Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Glycobiology ; 33(1): 17-37, 2023 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-36190502

RESUMO

O-linked ß-N-acetylglucosamine (O-GlcNAc)-modified proteins are post-translationally modified with GlcNAc conjugated to serine and threonine residues. This modification is associated with various physiological functions such as serine and threonine phosphorylation and Notch signaling. Here, we demonstrated that O-GlcNAc-modified proteins leaked from dead cells and GlcNAc-bearing polymers mimicking the multivalent GlcNAc moiety of these proteins induced anti-fibrotic activities, such as the suppression of α-smooth muscle actin and collagen and the induction of matrix metalloprotease 1 in myofibroblasts. We have previously reported that O-GlcNAc-modified proteins and GlcNAc-bearing polymers could interact with cell surface vimentin and desmin. In the current study, it was demonstrated that a multivalent GlcNAc moiety structure of these molecules activated PI3K/Akt and p38MAPK pathway and elicited these anti-fibrotic activities in myofibroblasts by interacting with cell surface vimentin. Since the interaction of O-GlcNAc-modified proteins with desmin was observed in the fibrotic liver of carbon tetrachloride-treated mice via an in situ proximity ligation assay, it was assumed that the activated stellate cells could bind to the O-GlcNAc-modified proteins from the damaged hepatocytes. In addition, the administration of anti-O-GlcNAc antibody to inhibit the interaction exacerbated liver fibrosis in the mice. Moreover, administration of the GlcNAc-bearing polymers into carbon tetrachloride-treated mice could ameliorate liver fibrosis. Thus, O-GlcNAc-modified proteins leaked from dead cells can interact with myofibroblasts and activated stellate cells and function as fibrosis suppressors. Moreover, we anticipate that GlcNAc-bearing polymers mimicking O-GlcNAc-modified proteins will be applied as novel therapeutic tools for fibrosis.


Assuntos
Acetilglucosamina , Miofibroblastos , Animais , Camundongos , Acetilglucosamina/metabolismo , Materiais Biomiméticos/farmacologia , Tetracloreto de Carbono , Desmina/metabolismo , Cirrose Hepática , Miofibroblastos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Polímeros/química , Polímeros/metabolismo , Processamento de Proteína Pós-Traducional , Serina/metabolismo , Vimentina/química , Vimentina/metabolismo , Células Estreladas do Fígado/metabolismo
2.
Genes Cells ; 25(6): 413-426, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32243065

RESUMO

Vimentin, desmin, glial fibrillary acidic protein (GFAP) and peripherin, classified as the type III intermediate filament family, maintain the integrity and architecture of various cell types. Recently, we reported their cell surface expression and binding to multivalent N-acetylglucosamine-conjugated polymers. Furthermore, the presence of vimentin on the surface of various cell types including malignant tumor cells and fibroblasts has been demonstrated. Type III intermediate filament proteins are traditionally considered intracellular proteins and do not possess signal peptides for cell membrane recruitment. Therefore, the mechanism of their transport to the cell surface is unclear. In the current study, we aimed to elucidate this mechanism by focusing on the relationship between their multimeric structure and lipid bilayer affinity. Blue native polyacrylamide gel electrophoresis demonstrated that cell surface-expressed type III intermediate filament proteins formed a multimeric mostly including 4-12-mers but not filamentous structure. Moreover, surface plasmon resonance analysis revealed that the multimeric structure of these recombinant proteins had high affinity to lipid bilayers, whereas their filament-like large multimeric structure did not. Our results suggest that type III intermediate filaments are incorporated into the cell membrane through alteration from a filamentous to a multimeric structure.


Assuntos
Membrana Celular/metabolismo , Proteínas de Filamentos Intermediários/metabolismo , Filamentos Intermediários/metabolismo , Bicamadas Lipídicas/metabolismo , Acetilglucosamina/metabolismo , Animais , Linhagem Celular Tumoral , Desmina/genética , Desmina/metabolismo , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Proteínas de Filamentos Intermediários/genética , Cinética , Camundongos , Microscopia de Força Atômica , Periferinas/genética , Periferinas/metabolismo , Ligação Proteica , Domínios Proteicos , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes , Ressonância de Plasmônio de Superfície , Fatores de Tempo , Vimentina/genética , Vimentina/metabolismo
3.
Genes Cells ; 22(10): 900-917, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28898551

RESUMO

Vimentin, desmin, glial fibrillary acidic protein (GFAP) and peripherin belong to type III intermediate filament family and are expressed in mesenchymal cells, skeletal muscle cells, astrocytes and peripheral neurons, respectively. Vimentin and desmin possess N-acetyl-d-glucosamine (GlcNAc)-binding properties on cell surfaces. The rod II domain of these proteins is a GlcNAc-binding site, which also exists in GFAP and peripherin. However, the GlcNAc-binding activities and behaviors of these proteins remain unclear. Here, we characterized the interaction and binding behaviors of these proteins, using various well-defined GlcNAc-bearing polymers synthesized by radical polymerization with a reversible addition-fragmentation chain transfer reagent. The small GlcNAc-bearing polymers strongly interacted with HeLa cells through vimentin expressed on the cell surface and interacted with vimentin-, desmin-, GFAP- and peripherin-transfected vimentin-deficient HeLa cells. These proteins present high affinity to GlcNAc-bearing polymers, as shown by surface plasmon resonance. These results show that type III intermediate filament proteins possess GlcNAc-binding activities on cell surfaces. These findings provide important insights into novel cellular functions and physiological significance of type III intermediate filaments.


Assuntos
Acetilglucosamina/análogos & derivados , Desmina/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Vimentina/metabolismo , Acetilglucosamina/química , Acetilglucosamina/metabolismo , Sítios de Ligação , Células HeLa , Humanos , Polimerização , Polímeros/química , Ligação Proteica
4.
Glycobiology ; 22(6): 788-805, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22345628

RESUMO

The clearance of apoptotic cells is important to maintain tissue homeostasis. The engulfment of apoptotic cells is performed by professional phagocytes, such as macrophages, and also by non-professional phagocytes, such as mesenchymal cells. Here, we show that vimentin, a cytoskeletal protein, functions as an engulfment receptor on neighboring phagocytes, which recognize O-linked ß-N-acetylglucosamine (O-GlcNAc)-modified proteins from apoptotic cells as "eat me" ligands. Previously, we reported that vimentin possesses a GlcNAc-binding lectin-like property on cell surface. However, the physiological relevance of the surface localization and GlcNAc-binding property of vimentin remained unclear. In the present study, we observed that O-GlcNAc proteins from apoptotic cells interacted with the surface vimentin of neighboring phagocytes and that this interaction induced serine 71-phosphorylation and recruitment of vimentin to the cell surface of the neighboring phagocytes. Moreover, tetrameric vimentin that was disassembled by serine 71-phosphorylation possessed a GlcNAc-binding activity and was localized to the cell surface. We demonstrated our findings in vimentin-expressing common cell lines such as HeLa cells. Furthermore, during normal developmental processes, the phagocytic engulfment and clearance of apoptotic footplate cells in mouse embryos was mediated by the interaction of surface vimentin with O-GlcNAc proteins. Our results suggest a common mechanism for the clearance of apoptotic cells, through the interaction of surface vimentin with O-GlcNAc-modified proteins.


Assuntos
Acetilglucosamina/metabolismo , Apoptose , Lectinas/metabolismo , Prenhez , Vimentina/metabolismo , Animais , Sítios de Ligação , Feminino , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos ICR , Gravidez , Propriedades de Superfície , Células Tumorais Cultivadas , Raios Ultravioleta
5.
Glycobiology ; 22(12): 1741-59, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22846177

RESUMO

The cytoskeleton protein vimentin is dramatically altered following pathological events such as fibrosis and tumorigenesis. Vimentin binds to multivalent N-acetylglucosamine (GlcNAc) molecules at the cell surface and interacts with O-linked ß-GlcNAc proteins. Moreover, dying cells can be engulfed by neighboring cells through surface interactions between vimentin and many O-GlcNAc proteins in cell debris. Here, we show that vimentin was altered by its interaction with GlcNAc-bearing molecules such as GlcNAc-bearing polymers. The interaction with GlcNAc-bearing polymers promoted the cell surface recruitment of vimentin followed by the phosphorylation of vimentin serine 71 and the increase in tetrameric vimentin disassembled from vimentin filaments in HeLa cells. Moreover, it was found that GlcNAc-bearing polymers and O-GlcNAc proteins from dying cells promoted vimentin expression and cell migration in the Madin-Darby canine kidney and Michigan Cancer Foundation-7 cells. These results suggest that interactions between surface vimentin and GlcNAc molecules, including the O-GlcNAc proteins from dying cells, may play a pivotal role in vimentin expression and the migration of cancer cells. We propose new mechanisms of vimentin expression in cancer cells.


Assuntos
Acetilglucosamina/metabolismo , Glicoproteínas de Membrana/metabolismo , Vimentina/metabolismo , Animais , Morte Celular , Movimento Celular , Cães , Regulação Neoplásica da Expressão Gênica , Células HeLa , Humanos , Células Madin Darby de Rim Canino , Mutação de Sentido Incorreto , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Serina/genética , Vimentina/química , Vimentina/genética
6.
Biotechnol Lett ; 34(9): 1765-71, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22661013

RESUMO

To enhance vascularization of hydrophobic implants in vivo, a VEGF-Fc fusion protein consisting of vascular endothelial growth factor (VEGF) fused to the immunoglobulin G Fc domain was prepared as an artificial extracellular matrix (ECM). VEGF-Fc was stably immobilized on a polystyrene plate due to the hydrophobicity of the Fc domain, and significantly enhanced the adhesion of human umbilical vein endothelial cells (HUVECs). Additionally, the use of VEGF-Fc as an ECM markedly promoted the proliferation of HUVECs longer than 72 h and induced the reorganization of actin filaments into larger stress fibers within these cells. The VEGF-Fc fusion protein may be a promising artificial ECM for enhancing endothelial cell growth.


Assuntos
Proliferação de Células , Células Endoteliais/fisiologia , Técnicas de Cultura de Células/métodos , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Poliestirenos/química , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
J Artif Organs ; 14(4): 301-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21809097

RESUMO

The targeted delivery of anti-inflammatory agents has great therapeutic potential for treating restenosis following percutaneous coronary intervention. To develop a drug delivery system targeted to injured blood vessels, we examined whether N-acetylglucosamine (GlcNAc)-bearing polymer-coated liposomes (GlcNAc-Ls) are specifically taken up by vascular smooth muscle cells (VSMCs). Flow cytometric analysis revealed that GlcNAc-Ls were taken up by VSMCs in vitro. Furthermore, GlcNAc-Ls were intravenously administered to mice that had undergone wire-mediated vascular injury. GlcNAc-Ls markedly accumulated at the intramural site of the injured vessel walls but not at the contralateral (uninjured) vessel walls. These results demonstrated that GlcNAc-Ls can be specifically taken up by VSMCs both in vitro and in vivo. We propose a novel strategy of using GlcNAc-Ls that has potential for application in drug delivery targeted to injured blood vessels.


Assuntos
Acetilglucosamina/farmacocinética , Sistemas de Liberação de Medicamentos , Lipossomos/farmacocinética , Células Musculares/metabolismo , Músculo Liso Vascular/metabolismo , Angioplastia , Animais , Animais Recém-Nascidos , Anti-Inflamatórios/administração & dosagem , Células Cultivadas , Reestenose Coronária/terapia , Injeções Intravenosas , Interleucina-6/metabolismo , NF-kappa B/farmacocinética , Ratos , Ratos Sprague-Dawley , Lesões do Sistema Vascular/tratamento farmacológico , Vimentina/metabolismo
8.
Colloids Surf B Biointerfaces ; 198: 111406, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33250416

RESUMO

We have reported that cytoskeletal proteins such as desmin and vimentin are expressed on the surface of muscle, mesenchymal and cancer cells, and possess N-acetyl-ß-D-glucosamine (ß-GlcNAc) residue-binding properties. As cell-recognizable ß-GlcNAc residue-bearing biopolymer, we prepared glycoconjugates (SF-GlcNAc) composed of silk fibroin (SF) and monosaccharide N-acetyl-D-glucosamine (GlcNAc) by chemical modification using cyanuric chloride. The covalent immobilization of GlcNAc into SF was assessed by 1H-NMR measurements. The 1H-NMR spectrum of SF-GlcNAc conjugates showed new peaks attributed to the methyl protons of the N-acetyl group in GlcNAc, and the integration of these peaks revealed that the GlcNAc content in the conjugates was 9 wt%. The existence of ß-GlcNAc residues in SF-GlcNAc was examined by the criteria using lectins such as wheat germ agglutinin (WGA). Addition of WGA to SF-GlcNAc solution caused an increase in the turbidity of the solution due to lectin-mediated aggregation. Solid-phase lectin binding assay based on the biotin-avidin interaction showed that biotinylated succinylated WGA bound more strongly onto SF-GlcNAc conjugate-coated wells compared to SF-coated well. Following the establishment of the existence of ß-GlcNAc residues in SF-GlcNAc, the interaction of SF-GlcNAc with desmin was examined by enzyme-linked immunosorbent assay using anti-desmin antibody. The stronger binding of desmin was observed for SF-GlcNAc conjugate-coated wells compared to SF-coated wells. The use of SF-GlcNAc conjugates as a substrate for culturing desmin-expressing human cardiac myocytes demonstrated an increase in the numbers of attached cells and proliferating cells on the conjugate-coated wells compared to SF-coated wells. These results suggest that the immobilization of monosaccharide GlcNAc is a useful method for the versatile functionalization of SF as an application in tissue engineering.


Assuntos
Fibroínas , Acetilglucosamina , Proteínas do Citoesqueleto , Glucosamina , Humanos , Lectinas , Miócitos Cardíacos
9.
Glycobiology ; 20(7): 843-64, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20332081

RESUMO

Vimentin and desmin are intermediate filament proteins found in various mesenchymal and skeletal muscle cells, respectively. These proteins play an important role in the stabilization of the cytoplasmic architecture. Here, we found, using artificial biomimicking glycopolymers, that vimentin and desmin possess N-acetylglucosamine (GlcNAc)-binding lectin-like properties on the cell surfaces of various vimentin- and desmin-expressing cells such as cardiomyocytes and vascular smooth muscle cells. The rod II domain of these proteins was demonstrated to be localized to the cell surface and to directly bind to the artificial biomimicking GlcNAc-bearing polymer, by confocal laser microscopy and surface plasmon resonance analysis. These glycopolymers strongly interact with lectins and are useful tools for the analysis of lectin-carbohydrate interactions, since glycopolymers binding to lectins can induce the clustering of lectins due to multivalent glycoside ligand binding. Moreover, immunocytochemistry and pull-down assay with His-tagged vimentin-rod II domain protein showed that the vimentin-rod II domain interacts with O-GlcNAc proteins. These results suggest that O-GlcNAc proteins might be one candidate for physiological GlcNAc-bearing ligands with which vimentin and desmin interact. These findings demonstrate a novel function of vimentin and desmin that does not involve stabilization of the cytoplasmic architecture by which these proteins interact with physiological GlcNAc-bearing ligands such as O-GlcNAc proteins on the cell surface through their GlcNAc-binding lectin-like properties.


Assuntos
Desmina/metabolismo , Receptores de N-Acetilglucosamina/química , Vimentina/metabolismo , Animais , Membrana Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Desmina/análise , Desmina/química , Células HeLa , Humanos , Imuno-Histoquímica , Lectinas/metabolismo , Ligantes , Camundongos , Modelos Moleculares , Receptores de N-Acetilglucosamina/metabolismo , Ressonância de Plasmônio de Superfície , Vimentina/análise , Vimentina/química
10.
Polymers (Basel) ; 12(7)2020 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-32645972

RESUMO

Targeting myofibroblasts and activated stellate cells in lesion sites of fibrotic tissues is an important approach to treat fibroses. Herein, we focused on targeting the cytoskeletal proteins vimentin, which are reportedly highly expressed on the surface of these cells and have N-acetylglucosamine (GlcNAc)-binding activity. A GlcNAc-bearing polymer synthesized via radical polymerization with a reversible addition-fragmentation chain transfer reagent has been previously found to interact with cell-surface vimentin-expressing cells. We designed a GlcNAc-bearing polymer-conjugated polyethyleneimine (PEI), as the gene carrier to target cell-surface vimentin-expressing cells and specifically deliver nuclear factor-κB decoy oligonucleotides (ODNs) and heat shock protein 47 (HSP47)-small interfering RNA (siRNA) to normal human dermal fibroblasts (NHDFs) that express cell-surface vimentin. The results showed that the expression of tumor necrosis factor-α in lipopolysaccharide-stimulated NHDFs and HSP47 in transforming growth factor-ß1-stimulated NHDFs was suppressed by cellular uptake of the GlcNAc-bearing polymer-conjugated PEI/nuclear factor (NF)-κB decoy ODNs and HSP47-siRNA complexes through cell-surface vimentin, respectively. These findings suggest that the effective and specific delivery of ODNs and siRNA for cell-surface vimentin-expressing cells such as myofibroblasts and activated stellate cells can be achieved using GlcNAc-bearing polymer-conjugated PEI. This therapeutic approach could prove advantageous to prevent the promotion of various fibroses.

11.
Circulation ; 117(24): 3079-87, 2008 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-18541743

RESUMO

BACKGROUND: Inflammatory cytokines such as interleukin (IL)-1 beta and IL-18 play an important role in the development of atherosclerosis and restenosis. Apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) is an adaptor protein that regulates caspase-1-dependent IL-1 beta and IL-18 generation; however, the role of ASC in vascular injury remains undefined. Here, we investigated the contribution of ASC to neointimal formation after vascular injury in ASC-deficient (ASC(-/-)) mice. METHODS AND RESULTS: Wire-mediated vascular injury was produced in the femoral artery of ASC(-/-) and wild-type mice. Immunohistochemical analysis revealed that ASC was markedly expressed at the site of vascular injury. Neointimal formation was significantly attenuated in ASC(-/-) mice after injury. IL-1 beta and IL-18 were expressed in the neointimal lesion in wild-type mice but showed decreased expression in the lesion of ASC(-/-) mice. To investigate the contribution of bone marrow-derived cells, we developed bone marrow-transplanted mice and found that neointimal formation was significantly decreased in wild-type mice in which bone marrow was replaced with ASC(-/-) bone marrow cells. Furthermore, in vitro experiments showed that the proliferation activity of ASC(-/-) vascular smooth muscle cells was not impaired. CONCLUSIONS: These findings suggest that bone marrow-derived ASC is critical for neointimal formation after vascular injury and identify ASC as a novel therapeutic target for atherosclerosis and restenosis.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Proteínas do Citoesqueleto/deficiência , Túnica Íntima/fisiopatologia , Doenças Vasculares/patologia , Doenças Vasculares/fisiopatologia , Animais , Apoptose , Proteínas Reguladoras de Apoptose , Transplante de Medula Óssea , Proteínas Adaptadoras de Sinalização CARD , Caspases/deficiência , Caspases/genética , Caspases/metabolismo , Técnicas de Cultura de Células , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/genética , Artéria Femoral/lesões , Artéria Femoral/patologia , Artéria Femoral/fisiopatologia , Imuno-Histoquímica , Inflamação/patologia , Inflamação/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Túnica Íntima/patologia
12.
Cardiovasc Res ; 78(3): 554-62, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18267955

RESUMO

AIMS: Monocyte chemoattractant protein-1 (MCP-1: CCL2) has been demonstrated to be involved in the pathophysiology of ischaemic heart disease; however, the precise role of MCP-1 in ischaemia/reperfusion (I/R) injury is controversial. Here, we investigated the role of cardiac MCP-1 expression on left ventricular (LV) dysfunction after global I/R in Langendorff-perfused hearts isolated from transgenic mice expressing the mouse JE-MCP-1 gene under the control of the alpha-cardiac myosin heavy chain promoter (MHC/MCP-1 mice). METHODS AND RESULTS: In vitro experiments showed that MCP-1 prevented the apoptosis of murine neonatal cardiomyocytes after hypoxia/reoxygenation. I/R significantly increased the mRNA expression of MCP-1 in the Langendorff-perfused hearts of wild-type mice. Cardiac MCP-1 overexpression in the MHC/MCP-1 mice improved LV dysfunction after I/R without affecting coronary flow; in particular, it ameliorated LV diastolic pressure after reperfusion. This improvement was independent of both sarcolemmal and mitochondrial K(ATP) channels. Cardiac MCP-1 overexpression prevented superoxide generation in the I/R hearts, and these hearts showed decreased expression of the NADPH oxidase family proteins Nox1, gp91phox, and Nox3 compared with the hearts of wild-type mice. Further, superoxide dismutase activity in the hearts of MHC/MCP-1 mice was significantly increased compared with that in the hearts of wild-type mice. CONCLUSION: These findings suggest that cardiac MCP-1 prevented LV dysfunction after global I/R through a reactive oxygen species-dependent but K(ATP) channel-independent pathway; this provides new insight into the beneficial role of MCP-1 in the pathophysiology of ischaemic heart diseases.


Assuntos
Cardiotônicos/metabolismo , Quimiocina CCL2/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Disfunção Ventricular Esquerda/prevenção & controle , Animais , Animais Recém-Nascidos , Apoptose , Hipóxia Celular , Células Cultivadas , Quimiocina CCL2/genética , Modelos Animais de Doenças , Isoenzimas/metabolismo , Canais KATP/antagonistas & inibidores , Canais KATP/metabolismo , Camundongos , Camundongos Transgênicos , Traumatismo por Reperfusão Miocárdica/complicações , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Cadeias Pesadas de Miosina/genética , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , RNA Mensageiro/metabolismo , Superóxido Dismutase/metabolismo , Fatores de Tempo , Disfunção Ventricular Esquerda/etiologia , Disfunção Ventricular Esquerda/metabolismo , Miosinas Ventriculares/genética , Pressão Ventricular
13.
Cardiovasc Res ; 80(2): 175-80, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18791204

RESUMO

AIMS: Neointimal formation after percutaneous coronary intervention (PCI), termed restenosis, limits therapeutic revascularization. Since it is now known that vascular injury involves an inflammatory response, we examined the role of tumour necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma) in the neointimal formation after injury. METHODS AND RESULTS: Control (BALB/c), TNF-alpha-deficient (Tnf(-/-)), IFN-gamma-deficient (Ifng(-/-)), or double-deficient (Tnf(-/-)Ifng(-/-)) mice were subjected to wire-mediated vascular injury of the right femoral artery. Neointimal formation after injury was significantly reduced after the injury in the Tnf(-/-)Ifng(-/-) mice, compared to that in the control, Tnf(-/-), and Ifng(-/-) mice. Immunohistochemical analysis showed that TNF-alpha and IFN-gamma were expressed in neointimal lesions in the control mice, but not in mice with deficiency of the corresponding cytokine. No significant difference in re-endothelialization was observed among these groups. The number of proliferating cell nuclear antigen in the neointimal lesions was significantly decreased in the Tnf(-/-)Ifng(-/-) mice. Bone marrow transplantation experiments revealed that deficiency of TNF-alpha and IFN-gamma specifically in bone marrow cells significantly inhibited neointimal formation after vascular injury. CONCLUSION: The absence of TNF-alpha and IFN-gamma in bone marrow cells synergistically inhibits neointimal formation following vascular injury, and thus, may provide new insights into the mechanisms underlying restenosis after PCI.


Assuntos
Arteriopatias Oclusivas/prevenção & controle , Células da Medula Óssea/imunologia , Transplante de Medula Óssea , Proliferação de Células , Artéria Femoral/patologia , Interferon gama/deficiência , Fator de Necrose Tumoral alfa/deficiência , Túnica Íntima/patologia , Animais , Arteriopatias Oclusivas/imunologia , Arteriopatias Oclusivas/patologia , Constrição Patológica , Modelos Animais de Doenças , Células Endoteliais/patologia , Artéria Femoral/imunologia , Artéria Femoral/lesões , Interferon gama/genética , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Miócitos de Músculo Liso/patologia , Fator de Necrose Tumoral alfa/genética , Túnica Íntima/imunologia , Túnica Íntima/lesões
14.
Stem Cells Int ; 2019: 4341286, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31814834

RESUMO

Mesenchymal stem cells (MSCs) in bone marrow and adipose tissues are expected to be effective tools for regenerative medicine to treat various diseases. To obtain MSCs that possess both high differentiation and tissue regenerative potential, it is necessary to establish an isolation system that does not require long-term culture. It has previously been reported that the cytoskeletal protein vimentin, expressed on the surfaces of multiple cell types, possesses N-acetylglucosamine- (GlcNAc-) binding activity. Therefore, we tried to exploit this interaction to efficiently isolate MSCs from rat bone marrow cells using GlcNAc-bearing polymer-coated dishes. Cells isolated by this method were identified as MSCs because they were CD34-, CD45-, and CD11b/c-negative and CD90-, CD29-, CD44-, CD54-, CD73-, and CD105-positive. Osteoblast, adipocyte, and chondrocyte differentiation was observed in these cells. In total, yields of rat MSCs were threefold to fourfold higher using GlcNAc-bearing polymer-coated dishes than yields using conventional tissue-culture dishes. Interestingly, MSCs isolated with GlcNAc-bearing polymer-coated dishes strongly expressed CD106, whereas those isolated with conventional tissue-culture dishes had low CD106 expression. Moreover, senescence-associated ß-galactosidase activity in MSCs from GlcNAc-bearing polymer-coated dishes was lower than that in MSCs from tissue-culture dishes. These results establish an improved isolation method for high-quality MSCs.

15.
Carcinogenesis ; 29(2): 448-54, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18174248

RESUMO

Bone marrow cells (BMCs) have been reported to behave as tissue-specific stem cells in some organs and to participate in tumorigenesis. However, the roles of BMCs in hepatic regeneration and carcinogenesis are still unknown. A choline-deficient, ethionine-supplemented (CDE) diet leads to the appearance of oval cells, a type of hepatic progenitor cell, and activates their replication. Furthermore, this type of diet induces preneoplastic nodules and hepatocellular carcinomas (HCCs) derived from oval cell progenitors. The aims of this study were to determine whether oval cells are derived from BMCs and whether preneoplastic nodules or HCCs originate from BMCs in the CDE diet rat model. To clarify the origin of constituent cells in the liver, we transplanted BMCs from green fluorescent protein (GFP) transgenic female rats into male Lewis rats, which were then exposed to a CDE diet to induce hepatocarcinogenesis. Some oval cells showed both donor-derived GFP expression and the recipient-specific Y chromosome, indicating that donor BMCs fused with recipient oval cells. Several preneoplastic nodules (precancerous lesions) identified by their glutathione S-transferase placental (GSTp) positivity were induced by CDE treatment. However, these preneoplastic GSTp-positive nodules were not GFP positive. In conclusion, this study has produced two major findings. First, BMCs fuse with some oval cells. Second, BMC-fused oval cells and BMCs might not have malignant potential in the CDE-treated rat model.


Assuntos
Células da Medula Óssea/metabolismo , Carcinoma Hepatocelular/metabolismo , Colina/farmacologia , Etionina/farmacologia , Regulação Neoplásica da Expressão Gênica , Hepatócitos/metabolismo , Neoplasias Hepáticas/metabolismo , Fígado/citologia , Ração Animal , Animais , Transformação Celular Neoplásica , Hepatócitos/citologia , Imuno-Histoquímica/métodos , Neoplasias Hepáticas Experimentais/metabolismo , Ratos , Ratos Endogâmicos Lew
16.
Circ Res ; 99(8): 891-9, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16990567

RESUMO

Myocardial infarction (MI) is accompanied by inflammatory responses that lead to the recruitment of leukocytes and subsequent myocardial damage, healing, and scar formation. Because monocyte chemoattractant protein-1 (MCP-1) (also known as CCL2) regulates monocytic inflammatory responses, we investigated the effect of cardiac MCP-1 overexpression on left ventricular (LV) dysfunction and remodeling in a murine MI model. Transgenic mice expressing the mouse JE-MCP-1 gene under the control of the alpha-cardiac myosin heavy chain promoter (MHC/MCP-1 mice) were used for this purpose. MHC/MCP-1 mice had reduced infarct area and scar formation and improved LV dysfunction after MI. These mice also showed induction of macrophage infiltration and neovascularization; however, few bone marrow-derived endothelial cells were detected in MHC/MCP-1 mice whose bone marrow was replaced with that of Tie2/LacZ transgenic mice. Flow cytometry analysis showed no increase in endothelial progenitor cells (CD34+/Flk-1+ cells) in MHC/MCP-1 mice. Marked myocardial interleukin (IL)-6 secretion, STAT3 activation, and LV hypertrophy were observed after MI in MHC/MCP-1 mice. Furthermore, cardiac myofibroblasts accumulated after MI in MHC/MCP-1 mice. In vitro experiments revealed that a combination of IL-6 with MCP-1 synergistically stimulated and sustained STAT3 activation in cardiomyocytes. MCP-1, IL-6, and hypoxia directly promoted the differentiation of cardiac fibroblasts into myofibroblasts. Our results suggest that cardiac overexpression of MCP-1 induced macrophage infiltration, neovascularization, myocardial IL-6 secretion, and accumulation of cardiac myofibroblasts, thereby resulting in the prevention of LV dysfunction and remodeling after MI. They also provide a new insight into the role of cardiac MCP-1 in the pathophysiology of MI.


Assuntos
Quimiocina CCL2/fisiologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Disfunção Ventricular Esquerda/prevenção & controle , Remodelação Ventricular , Animais , Células da Medula Óssea/citologia , Capilares/fisiopatologia , Cardiomegalia/etiologia , Diferenciação Celular , Quimiocina CCL2/metabolismo , Cicatriz/patologia , Vasos Coronários/fisiopatologia , Citocinas/fisiologia , Ecocardiografia , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Mediadores da Inflamação/fisiologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Infarto do Miocárdio/complicações , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/patologia , Neovascularização Fisiológica , Fator de Transcrição STAT3/metabolismo , Função Ventricular Esquerda
17.
Arterioscler Thromb Vasc Biol ; 27(2): 283-9, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17053169

RESUMO

OBJECTIVE: Since the macrophage colony-stimulating factor (M-CSF) has been shown to stimulate differentiation and proliferation of monocyte/macrophage lineage and to be involved in the process of neointimal formation after vascular injury, we tested the effects of M-CSF on the recruitment of bone marrow-derived progenitor cells in neointimal formation after vascular injury in mice. METHODS AND RESULTS: Wire-mediated vascular injury was produced in the femoral artery of C57BL/6 mice. Recombinant human M-CSF [500 microg/(kg x day)] or saline (control) was administered for 10 consecutive days, starting 4 days before the injury. Treatment with M-CSF accelerated neointimal formation in the early phase after injury, and this neointimal lesion mainly consisted of bone marrow-derived cells. M-CSF treatment had no effect on the mobilization of endothelial progenitor cells (EPCs: CD34+/Flk-1+) and reendothelialization after injury. The stromal cell-derived factor-1 (SDF-1) was markedly expressed in the neointima and media after injury, whereas CXCR4+ cells were observed in the neointima. Further, a novel CXCR4 antagonist, AMD3100, significantly attenuated the M-CSF-induced neointimal formation. CONCLUSIONS: These findings suggest that M-CSF accelerated neointimal formation after vascular injury via the SDF-1-CXCR4 system, and the inhibition of this system has therapeutic potential for the treatment of cardiovascular diseases.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Quimiocinas CXC/fisiologia , Endotélio Vascular/efeitos dos fármacos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Receptores CXCR4/fisiologia , Células-Tronco/efeitos dos fármacos , Animais , Benzilaminas , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Quimiocina CCL2/genética , Quimiocina CCL2/fisiologia , Quimiocina CXCL12 , Quimiocinas CXC/genética , Ciclamos , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Compostos Heterocíclicos/farmacologia , Interleucina-10/genética , Interleucina-10/fisiologia , Interleucina-6/genética , Interleucina-6/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/efeitos dos fármacos , Receptores CXCR4/genética , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/fisiologia , Túnica Íntima/citologia , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/fisiologia
18.
Cardiovasc Res ; 70(1): 61-9, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16448633

RESUMO

OBJECTIVE: Neointimal formation following percutaneous coronary intervention (PCI), termed restenosis, limits therapeutic revascularization. Since reendothelialization is one of the determinant factors for the development of neointimal formation, we examined the effects of granulocyte colony-stimulating factor (G-CSF) on reendothelialization and neointimal formation after vascular injury in mice. METHODS AND RESULTS: Wire-mediated vascular injury was produced in the femoral artery of C57BL/6 mice. G-CSF pretreatment significantly accelerated reendothelialization and decreased neointimal formation following vascular injury; however, this inhibitory effect of G-CSF was diminished when G-CSF was started following the injury. Flow cytometry analysis revealed that G-CSF treatment increased the number of endothelial progenitor cells (EPCs: CD34+/Flk-1+) in the peripheral circulation. Vascular injury was also produced in 2 types of mice whose bone marrow was replaced with that of enhanced green fluorescent protein- and Tie2/LacZ-transgenic mice. In the reendothelialized artery of these mice, few bone marrow-derived EPCs were detected. Furthermore, G-CSF treatment reduced the serum level of interleukin (IL)-6. CONCLUSION: G-CSF treatment accelerated reendothelialization and decreased neointimal formation following vascular injury, although there was little contribution of bone marrow-derived EPCs to the reendothelialization of the artery. These results suggest that G-CSF pretreatment has a therapeutic potential for prevention of restenosis following PCI.


Assuntos
Angioplastia Coronária com Balão/efeitos adversos , Endotélio Vascular/patologia , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Neovascularização Patológica/prevenção & controle , Animais , Transplante de Medula Óssea , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/imunologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Artéria Femoral/imunologia , Artéria Femoral/lesões , Artéria Femoral/patologia , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Humanos , Imuno-Histoquímica/métodos , Interleucina-6/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Transgênicos , Modelos Animais , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Proteínas Recombinantes , Células-Tronco/patologia , Túnica Íntima/patologia , beta-Galactosidase/genética
19.
Cardiovasc Res ; 69(2): 545-55, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16376325

RESUMO

OBJECTIVE: Sepsis accompanies myocardial dysfunction and dynamic alterations of cardiac metabolism. We have recently demonstrated that the very low-density lipoprotein receptor (VLDL-R), which is abundantly expressed in the heart, plays a key role in energy metabolism of the fasting heart. However, little is known about the function and regulation of the VLDL-R during sepsis. In the present study, we explored lipid accumulation and VLDL-R expression in the lipopolysaccharide (LPS)-stimulated heart in vivo and regulation of VLDL-R expression in vitro. METHODS AND RESULTS: Electron microscopy and immunohistochemistry demonstrated that LPS significantly decreased both lipid accumulation and VLDL-R expression in the hearts of fasting mice. Treatment with LPS also downregulated VLDL-R in rat neonatal cardiac myocytes, and this downregulation was completely reversed by interleukin (IL)-1beta receptor antagonist. IL-1beta downregulated the expression of VLDL-R in a time- and dose-dependent manner and markedly reduced the uptake of DiI-labeled beta-VLDL but not DiI-labeled low-density lipoprotein (LDL). Use of specific pharmacologic inhibitors and short interference RNA revealed that Hsp90 was required for IL-1beta to downregulate VLDL-R expression. CONCLUSIONS: These findings suggest that IL-1beta is a principle mediator of changes in cardiac lipid and energy metabolism during sepsis through the downregulation of myocardial VLDL-R expression.


Assuntos
Metabolismo dos Lipídeos , Miocárdio/metabolismo , Receptores de LDL/fisiologia , Sepse/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Regulação para Baixo , Jejum , Feminino , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Immunoblotting/métodos , Interleucina-1/metabolismo , Macrolídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão , Miócitos Cardíacos/metabolismo , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Curr Vasc Pharmacol ; 4(1): 59-65, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16472177

RESUMO

In the past decade, researchers have defined committed stem or progenitor cells from various tissues, including bone marrow, peripheral blood, brain, liver and reproductive organs, in both adult animals and humans. Recently, endothelial progenitor cells (EPCs) were isolated from peripheral blood mononuclear cells and were shown to be incorporated into foci of neovascularization. This finding that circulating EPCs may home into sites of neovascularization and differentiate into mature endothelial cells in situ is consistent with the concept of 'vasculogenesis' and suggests that vasculogenesis and angiogenesis might constitute complementary mechanisms for postnatal neovascularization. Furthermore, experimental and clinical studies on ischemic cardiovascular diseases suggest a therapeutic potential for EPC transplantation. In this review, we summarize the biological features of EPCs and discuss their therapeutic potential for the treatment of cardiovascular diseases.


Assuntos
Doenças Cardiovasculares/terapia , Neovascularização Fisiológica/fisiologia , Transplante de Células-Tronco , Células-Tronco/fisiologia , Adulto , Animais , Humanos , Estudos Multicêntricos como Assunto , Neovascularização Fisiológica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA