Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Matrix Biol ; 37: 35-48, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24589453

ABSTRACT

Thrombospondins (TSPs) are multifunctional proteins that are deposited in the extracellular matrix where they directly affect the function of vascular and other cell types. TSP-4, one of the 5 TSP family members, is expressed abundantly in tendon and muscle. We have examined the effect of TSP-4 deficiency on tendon collagen and skeletal muscle morphology and function. In Thbs4(-/-) mice, tendon collagen fibrils are significantly larger than in wild-type mice, and there is no compensatory over-expression of TSP-3 and TSP-5, the two TSPs most highly homologous to TSP-4, in the deficient mice. TSP-4 is expressed in skeletal muscle, and higher levels of TSP-4 protein are associated with the microvasculature of red skeletal muscle with high oxidative metabolism. Lack of TSP-4 in medial soleus, red skeletal muscle with predominant oxidative metabolism, is associated with decreased levels of several specific glycosaminoglycan modifications, decreased expression of a TGFß receptor beta-glycan, decreased activity of lipoprotein lipase, which associates with vascular cell surfaces by binding to glycosaminoglycans, and decreased uptake of VLDL. The soleus muscle is smaller and hind- and fore-limb grip strength is reduced in Thbs4(-/-) mice compared to wild-type mice. These observations suggest that TSP-4 regulates the composition of the ECM at major sites of its deposition, tendon and muscle, and the absence of TSP-4 alters the organization, composition and physiological functions of these tissues.


Subject(s)
Collagen/metabolism , Extracellular Matrix Proteins/physiology , Muscle, Skeletal/physiology , Tendons/physiology , Thrombospondins/genetics , Thrombospondins/physiology , Animals , Blotting, Western , DNA Primers/genetics , Endothelial Cells/metabolism , Extracellular Matrix Proteins/metabolism , Glycosaminoglycans/metabolism , Heparitin Sulfate/metabolism , Immunohistochemistry , Lipoproteins, VLDL/metabolism , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron, Transmission , Muscle Strength/physiology , Muscle, Skeletal/metabolism , Tendons/metabolism , Thrombospondins/metabolism
2.
Circulation ; 127(11): 1209-18, e1-16, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23401155

ABSTRACT

BACKGROUND: Deciphering the molecular and cellular processes that govern macrophage foam cell formation is critical to understanding the basic mechanisms underlying atherosclerosis and other vascular pathologies. METHODS AND RESULTS: Here, we identify a pivotal role of plasminogen (Plg) in regulating foam cell formation. Deficiency of Plg inhibited macrophage cholesterol accumulation on exposure to hyperlipidemic conditions in vitro, ex vivo, and in vivo. Gene expression analysis identified CD36 as a regulated target of Plg, and macrophages from Plg(-/-) mice had decreased CD36 expression and diminished foam cell formation. The Plg-dependent CD36 expression and foam cell formation depended on conversion of Plg to plasmin, binding to the macrophage surface, and the consequent intracellular signaling that leads to production of leukotriene B4. Leukotriene B4 rescued the suppression of CD36 expression and foam cell formation arising from Plg deficiency. CONCLUSIONS: Our findings demonstrate an unanticipated role of Plg in the regulation of gene expression and cholesterol metabolism by macrophages and identify Plg-mediated regulation of leukotriene B4 as an underlying mechanism.


Subject(s)
Cell Differentiation/physiology , Foam Cells/cytology , Foam Cells/metabolism , Gene Expression Regulation/physiology , Macrophages/cytology , Macrophages/metabolism , Plasminogen/physiology , Animals , CD36 Antigens/metabolism , Cholesterol/metabolism , In Vitro Techniques , Leukotriene B4/metabolism , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Plasminogen/deficiency , Plasminogen/genetics , Signal Transduction/physiology
3.
FASEB J ; 26(6): 2363-73, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22362893

ABSTRACT

Thrombospondin-4 (TSP-4) expression increases dramatically in hypertrophic and failing hearts in rodent models and in humans. The aim of this study was to address the function of TSP-4 in the heart. TSP-4-knockout (Thbs4(-/-)) and wild-type (WT) mice were subjected to transverse aortic constriction (TAC) to increase left ventricle load. After 2 wk, Thbs4(-/-) mice had a significantly higher heart weight/body weight ratio than WT mice. The additional increase in the heart weight in TAC Thbs4(-/-) mice was due to increased deposition of extracellular matrix (ECM). The levels of interstitial collagens were higher in the knockout mice, but the size of cardiomyocytes and apoptosis in the myocardium was unaffected by TSP-4 deficiency, suggesting that increased reactive fibrosis was the primary cause of the higher heart weight. The increased ECM deposition in Thbs4(-/-) mice was accompanied by changes in functional parameters of the heart and decreased vessel density. The expression of inflammatory and fibrotic genes known to be influential in myocardial remodeling changed as a result of TSP-4 deficiency in vivo and as a result of incubation of cells with recombinant TSP-4 in vitro. Thus, TSP-4 is involved in regulating the adaptive responses of the heart to pressure overload, suggesting its important role in myocardial remodeling. Our study showed a direct influence of TSP-4 on heart function and to identify the mechanism of its effects on heart remodeling.


Subject(s)
Cardiomegaly/physiopathology , Myocardium/pathology , Thrombospondins/deficiency , Ventricular Remodeling/physiology , Animals , Aorta/pathology , Collagen/biosynthesis , Constriction, Pathologic/physiopathology , Extracellular Matrix/metabolism , Heart Failure/physiopathology , Heart Ventricles/pathology , Mice , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/pathology , Thrombospondins/physiology
4.
Circ Res ; 107(11): 1313-25, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-20884877

ABSTRACT

RATIONALE: Thrombospondin (TSP)-4 is an extracellular protein that has been linked to several cardiovascular pathologies. However, a role for TSP-4 in vascular wall biology remains unknown. OBJECTIVE: We have examined the effects of TSP-4 gene (Thbs4) knockout on the development of atherosclerotic lesions in ApoE(-/-) mice. METHODS AND RESULTS: Deficiency in TSP-4 reduced atherosclerotic lesions: at 20 weeks of age, the size of the aortic root lesions in Thbs4(-/-)/ApoE(-/-) mice was decreased by 48% in females and by 39% in males on chow diets; in mice on Western diets, lesions in the descending aorta were reduced by 30% in females and 33% in males. In ApoE(-/-) mice, TSP-4 was abundant in vessel areas prone to lesion development and in the matrix of the lesions themselves. TSP-4 deficiency reduced the number of macrophages in lesions in all groups by ≥ 2-fold. In addition, TSP-4 deficiency reduced endothelial cell activation (expression of surface adhesion molecules) and other markers of inflammation in the vascular wall (decreased production of monocyte chemoattractant protein-1 and activation of p38). In vitro, both the adhesion and migration of wild-type macrophages increased in the presence of purified recombinant TSP-4 in a dose-dependent manner (up to 7- and 4.7-fold, respectively). These responses led to p38-MAPkinase activation and were dependent on ß(2) and ß(3) integrins, which recognize TSP-4 as a ligand. CONCLUSIONS: TSP-4 is abundant in atherosclerotic lesions and in areas prone to development of lesions and may influence the recruitment of macrophages by activating endothelial cells and directly interacting with macrophages to increase their adhesion and migration. Our observations suggest an important role for this matricellular protein in the local regulation of inflammation associated with atherogenesis.


Subject(s)
Atherosclerosis/metabolism , Inflammation Mediators/physiology , Thrombospondins/physiology , Vascular Diseases/metabolism , Animals , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/physiology , Female , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Thrombospondins/deficiency , Vascular Diseases/pathology , Vascular Diseases/physiopathology
5.
Vascular ; 13(4): 230-5, 2005.
Article in English | MEDLINE | ID: mdl-16229796

ABSTRACT

Biomaterial implants induce a local inflammatory response. A comparison of the inflammatory cell response was made between several biomaterials commonly used as vascular prostheses. Disks of polyethylene terephthalate (PET), polytetrafluoroethylene (PTFE), aluminum, titanium, copper, and stainless steel were surgically placed into the peritoneum of mice. Recruited macrophage and neutrophil populations were measured after recovery from the disk surface and peritoneal lavage. Following peritoneal biomaterial implants, there was no difference in total neutrophil or macrophage recruitment between mice implanted with PET, PTFE, aluminum, or titanium disks. However, there was significant attenuation of total neutrophil and macrophage recruitment to stainless steel compared with the other implants. Similarly, there was no significant difference in the percentage of leukocytes adherent to the PET, aluminum, or titanium disks. Macrophage adherence to the stainless steel disks was attenuated by 19.1%, and the number of neutrophils was attenuated by 69.1% when compared with PET implant mice. Mice implanted with copper disks universally expired. Leukocyte recruitment did not differ between PET, PTFE, aluminum, or titanium disks, suggesting that these materials stimulate similar inflammatory responses. Stainless steel disks recruited both fewer neutrophils and fewer macrophages and support lower adherence of these cells than the other biomaterials. Copper incited an overwhelming and fatal response.


Subject(s)
Biocompatible Materials/adverse effects , Inflammation/immunology , Macrophages/immunology , Neutrophils/immunology , Animals , Mice , Mice, Inbred C57BL , Prostheses and Implants
SELECTION OF CITATIONS
SEARCH DETAIL
...