Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Commun ; 7: 13516, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27869117

ABSTRACT

In humans and animals lacking functional LDL receptor (LDLR), LDL from plasma still readily traverses the endothelium. To identify the pathways of LDL uptake, a genome-wide RNAi screen was performed in endothelial cells and cross-referenced with GWAS-data sets. Here we show that the activin-like kinase 1 (ALK1) mediates LDL uptake into endothelial cells. ALK1 binds LDL with lower affinity than LDLR and saturates only at hypercholesterolemic concentrations. ALK1 mediates uptake of LDL into endothelial cells via an unusual endocytic pathway that diverts the ligand from lysosomal degradation and promotes LDL transcytosis. The endothelium-specific genetic ablation of Alk1 in Ldlr-KO animals leads to less LDL uptake into the aortic endothelium, showing its physiological role in endothelial lipoprotein metabolism. In summary, identification of pathways mediating LDLR-independent uptake of LDL may provide unique opportunities to block the initiation of LDL accumulation in the vessel wall or augment hepatic LDLR-dependent clearance of LDL.


Subject(s)
Activin Receptors, Type II/metabolism , Cholesterol, LDL/metabolism , Endothelial Cells/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Activin Receptors, Type II/genetics , Animals , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Biological Transport , Cells, Cultured , Cholesterol, LDL/genetics , Cloning, Molecular , Gene Knockdown Techniques , Genome-Wide Association Study , Humans , Male , Mice , RNA Interference
2.
Inflamm Bowel Dis ; 17(7): 1501-12, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21674706

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) consists of Crohn's disease (CD) and ulcerative colitis (UC), two widespread diseases of unknown, multifactorial etiology. Colitis pathology involves a pathological angiogenic response where increases in vascular density participate in colitis histopathology. Vascular endothelial growth factor-A (VEGF-A) is a potent angiogenesis stimulator known to be involved in pathological angiogenesis in several diseases including colitis. However, the pathogenic importance of specific VEGF-A isoforms during T-cell-mediated experimental colitis remains largely unknown. METHODS: The CD4⁺ CD45RB(high) T-cell transfer model of experimental colitis was used for these studies. The VEGF lac-Z transgenic reporter mouse was used to examine specific cellular sources of VEGF-A production. The VEGF164 aptamer (Macugen), adenoviral VEGF164, and the VEGF Trap were used to evaluate pathological importance. RESULTS: VEGF lac-Z reporter mice experiments showed that both infiltrating T cells and local tissue cells produce VEGF-A in the colon during disease. Inhibition of VEGF164 using a highly selective RNA aptamer significantly attenuated CD4⁺ CD45RB(high) T-cell-dependent experimental colitis by reducing pathological angiogenesis and inflammatory pathology. Conversely, broad-spectrum VEGF inhibition with VEGF Trap did not attenuate disease, nor did adenoviral VEGF164 overexpression significantly alter colitis pathology. CONCLUSIONS: VEGF164 is actively produced by multiple cell types during T-cell-mediated colitis. Importantly, specific inhibition of the VEGF164 isoform during T-cell-mediated colitis dose-dependently attenuated disease progression, while broad-scale inhibition of all VEGF-A isoforms was not therapeutically beneficial.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Colitis/etiology , Colitis/metabolism , Disease Models, Animal , T-Lymphocytes, Regulatory/immunology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/physiology , Animals , Aptamers, Nucleotide/pharmacology , Colitis/pathology , Colon/immunology , Colon/metabolism , Colon/pathology , Enzyme-Linked Immunosorbent Assay , Homeodomain Proteins , Humans , Inflammation/prevention & control , Leukocyte Common Antigens/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic/prevention & control , Protein Isoforms , Vascular Endothelial Growth Factor A/genetics
3.
Am J Physiol Gastrointest Liver Physiol ; 299(6): G1361-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20884890

ABSTRACT

Leukocyte recruitment to inflamed tissues is the cornerstone of inflammatory responses and the driving force behind the establishment of inflammatory bowel disease, consisting of Crohn's disease and ulcerative colitis. It has been reported that angiogenic cytokines contribute to this inflammatory response that facilitates the chronic nature of disease. We have previously reported (Goebel S, Huang M, Davis WC, Jennings M, Siahaan TJ, Alexander JS, Kevil CG. Am J Physiol Gastrointest Liver Physiol 290: G648-G654, 2006) that vascular endothelial growth factor (VEGF)-A can stimulate neutrophil adhesion to colon microvascular endothelial cells in a ß2-integrin (Itgb2)-dependent manner. However, it is not known which of the specific leukocyte integrins are critical for VEGF-A-dependent neutrophil and T cell recruitment. Here we examine the differential importance of either α-integrin (Itga)L or ItgaM in governing neutrophil and T cell adhesion to VEGF-A-activated colonic endothelium. Using an in vitro parallel-plate flow chamber model, we found that genetic deficiency of ItgaM completely blunted neutrophil adhesion to VEGF-A-stimulated endothelium, whereas ItgaL deficiency only partly blocked neutrophil adhesion. Deficiency of ItgaM did significantly decrease neutrophil rolling, whereas deficiency of ItgaL did not. We found that genetic deficiency of either ItgaL or ItgaM did significantly blunt T cell adhesion to VEGF-A-stimulated colon endothelium. We also found that genetic deficiency of these Itgas significantly attenuated T cell rolling behavior. Lastly, we examined whether VEGF-A-mediated leukocyte recruitment occurred through different VEGF receptor (VEGFR) pathways and found that VEGFR2 activation regulates neutrophil recruitment, whereas both VEGFR1 and VEGFR2 modulate T cell recruitment. Together, these data identify differential molecular mechanisms of VEGF-A-mediated leukocyte recruitment.


Subject(s)
CD11a Antigen/metabolism , CD11b Antigen/metabolism , Neutrophils/physiology , T-Lymphocytes/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Bone Marrow Cells/metabolism , CD11a Antigen/genetics , CD11b Antigen/genetics , Cell Adhesion , Cell Line , Colon/physiology , Endothelium/physiology , Gene Expression Regulation/physiology , Ligands , Mice , Mice, Knockout , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/genetics
4.
Cardiovasc Res ; 86(2): 219-25, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20202978

ABSTRACT

Caveolae are specialized lipid rafts that form flask-shaped invaginations of the plasma membrane. They are involved in cell signalling and transport and have been shown critically regulate vascular reactivity and blood pressure. The organization and functions of caveolae are mediated by coat proteins (caveolins) and support or adapter proteins (cavins). The caveolins, caveolin-1, -2, and -3, form the structural backbone of caveolae. These proteins are also highly integrated into caveolae function and have their own activity independent of caveolae. The cavins, cavins 1-4, are involved in regulation of caveolae and modulate the function of caveolins by promoting the membrane remodelling and trafficking of caveolin-derived structures. The relationships between these different proteins are complex and intersect with many aspects of cell function. Caveolae have also been implicated in chronic inflammatory conditions and other pathologies including atherosclerosis, inflammatory bowel disease, muscular dystrophy, and generalized dyslipidaemia. The pathogenic role of the caveolins is an emerging area, however, the roles of cavins in disease is just beginning to be explored. This review will examine the relationship between caveolins and cavins and explore the role of caveolae in inflammatory signalling mechanisms.


Subject(s)
Caveolae/immunology , Caveolins/immunology , Endothelium, Vascular/immunology , Inflammation Mediators/immunology , Inflammation/immunology , Intracellular Signaling Peptides and Proteins/immunology , Signal Transduction , Animals , Endothelium, Vascular/physiopathology , Humans , Inflammation/physiopathology
5.
Gastroenterology ; 136(2): 575-84.e2, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19111727

ABSTRACT

BACKGROUND & AIMS: Increased vascular density has been associated with progression of human inflammatory bowel diseases (IBDs) and animal models of colitis. Pathologic angiogenesis in chronically inflamed tissues is mediated by several factors that are regulated at specialized lipid rafts known as caveolae. Caveolin-1 (Cav-1), the major structural protein of caveolae in endothelial cells, is involved in the regulation of angiogenesis, so we investigated its role in experimental colitis. METHODS: Colitis was induced by administration of dextran sodium sulfate to wild-type and Cav-1(-/-) mice, as well as Cav-1(-/-) mice that overexpress Cav-1 only in the endothelium. Colon tissues were analyzed by histologic analyses. Leukocyte recruitment was analyzed by intravital microscopy; angiogenesis was evaluated by immunohistochemistry and in vivo disk assays. RESULTS: Cav-1 protein levels increased after the induction of colitis in wild-type mice. In Cav-1(-/-) mice or mice given a Cav-1 inhibitory peptide, the colitis histopathology scores, vascular densities, and levels of inflammatory infiltrates decreased significantly compared with controls. Lower levels of leukocyte and platelet rolling and adhesion colitis also were observed in Cav-1(-/-) mice and mice given a Cav-1 inhibitory peptide, compared with controls. Cav-1(-/-) mice that received transplants of wild-type bone marrow had a lower colitis score than wild-type mice. Data from mice that overexpress Cav-1 only in the endothelium indicated that endothelial Cav-1 is the critical regulator of colitis. Genetic deletion or pharmacologic inhibition of endothelial Cav-1 also significantly decreased vascular densities and angiogenesis scores, compared with controls. CONCLUSIONS: Endothelial Cav-1 mediates angiogenesis in experimental colitis. Modulation of Cav-1 could provide a novel therapeutic target for IBD.


Subject(s)
Caveolin 1/metabolism , Colitis/metabolism , Neovascularization, Pathologic/metabolism , Animals , Caveolae/pathology , Caveolin 1/genetics , Cell Adhesion/physiology , Colitis/chemically induced , Colitis/physiopathology , Dextran Sulfate , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Leukocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/physiopathology
6.
Proc Natl Acad Sci U S A ; 105(21): 7540-5, 2008 May 27.
Article in English | MEDLINE | ID: mdl-18508974

ABSTRACT

Chronic tissue ischemia due to defective vascular perfusion is a hallmark feature of peripheral artery disease for which minimal therapeutic options exist. We have reported that sodium nitrite therapy exerts cytoprotective effects against acute ischemia/reperfusion injury in both heart and liver, consistent with the model of bioactive NO formation from nitrite during ischemic stress. Here, we test the hypothesis that chronic sodium nitrite therapy can selectively augment angiogenic activity and tissue perfusion in the murine hind-limb ischemia model. Various therapeutic doses (8.25-3,300 mug/kg) of sodium nitrite or PBS were administered. Sodium nitrite significantly restored ischemic hind-limb blood flow in a time-dependent manner, with low-dose sodium nitrite being most effective. Nitrite therapy significantly increased ischemic limb vascular density and stimulated endothelial cell proliferation. Remarkably, the effects of sodium nitrite therapy were evident within 3 days of the ischemic insult demonstrating the potency and efficacy of chronic sodium nitrite therapy. Sodium nitrite therapy also increased ischemic tissue nitrite and NO metabolites compared to nonischemic limbs. Use of the NO scavenger carboxy PTIO completely abolished sodium nitrite-dependent ischemic tissue blood flow and angiogenic activity consistent with nitrite reduction to NO being the proangiogenic mechanism. These data demonstrate that chronic sodium nitrite therapy is a recently discovered therapeutic treatment for peripheral artery disease and critical limb ischemia.


Subject(s)
Arteries/drug effects , Cytoprotection , Hindlimb/blood supply , Ischemia/drug therapy , Neovascularization, Physiologic/drug effects , Sodium Nitrite/administration & dosage , Animals , Arteries/growth & development , Cyclic N-Oxides/pharmacology , Disease Models, Animal , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Free Radical Scavengers/pharmacology , Imidazoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Peripheral Vascular Diseases/drug therapy , Sodium Nitrite/therapeutic use
7.
Arterioscler Thromb Vasc Biol ; 27(9): 1947-54, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17585066

ABSTRACT

BACKGROUND: Peripheral artery disease (PAD) is a prevalent cardiovascular disorder that results in tissue ischemia which can progress to critical limb ischemia. Restoration of tissue perfusion in the setting of chronic ischemia through stimulation of arteriogenesis and angiogenesis remains a key therapeutic target for PAD. However, experimental therapeutics, including growth factor and gene therapy, have had little clinical success indicating the need for a better understanding of molecular pathways required for therapeutic angiogenesis. METHODS AND RESULTS: Here we report that phosphodiesterase-5 inhibition by sildenafil significantly increases vascular perfusion, tissue blood flow, and vascular density during chronic ischemia of the mouse hind limb. Importantly, sildenafil therapy did not alter any of these parameters in nonischemic limbs. Sildenafil increased tissue cGMP levels independently of increases in nitric oxide production, and sildenafil therapy stimulated angiogenesis in ischemic limbs of eNOS-/- and iNOS-/- mice. Lastly, sildenafil-mediated angiogenic activity was blocked by inhibition of protein kinase G using the PKG antagonist DT-3. CONCLUSIONS: These data demonstrate that sildenafil therapy results in increased angiogenic activity through a PKG-dependent pathway that is independent of nitric oxide production or NOS activity and identify the angiogenic therapeutic potential of sildenafil for critical limb ischemia.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/drug effects , Ischemia/drug therapy , Neovascularization, Physiologic/drug effects , Phosphodiesterase Inhibitors/pharmacology , Piperazines/pharmacology , Sulfones/pharmacology , Animals , Cyclic GMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Hindlimb/blood supply , Ischemia/physiopathology , Male , Mice , Peripheral Vascular Diseases/drug therapy , Purines/pharmacology , Regional Blood Flow/drug effects , Signal Transduction/drug effects , Sildenafil Citrate
8.
Am J Physiol Gastrointest Liver Physiol ; 293(1): G5-G18, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17463183

ABSTRACT

Angiogenesis is now understood to play a major role in the pathology of chronic inflammatory diseases and is indicated to exacerbate disease pathology. Recent evidence shows that angiogenesis is crucial during inflammatory bowel disease (IBD) and in experimental models of colitis. Examination of the relationship between angiogenesis and inflammation in experimental colitis shows that initiating factors for these responses simultaneously increase as disease progresses and correlate in magnitude. Recent studies show that inhibition of the inflammatory response attenuates angiogenesis to a similar degree and, importantly, that inhibition of angiogenesis does the same to inflammation. Recent data provide evidence that differential regulation of the angiogenic mediators involved in IBD-associated chronic inflammation is the root of this pathological angiogenesis. Many factors are involved in this phenomenon, including growth factors/cytokines, chemokines, adhesion molecules, integrins, matrix-associated molecules, and signaling targets. These factors are produced by various vascular, inflammatory, and immune cell types that are involved in IBD pathology. Moreover, recent studies provide evidence that antiangiogenic therapy is a novel and effective approach for IBD treatment. Here we review the role of pathological angiogenesis during IBD and experimental colitis and discuss the therapeutic avenues this recent knowledge has revealed.


Subject(s)
Colitis/physiopathology , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/therapy , Neovascularization, Pathologic , Angiogenesis Inhibitors/therapeutic use , Animals , Antigens, CD/physiology , Cadherins/physiology , Cell Adhesion Molecules , Colon/chemistry , Disease Models, Animal , Humans , Immunoglobulins/physiology , Inflammation Mediators/physiology , Integrin alphaV/drug effects , Integrin alphaV/physiology , Intercellular Adhesion Molecule-1/physiology , Intercellular Signaling Peptides and Proteins/physiology , Matrix Metalloproteinases/physiology , Mice , Microcirculation , Mucoproteins/physiology , Neovascularization, Pathologic/prevention & control , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/physiology , Vascular Endothelial Growth Factor A/physiology
9.
Free Radic Biol Med ; 42(5): 720-9, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17291995

ABSTRACT

Previous studies suggest that inflammatory cell adhesion molecules may modulate endothelial cell migration and angiogenesis through unknown mechanisms. Using a combination of in vitro and in vivo approaches, herein we reveal a novel redox-sensitive mechanism by which ICAM-1 modulates endothelial GSH that controls VEGF-A-induced eNOS activity, endothelial chemotaxis, and angiogenesis. In vivo disk angiogenesis assays showed attenuated VEGF-A-mediated angiogenesis in ICAM-1(-/-) mice. Moreover, VEGF-A-dependent chemotaxis, eNOS phosphorylation, and nitric oxide production were impaired in ICAM-1(-/-) mouse aortic endothelial cells (MAEC) compared to WT MAEC. Decreasing intracellular GSH in ICAM-1(-/-) MAEC to levels observed in WT MAEC with 150 microM buthionine sulfoximine restored VEGF-A responses. Conversely, GSH supplementation of WT MAEC with 5 mM glutathione ethyl ester mimicked defects observed in ICAM-1(-/-) cells. Deficient angiogenic responses in ICAM-1(-/-) cells were associated with increased expression of the lipid phosphatase PTEN, consistent with antagonism of signaling pathways leading to eNOS activation. PTEN expression was also sensitive to GSH status, decreasing or increasing in proportion to intracellular GSH concentrations. These data suggest a novel role for ICAM-1 in modulating VEGF-A-induced angiogenesis and eNOS activity through regulation of PTEN expression via modulation of intracellular GSH status.


Subject(s)
Glutathione/metabolism , Intercellular Adhesion Molecule-1/physiology , Neovascularization, Physiologic , Nitric Oxide Synthase Type II/metabolism , Vascular Endothelial Growth Factor A/physiology , Animals , Aorta/metabolism , Cell Movement , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Nitric Oxide Synthase Type III , PTEN Phosphohydrolase/metabolism
10.
Am J Pathol ; 169(6): 2014-30, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17148665

ABSTRACT

Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders of the intestinal tract with unknown multifactorial etiology that, among other things, result in alteration and dysfunction of the intestinal microvasculature. Clinical observations of increased colon microvascular density during IBD have been made. However, there have been no reports investigating the physiological or pathological importance of angiogenic stimulation during the development of intestinal inflammation. Here we report that the dextran sodium sulfate and CD4+CD45RBhigh T-cell transfer models of colitis stimulate angiogenesis that results in increased blood vessel density concomitant with increased histopathology, suggesting that the neovasculature contributes to tissue damage during colitis. We also show that leukocyte infiltration is an obligatory requirement for the stimulation of angiogenesis. The angiogenic response during experimental colitis was differentially regulated in that the production of various angiogenic mediators was diverse between the two models with only a small group of molecules being similarly controlled. Importantly, treatment with the anti-angiogenic agent thalidomide or ATN-161 significantly reduced angiogenic activity and associated tissue histopathology during experimental colitis. Our findings identify a direct pathological link between angiogenesis and the development of experimental colitis, representing a novel therapeutic target for IBD.


Subject(s)
Blood Vessels/physiopathology , Colitis/chemically induced , Colon/blood supply , Neovascularization, Pathologic/etiology , Thalidomide/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , Colitis/pathology , Colon/pathology , Disease Models, Animal , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration
11.
Lab Invest ; 86(4): 380-90, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16482101

ABSTRACT

Inflammatory bowel diseases (IBD) are chronic inflammatory disorders whose etiology remains unknown. Reports have shown that infiltration of leukocytes into intestinal tissue is a pathognomonic hallmark for this disease. Leukocyte beta(2) integrins are heterodimeric adhesion membrane proteins that are exclusively expressed on leukocytes and participate in immune cell adhesion and activation. In this study, we examined the pathophysiological role of the beta(2) integrins CD18, CD11a, and CD11b in the pathogenesis of dextran sodium sulfte (DSS)-induced experimental colitis. Disease activity was measured by daily assessment of clinical parameters including stool consistency, weight loss, occult blood, and gross rectal bleeding. Histopathological changes including severity of inflammation, surface epithelial/crypt damage, and depth of injury were also determined. The CD18 null and CD11a null mice had significantly lower disease activity and cumulative histopathological scores compared to wild-type mice. Interestingly, CD11b null mice did not show protection against DSS colitis and displayed increased disease activity compared to wild-type mice. Examination of specific leukocyte populations in the distal colon from various mice revealed significant attenuation of neutrophil and macrophage infiltrates in CD18, CD11a, and CD11b null mice. Surprisingly, the CD11b null mice showed a significant increase in plasma cell infiltration in response to DSS suggesting that this molecule may influence plasma cell function during colitis. This study demonstrates that genetic loss of CD18 or CD11a is protective during experimental colitis and that CD11b may serve a regulatory role during development of disease.


Subject(s)
Colitis, Ulcerative/physiopathology , Integrins/physiology , Animals , Bacterial Translocation , CD11a Antigen/metabolism , CD11b Antigen/metabolism , CD18 Antigens/metabolism , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Colon/pathology , Dextran Sulfate , Integrins/metabolism , Leukocytes/metabolism , Leukocytes/physiology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
12.
Am J Pathol ; 165(6): 1849-52, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15579429

ABSTRACT

Leukocyte recruitment into pancreatic islets is believed to play an important pathophysiological role in autoimmune diabetes. Previous reports have suggested that several different adhesion molecules may be involved in leukocyte recruitment during autoimmune diabetes, including members of the leukocyte beta(2) integrins. Here we report that a gene-targeted deficiency of the beta(2) integrin, CD18, protects against multiple low-dose streptozotocin-induced autoimmune diabetes. CD18 null mice displayed lower blood glucose values throughout the study, with only 10% of these mice eventually developing diabetes compared to 95% in the control group. Importantly, the development of insulitis was markedly absent in the CD18 null mice, suggesting that members of this integrin subfamily predominately regulate leukocyte infiltration into pancreatic islets. This study demonstrates that the beta(2) integrins play a key pathophysiological role in the development of multiple low-dose streptozotocin-induced autoimmune diabetes.


Subject(s)
CD18 Antigens/physiology , Diabetes Mellitus, Experimental/prevention & control , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/etiology , Dose-Response Relationship, Drug , Gene Targeting , Hyperglycemia/pathology , Islets of Langerhans/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
Am J Physiol Heart Circ Physiol ; 287(6): H2555-60, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15308480

ABSTRACT

Although the beta2-integrins have been implicated in the pathogenesis of cerebral ischemia-reperfusion (I/R) injury, the relative contributions of the alpha-subunits to the pathogenesis of ischemic stroke remains unclear. The objective of this study was to determine whether and how genetic deficiency of either lymphocyte function-associated antigen-1 (LFA-1) or macrophage-1 (Mac-1) alters the blood cell-endothelial cell interactions, tissue injury, and organ dysfunction in the mouse brain exposed to focal I/R. Middle cerebral artery occlusion was induced for 1 h (followed by either 4 or 24 h of reperfusion) in wild-type mice and in mice with null mutations for either LFA-1 or Mac-1. Neurological deficit and infarct volume were monitored for 24 h after reperfusion. Platelet- and leukocyte-vessel wall adhesive interactions were monitored in cortical venules by intravital microscopy. Mice with null mutations for LFA-1 or Mac-1 exhibited significant reductions in infarct volume. This was associated with a significant improvement in the I/R-induced neurological deficit. Leukocyte adhesion in cerebral venules did not differ between wild-type and mutant mice at 4 h after reperfusion. However, after 24 h of reperfusion, leukocyte adhesion was reduced in both LFA-1- and Mac-1-deficient mice compared with their wild-type counterparts. Platelet adhesion was also reduced at both 4 and 24 h after reperfusion in the LFA-1- and Mac-1-deficient mice. These findings indicate that both alpha-subunits of the beta2-integrins contribute to the brain injury and blood cell-vessel wall interactions that are associated with transient focal cerebral ischemia.


Subject(s)
Cerebrovascular Circulation , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Lymphocyte Function-Associated Antigen-1/genetics , Macrophage-1 Antigen/genetics , Animals , Blood Platelets/cytology , Blood Platelets/physiology , Cell Communication , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Infarction, Middle Cerebral Artery/mortality , Leukocytes/cytology , Leukocytes/physiology , Lymphocyte Function-Associated Antigen-1/metabolism , Macrophage-1 Antigen/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microcirculation
14.
Am J Physiol Cell Physiol ; 285(1): C112-8, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12637264

ABSTRACT

Leukocyte rolling, adhesion, and migration on vascular endothelium involve several sets of adhesion molecules that interact simultaneously. Each of these receptor-ligand pairs may play multiple roles. We examined the role of ICAM-1 in adhesive interactions with mouse aortic endothelial cells (MAECs) in an in vitro flow system. Average rolling velocity of the monocytic cell line WEHI 274.1 was increased on ICAM-1-deficient MAECs compared with wild-type MAECs, both with and without TNF-alpha stimulation. High-temporal-resolution analysis provided insights into the underlying basis for these differences. Without TNF-alpha stimulation, average rolling velocity was slower on wild-type than on ICAM-1-deficient endothelium because of brief (<1 s) pauses. On TNF-alpha-stimulated ICAM-1-deficient endothelium, cells rolled faster because of transient accelerations, producing "jerky" rolling. Firm adhesion to ICAM-1-deficient MAECs was significantly reduced compared with wild-type MAECs, although the number of rolling cells was similar. These results demonstrate directly that ICAM-1 affects rolling velocity by stabilizing leukocyte rolling.


Subject(s)
Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Intercellular Adhesion Molecule-1/metabolism , Monocytes/cytology , Monocytes/metabolism , Animals , Antineoplastic Agents/pharmacology , Aorta/cytology , Cell Adhesion/immunology , Cell Communication/immunology , Cells, Cultured , Endothelium, Vascular/drug effects , Intercellular Adhesion Molecule-1/genetics , Mice , Mice, Mutant Strains , Microscopy, Video , P-Selectin/metabolism , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...