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.
Endocrinology ; 151(6): 2567-76, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20382694

ABSTRACT

We previously reported that 3T3-L1 and rat primary adipocytes secreted microvesicles, known as adipocyte-derived microvesicles (ADMs). In the present study, we further characterized the 3T3-L1 ADMs and found that they exhibited angiogenic activity in vivo. Antibody arrays and gelatin zymography analyses revealed that several angiogenic and antiangiogenic proteins, including leptin, TNFalpha, acidic fibroblast growth factor (FGFa), interferon-gamma, and matrix metalloprotease (MMP)-2 and MMP-9, were present in the ADMs. Gene expression of most of these angiogenic factors was induced in the adipose tissue of diet-induced obese mice. Furthermore, leptin, TNFalpha, and MMP-2 were up-regulated at the protein level in the adipocyte fractions prepared from epididymal adipose tissues of high-fat-diet-induced obese mice. ADMs induced cell migration and tube formation of human umbilical vein endothelial cells, which were partially suppressed by neutralizing antibodies to leptin, TNFalpha, or FGFa but not to interferon-gamma. Supporting these data, a mixture of leptin, TNFalpha, and FGFa induced tube formation. ADMs also promoted cell invasion of human umbilical vein endothelial cells through Matrigel, which was suppressed by the addition of the MMP inhibitor 1,10'-phenanthroline and a neutralizing antibody to MMP-2 but not to MMP-9. These results suggest that ADMs are associated with multiple angiogenic factors and play a role in angiogenesis in adipose tissue.


Subject(s)
Adipocytes/metabolism , Cell-Derived Microparticles/metabolism , Fibroblast Growth Factors/metabolism , Interferon-gamma/metabolism , Leptin/metabolism , Neovascularization, Physiologic/physiology , Tumor Necrosis Factor-alpha/metabolism , 3T3-L1 Cells , Animals , Cell Line , Cell Movement/physiology , Electrophoresis, Polyacrylamide Gel , Endothelial Cells/cytology , Humans , Immunoblotting , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Umbilical Veins/cytology
2.
Histol Histopathol ; 25(6): 765-70, 2010 06.
Article in English | MEDLINE | ID: mdl-20376783

ABSTRACT

Ischemic tissue regeneration depends on neovascularization, the growth of new blood vessels. Bone marrow (BM)-derived cells, including neutrophils, have been shown to contribute to neovascularization during hind limb ischemia and inflammation. Neutrophils produce a broad array of angiogenic growth factors and proteases, which promote remodeling of arterioles into arteries through proteolytic mechanisms. Matrix metalloproteinases (MMPs) have been shown to play a role in the recruitment of neutrophils to sites of inflammation, which requires the extravascular migration of neutrophils through the extracellular matrix. Neutrophils control critical steps during angiogenesis and neutrophil-derived MMPs can promote neoangiogenesis, and collateral growth and perfusion recovery, in part by liberating vital angiogenic growth factors, including vascular endothelial growth factor-A (VEGF-A). This review focuses on the role of neutrophils as key players in the control of the angiogenic process during ischemic tissue regeneration. Aspects of neutrophil regulation, in particular regulation by its major growth factor granulocyte colony-stimulating factor (G-CSF), the role of the unique, readily available, neutrophil-derived MMP-9, and the functional consequences of this MMP-9 activation for angiogenesis, such as MMP-mediated release of biologically relevant cytokines from the matrix and cell surfaces, will be discussed.


Subject(s)
Ischemia/enzymology , Matrix Metalloproteinase 9/metabolism , Neovascularization, Physiologic/physiology , Neutrophils/enzymology , Regeneration/physiology , Animals , Disease Models, Animal , Granulocyte Colony-Stimulating Factor/physiology , Humans , Ischemia/physiopathology , Mice , Mice, Knockout
3.
Blood ; 115(21): 4302-12, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20110420

ABSTRACT

Ischemia of the heart, brain, and limbs is a leading cause of morbidity and mortality worldwide. Treatment with tissue type plasminogen activator (tPA) can dissolve blood clots and can ameliorate the clinical outcome in ischemic diseases. But the underlying mechanism by which tPA improves ischemic tissue regeneration is not well understood. Bone marrow (BM)-derived myeloid cells facilitate angiogenesis during tissue regeneration. Here, we report that a serpin-resistant form of tPA by activating the extracellular proteases matrix metalloproteinase-9 and plasmin expands the myeloid cell pool and mobilizes CD45(+)CD11b(+) proangiogenic, myeloid cells, a process dependent on vascular endothelial growth factor-A (VEGF-A) and Kit ligand signaling. tPA improves the incorporation of CD11b(+) cells into ischemic tissues and increases expression of neoangiogenesis-related genes, including VEGF-A. Remarkably, transplantation of BM-derived tPA-mobilized CD11b(+) cells and VEGFR-1(+) cells, but not carrier-mobilized cells or CD11b(-) cells, accelerates neovascularization and ischemic tissue regeneration. Inhibition of VEGF signaling suppresses tPA-induced neovascularization in a model of hind limb ischemia. Thus, tPA mobilizes CD11b(+) cells from the BM and increases systemic and local (cellular) VEGF-A, which can locally promote angiogenesis during ischemic recovery. tPA might be useful to induce therapeutic revascularization in the growing field of regenerative medicine.


Subject(s)
Myeloid Cells/drug effects , Myeloid Cells/physiology , Neovascularization, Physiologic/drug effects , Regeneration/drug effects , Tissue Plasminogen Activator/pharmacology , Animals , Base Sequence , Bone Marrow Transplantation , CD11b Antigen/metabolism , DNA Primers/genetics , Female , Gene Expression/drug effects , Ischemia/drug therapy , Ischemia/pathology , Ischemia/physiopathology , Male , Matrix Metalloproteinase 9/deficiency , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutant Proteins/pharmacology , Neovascularization, Physiologic/genetics , Plasminogen/deficiency , Plasminogen/genetics , Plasminogen/metabolism , Recombinant Proteins/pharmacology , Regeneration/physiology , Signal Transduction , Stem Cell Factor/metabolism , Tissue Plasminogen Activator/deficiency , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/physiology , Transplantation Chimera , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
4.
J Cell Physiol ; 221(3): 521-5, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19681053

ABSTRACT

Hematopoietic stem cells (HSCs) can differentiate and proliferate in response to hematopoietic stress (e.g., myelosuppression, infections, and allergic reactions), thereby ensuring a well-regulated supply of mature and immature hematopoietic cells within the circulation and prompt adjustment of blood cell levels within normal ranges. The recovery of tissues and organs from hematopoietic stress (e.g., myelosuppression or ionizing irradiation) is dependent on two cell types: resident HSCs which repopulate the bone marrow (BM) cavity, and stromal cells. BM regeneration critically depends on the release of soluble factors from cells such as stromal cells, a process regulated by proteases. Two proteolytic systems, the fibrinolytic system and the matrix metalloproteinases (MMPs), have recently been shown to be involved in this process (Heissig B, 2007, Cell Stem Cell 1: 658-670). The plasminogen/plasmin system is mostly recognized for its fibrinolytic activity, but it is also involved in processes such as cell invasion, chemotaxis, growth factor activity modulation, and tissue remodeling. This review focuses on the role of plasmin and its activators as key players in controlling the hematopoietic stress response after myelosuppression (hematopoietic regeneration). Aspects of plasmin regulation, especially regulation of its ability to activate MMPs and the functional consequences of this enzyme activation, such as plasmin-mediated release of biologically relevant cytokines from the matrix and cell surfaces, will be discussed.


Subject(s)
Fibrinolysis/physiology , Hematopoiesis/physiology , Regeneration/physiology , Animals , Bone Marrow/metabolism , Fibrinolysin/metabolism , Humans , Matrix Metalloproteinases/metabolism , Models, Biological , Plasminogen Activators/metabolism
5.
Cell Stem Cell ; 1(6): 658-70, 2007 Dec 13.
Article in English | MEDLINE | ID: mdl-18371407

ABSTRACT

Hematopoietic stem cells within the bone marrow exist in a quiescent state. They can differentiate and proliferate in response to hematopoietic stress (e.g., myelosuppression), thereby ensuring a well-regulated supply of mature and immature hematopoietic cells within the circulation. However, little is known about how this stress response is coordinated. Here, we show that plasminogen (Plg), a classical fibrinolytic factor, is a key player in controlling this stress response. Deletion of Plg in mice prevented hematopoietic stem cells from entering the cell cycle and undergoing multilineage differentiation after myelosuppression, leading to the death of the mice. Activation of Plg by administration of tissue-type plasminogen activator promoted matrix metalloproteinase-mediated release of Kit ligand from stromal cells, thereby promoting hematopoietic progenitor cell proliferation and differentiation. Thus, activation of the fibrinolytic cascade is a critical step in regulating the hematopoietic stress response.


Subject(s)
Fibrinolysis , Hematopoiesis , Hematopoietic Stem Cells/cytology , Plasminogen/genetics , Animals , Bone Marrow Transplantation , Cell Differentiation , Cell Proliferation , Cells, Cultured , Gene Deletion , Hematopoiesis/genetics , Hematopoiesis/physiology , Matrix Metalloproteinases/drug effects , Mice , Mice, Knockout , Plasminogen/pharmacology , Signal Transduction , Stem Cell Factor/drug effects , Stem Cell Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...