited the expression of VCAM-1 in HIMECs through blockade of Akt, p38 MAPK, and NF-kappaB. Curcumin may represent a novel therapeutic agent targeting endothelial activation in IBD.


Subject(s)
Curcumin/pharmacology , Endothelial Cells/drug effects , Gastrointestinal Agents/pharmacology , Intestines/blood supply , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor RelA/antagonists & inhibitors , Vascular Cell Adhesion Molecule-1/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Active Transport, Cell Nucleus , Cell Adhesion/drug effects , Cell Adhesion Molecules , Cells, Cultured , Endothelial Cells/enzymology , Humans , Immunoglobulins/metabolism , Leukocytes/drug effects , Leukocytes/metabolism , Lipopolysaccharides/pharmacology , Microvessels/drug effects , Microvessels/enzymology , Mucoproteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , Signal Transduction/drug effects , Time Factors , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Am J Surg ; 191(2): 276-80, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16442960

ABSTRACT

BACKGROUND: The Trauma Evaluation and Management (TEAM) module orients medical students to the initial assessment of an injured patient. At the Medical College of Wisconsin, a course based on expanded TEAM (eTEAM) was developed for junior medical students. This study determined whether eTEAM improved the ability to perform and retain primary survey skills. METHODS: Objective Structured Clinical Examination methodology was used to compare 2 groups of senior medical students 1 year after receiving either a 2-hour lecture or eTEAM. RESULTS: Students receiving eTEAM performed the primary survey much better than those receiving lecture alone. The overall Objective Structured Clinical Examination scores did not differ between groups. CONCLUSIONS: Medical students participating in eTEAM retained the ability to perform a primary survey in proper sequence 1 year later better than students receiving the information in lecture format only.


Subject(s)
Data Collection , Retention, Psychology , Students, Medical/psychology , Traumatology/education , Curriculum , Wisconsin
4.
Cell Commun Signal ; 2(1): 3, 2004 Jun 02.
Article in English | MEDLINE | ID: mdl-15175101

ABSTRACT

The immunosuppressive agent cyclosporin A (CsA), a calcineurin inhibitor which blocks T cell activation has provided the pharmacologic foundation for organ transplantation. CsA exerts additional effects on non-immune cell populations and may adversely effect microvascular endothelial cells, contributing to chronic rejection, a long-term clinical complication and significant cause of mortality in solid-organ transplants, including patients with small bowel allografts. Growth of new blood vessels, or angiogenesis, is a critical homeostatic mechanism in organs and tissues, and regulates vascular populations in response to physiologic requirements. We hypothesized that CsA would inhibit the angiogenic capacity of human gut microvessels. Primary cultures of human intestinal microvascular endothelial cells (HIMEC) were used to evaluate CsA's effect on four in vitro measures of angiogenesis, including endothelial stress fiber assembly, migration, proliferation and tube formation, in response to the endothelial growth factor VEGF. We characterized the effect of CsA on intracellular signaling mechanisms following VEGF stimulation. CsA affected all VEGF induced angiogenic events assessed in HIMEC. CsA differentially inhibited signaling pathways which mediated distinct steps of the angiogenic process. CsA blocked VEGF induced nuclear translocation of the transcription factor NFAT, activation of p44/42 MAPK, and partially inhibited JNK and p38 MAPK. CsA differentially affected signaling cascades in a dose dependent fashion and completely blocked expression of COX-2, which was integrally linked to HIMEC angiogenesis. These data suggest that CsA inhibits the ability of microvascular endothelial cells to undergo angiogenesis, impairing vascular homeostatic mechanisms and contributing to the vasculopathy associated with chronic rejection.

5.
Am J Physiol Gastrointest Liver Physiol ; 285(6): G1277-92, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12919942

ABSTRACT

Gastroesophageal reflux disease is the most common malady of the esophagus, affecting 7% of the United States population. Histological assessment demonstrates classic inflammatory mechanisms including selective leukocyte recruitment and hemorrhage, suggesting a prominent role for the microvasculature. We isolated and characterized human esophageal microvascular endothelial cells (EC) (HEMEC), examined inflammatory activation in response to cytokines, LPS, and acidic pH exposure, and identified signaling pathways that underlie activation. HEMEC displayed characteristic morphological and phenotypic features including acetylated LDL uptake. TNF-alpha/LPS activation of HEMEC resulted in upregulation of the cell adhesion molecules (CAM) ICAM-1, VCAM-1, E-selectin, and mucosal addressin CAM-1 (MAdCAM-1), increased IL-8 production, and enhanced leukocyte binding. Both acid and TNF-alpha/LPS activation lead to activation of SAPK/JNK in HEMEC that was linked to VCAM-1 expression and U-937 leukocyte adhesion. Expression of constitutive inducible nitric oxide synthase in HEMEC was in marked contrast to intestinal microvascular endothelial cells. In this study, we demonstrate that HEMECs are phenotypically and functionally distinct from lower gut-derived endothelial cells and will facilitate understanding of inflammatory mechanisms in esophageal inflammation.


Subject(s)
Endothelium, Vascular/physiology , Esophagus/blood supply , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Separation , Cells, Cultured , Chemokines/metabolism , Curcumin/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Esophagitis/etiology , Humans , Immunoglobulins/metabolism , Intestines/blood supply , JNK Mitogen-Activated Protein Kinases , Leukocytes/physiology , Microcirculation , Mitogen-Activated Protein Kinases/metabolism , Mucoproteins/metabolism , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Phenotype , Signal Transduction , Vascular Cell Adhesion Molecule-1/drug effects
6.
J Biol Chem ; 277(38): 35605-15, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12110686

ABSTRACT

The calcineurin inhibitor cyclosporine A (CsA) modulates leukocyte cytokine production but may also effect nonimmune cells, including microvascular endothelial cells, which regulate the inflammatory process through leukocyte recruitment. We hypothesized that CsA would promote a proinflammatory phenotype in human intestinal microvascular endothelial cells (HIMEC), by inhibiting inducible nitric-oxide synthase (iNOS, NOS2)-derived NO, normally an important mechanism in limiting endothelial activation and leukocyte adhesion. Primary cultures of HIMEC were used to assess CsA effects on endothelial activation, leukocyte interaction, and the expression of iNOS as well as cell adhesion molecules. CsA significantly increased leukocyte binding to activated HIMEC, but paradoxically decreased endothelial expression of cell adhesion molecules (E-selectin, intercellular adhesion molecule 1, and vascular cell adhesion molecule-1). In contrast, CsA completely inhibited the expression of iNOS in tumor necrosis factor-alpha/lipopolysaccharide-activated HIMEC. CsA blocked p38 MAPK phosphorylation in activated HIMEC, a key pathway in iNOS expression, but failed to inhibit NFkappaB activation. These studies demonstrate that CsA exerts a proinflammatory effect on HIMEC by blocking iNOS expression. CsA exerts a proinflammatory effect on the microvascular endothelium, and this drug-induced endothelial dysfunction may help explain its lack of efficacy in the long-term treatment of chronically active inflammatory bowel disease.


Subject(s)
Cell Adhesion/drug effects , Cyclosporine/pharmacology , Endothelium, Vascular/drug effects , Immunosuppressive Agents/pharmacology , Intestines/blood supply , Leukocytes/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Base Sequence , Cells, Cultured , DNA Primers , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Humans , Leukocytes/cytology , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/physiology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...