Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 35(35): 12241-7, 2015 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-26338334

RESUMO

UNLABELLED: The identification of the molecular network that supports oligodendrocyte (OL) regeneration under demyelinating conditions has been a primary goal for regenerative medicine in demyelinating disorders. We recently described an essential function for TACE/ADAM17 in regulating oligodendrogenesis during postnatal myelination, but it is unknown whether this protein also plays a role in OL regeneration and remyelination under demyelinating conditions. By using genetic mouse models to achieve selective gain- or loss-of-function of TACE or EGFR in OL lineage cells in vivo, we found that TACE is critical for EGFR activation in OLs following demyelination, and therefore, for sustaining OL regeneration and CNS remyelination. TACE deficiency in oligodendrocyte progenitor cells following demyelination disturbs OL lineage cell expansion and survival, leading to a delay in the remyelination process. EGFR overexpression in TACE deficient OLs in vivo restores OL development and postnatal CNS myelination, but also OL regeneration and CNS remyelination following demyelination. Our study reveals an essential function of TACE in supporting OL regeneration and CNS remyelination that may contribute to the design of new strategies for therapeutic intervention in demyelinating disorders by promoting oligodendrocyte regeneration and myelin repair. SIGNIFICANCE STATEMENT: Oligodendrocyte (OL) regeneration has emerged as a promising new approach for the treatment of demyelinating disorders. By using genetic mouse models to selectively delete TACE expression in oligodendrocyte progenitors cells (OPs), we found that TACE/ADAM17 is required for supporting OL regeneration following demyelination. TACE genetic depletion in OPs abrogates EGFR activation in OL lineage cells, and perturbs cell expansion and survival, blunting the process of CNS remyelination. Moreover, EGFR overexpression in TACE-deficient OPs in vivo overcomes the defects in OL development during postnatal development but also OL regeneration during CNS remyelination. Our study identifies TACE as an essential player in OL regeneration that may provide new insights in the development of new strategies for promoting myelin repair in demyelinating disorders.


Assuntos
Proteínas ADAM/metabolismo , Sistema Nervoso Central/patologia , Doenças Desmielinizantes/patologia , Regulação da Expressão Gênica/fisiologia , Esclerose Múltipla/patologia , Oligodendroglia/fisiologia , Regeneração/fisiologia , 2',3'-Nucleotídeo Cíclico 3'-Fosfodiesterase/genética , 2',3'-Nucleotídeo Cíclico 3'-Fosfodiesterase/metabolismo , Proteína ADAM17 , Animais , Antígenos/genética , Antígenos/metabolismo , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/genética , Doenças Desmielinizantes/induzido quimicamente , Modelos Animais de Doenças , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/ultraestrutura , Proteoglicanas/genética , Proteoglicanas/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Regeneração/efeitos dos fármacos
2.
Proc Natl Acad Sci U S A ; 110(40): 15919-24, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24046373

RESUMO

Most current cancer therapies focus on killing malignant cells, but these cells are often genetically unstable and can become resistant to chemotherapy. Tumor-associated macrophages (TAMs) facilitate disease progression by promoting angiogenesis and tumor cell growth, as well as by suppressing the adaptive immune response. TAMs are therefore potential targets for adjuvant anticancer therapies. However, resident macrophages are critical to host defense, and preferential ablation of TAMs remains challenging. Macrophage activation is broadly categorized as classically activated, or M1, and alternatively activated, or M2, and TAMs in the tumor microenvironment have been shown to adopt the anti-inflammatory, M2-like phenotype. To date, there are no methods for specific molecular targeting of TAMs. In this work, we report the discovery of a unique peptide sequence, M2pep, identified using a subtractive phage biopanning strategy against whole cells. The peptide preferentially binds to murine M2 cells, including TAMs, with low affinity for other leukocytes. Confocal imaging demonstrates the accumulation of M2pep in TAMs in vivo after tail vein injection. Finally, tail vein injection of an M2pep fusion peptide with a proapoptotic peptide delays mortality and selectively reduces the M2-like TAM population. This work therefore describes a molecularly targeted construct for murine TAMs and provides proof of concept of this approach as an anticancer treatment. In addition, M2pep is a useful tool for murine M2 macrophage identification and for modulating M2 macrophages in other murine models of disease involving M2 cells.


Assuntos
Apoptose/imunologia , Sistemas de Liberação de Medicamentos/métodos , Imunidade Inata/imunologia , Macrófagos/metabolismo , Neoplasias/imunologia , Peptídeos/metabolismo , Animais , Citometria de Fluxo , Macrófagos/imunologia , Camundongos , Microscopia Confocal , Biblioteca de Peptídeos , Peptídeos/imunologia , Análise de Sobrevida
3.
J Neurosci ; 34(36): 11884-96, 2014 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-25186737

RESUMO

Several studies have elucidated the significance of a disintegrin and metalloproteinase proteins (ADAMs) in PNS myelination, but there is no evidence if they also play a role in oligodendrogenesis and CNS myelination. Our study identifies ADAM17, also called tumor necrosis factor-α converting enzyme (TACE), as a novel key modulator of oligodendrocyte (OL) development and CNS myelination. Genetic deletion of TACE in oligodendrocyte progenitor cells (OPs) induces premature cell cycle exit and reduces OL cell survival during postnatal myelination of the subcortical white matter (SCWM). These cellular and molecular changes lead to deficits in SCWM myelination and motor behavior. Mechanistically, TACE regulates oligodendrogenesis by modulating the shedding of EGFR ligands TGFα and HB-EGF and, consequently, EGFR signaling activation in OL lineage cells. Constitutive TACE depletion in OPs in vivo leads to similar alterations in CNS myelination and motor behavior as to what is observed in the EGFR hypofunctional mouse line EgfrWa2. EGFR overexpression in TACE-deficient OPs restores OL survival and development. Our study reveals an essential function of TACE in oligodendrogenesis, and demonstrates how this molecule modulates EGFR signaling activation to regulate postnatal CNS myelination.


Assuntos
Proteínas ADAM/metabolismo , Encéfalo/metabolismo , Bainha de Mielina/metabolismo , Neurogênese , Oligodendroglia/metabolismo , Proteínas ADAM/genética , Proteína ADAM17 , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiologia , Linhagem Celular , Linhagem da Célula , Células Cultivadas , Receptores ErbB/genética , Receptores ErbB/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Locomoção , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Oligodendroglia/citologia , Fator de Crescimento Transformador alfa/metabolismo
4.
Circ Res ; 113(1): 52-61, 2013 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-23584255

RESUMO

RATIONALE: Apoptotic cell phagocytosis (efferocytosis) is mediated by specific receptors and is essential for resolution of inflammation. In chronic inflammation, apoptotic cell clearance is dysfunctional and soluble levels of several apoptotic cell receptors are elevated. Reports have identified proteolytic cleavage as a mechanism capable of releasing soluble apoptotic cell receptors, but the functional implications of their proteolysis are unclear. OBJECTIVE: To test the hypothesis that ADAM17-mediated cleavage of apoptotic cell receptors limits efferocytosis in vivo. METHODS AND RESULTS: In vivo comparison of macrophage efferocytosis in wild-type and Adam17-null hematopoietic chimeras demonstrates that ADAM17 deficiency leads to a 60% increase in efferocytosis and an enhanced anti-inflammatory phenotype in a model of peritonitis. In vitro uptake of phosphatidylserine liposomes identifies the dual-pass apoptotic cell receptor CD36 as a major contributor to enhanced efferocytosis, and CD36 surface levels are elevated on macrophages from Adam17-null mice. Further, temporal elevation of CD36 expression with inflammation may also contribute to its impact. Soluble CD36 from macrophage-conditioned media comprises 2 species based on Western blotting, and mass spectrometry identifies 3 N-terminal peptides that represent probable cleavage sites. Levels of soluble CD36 are decreased in Adam17-null conditioned media, providing evidence for involvement of ADAM17 in CD36 cleavage. Importantly, enhanced efferocytosis in vivo by macrophages lacking ADAM17 is CD36 dependent and accelerates macrophage clearance from the peritoneum, thus promoting resolution of inflammation and highlighting the impact of increased apoptotic cell uptake. CONCLUSIONS: Our studies demonstrate the importance of ADAM17-mediated proteolysis for in vivo efferocytosis regulation and suggest a possible mechanistic link between chronic inflammation and defective efferocytosis.


Assuntos
Proteínas ADAM/fisiologia , Apoptose/fisiologia , Antígenos CD36/fisiologia , Macrófagos Peritoneais/enzimologia , Peritonite/enzimologia , Fagocitose/fisiologia , Proteínas ADAM/deficiência , Proteínas ADAM/genética , Proteína ADAM17 , Sequência de Aminoácidos , Animais , Transplante de Medula Óssea , Antígenos CD36/química , Quimera , Meios de Cultivo Condicionados/farmacologia , Dexametasona/farmacologia , Células-Tronco Embrionárias/transplante , Lipossomos , Macrófagos Peritoneais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Peritonite/induzido quimicamente , Peritonite/patologia , Fenótipo , Tioglicolatos/toxicidade , Timócitos/patologia , Timócitos/transplante
5.
J Immunol ; 190(8): 4236-44, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23479224

RESUMO

Despite expanded definition of the leukocyte adhesion cascade and mechanisms underlying individual steps, very little is known about regulatory mechanisms controlling sequential shifts between steps. We tested the hypothesis that metalloproteinases provide a mechanism to rapidly transition monocytes between different steps. Our study identifies diapedesis as a step targeted by metalloproteinase activity. Time-lapse video microscopy shows that the presence of a metalloproteinase inhibitor results in a doubling of the time required for human monocytes to complete diapedesis on unactivated or inflamed human endothelium, under both static and physiological-flow conditions. Thus, diapedesis is promoted by metalloproteinase activity. In contrast, neither adhesion of monocytes nor their locomotion over the endothelium is altered by metalloproteinase inhibition. We further demonstrate that metalloproteinase inhibition significantly elevates monocyte cell surface levels of integrins CD11b/CD18 (Mac-1), specifically during transendothelial migration. Interestingly, such alterations are not detected for other endothelial- and monocyte-adhesion molecules that are presumed metalloproteinase substrates. Two major transmembrane metalloproteinases, a disintegrin and metalloproteinase (ADAM)17 and ADAM10, are identified as enzymes that control constitutive cleavage of Mac-1. We further establish that knockdown of monocyte ADAM17, but not endothelial ADAM10 or ADAM17 or monocyte ADAM10, reproduces the diapedesis delay observed with metalloproteinase inhibition. Therefore, we conclude that monocyte ADAM17 facilitates the completion of transendothelial migration by accelerating the rate of diapedesis. We propose that the progression of diapedesis may be regulated by spatial and temporal cleavage of Mac-1, which is triggered upon interaction with endothelium.


Assuntos
Proteínas ADAM/fisiologia , Metaloproteases/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Migração Transendotelial e Transepitelial/imunologia , Proteínas ADAM/deficiência , Proteínas ADAM/metabolismo , Proteína ADAM17 , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana , Humanos , Antígeno de Macrófago 1/metabolismo , Metaloproteases/antagonistas & inibidores , Monócitos/enzimologia , Especificidade por Substrato/imunologia , Imagem com Lapso de Tempo/métodos
6.
J Biol Chem ; 288(42): 30742-30751, 2013 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-24043629

RESUMO

p40, a Lactobacillus rhamnosus GG (LGG)-derived soluble protein, ameliorates intestinal injury and colitis, reduces apoptosis, and preserves barrier function by transactivation of the EGF receptor (EGFR) in intestinal epithelial cells. The aim of this study is to determine the mechanisms by which p40 transactivates the EGFR in intestinal epithelial cells. Here we show that p40-conditioned medium activates EGFR in young adult mouse colon epithelial cells and human colonic epithelial cell line, T84 cells. p40 up-regulates a disintegrin and metalloproteinase domain-containing protein 17 (ADAM17) catalytic activity, and broad spectrum metalloproteinase inhibitors block EGFR transactivation by p40 in these two cell lines. In ADAM17-deficient mouse colonic epithelial (ADAM17(-/-) MCE) cells, p40 transactivation of EGFR is blocked, but can be rescued by re-expression with WT ADAM17. Furthermore, p40 stimulates release of heparin binding (HB)-EGF, but not transforming growth factor (TGF)α or amphiregulin, in young adult mouse colon cells and ADAM17(-/-) MCE cells overexpressing WT ADAM17. Knockdown of HB-EGF expression by siRNA suppresses p40 effects on transactivating EGFR and Akt, preventing apoptosis, and preserving tight junction function. The effects of p40 on HB-EGF release and ADAM17 activation in vivo are examined after administration of p40-containing pectin/zein hydrogel beads to mice. p40 stimulates ADAM17 activity and EGFR activation in colonic epithelial cells and increases HB-EGF levels in blood from WT mice, but not from mice with intestinal epithelial cell-specific ADAM17 deletion. Thus, these data define a mechanism of a probiotic-derived soluble protein in modulating intestinal epithelial cell homeostasis through ADAM17-mediated HB-EGF release, leading to transactivation of EGFR.


Assuntos
Proteínas de Bactérias/metabolismo , Células Epiteliais/metabolismo , Receptores ErbB/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mucosa Intestinal/metabolismo , Lacticaseibacillus rhamnosus/metabolismo , Probióticos/metabolismo , Ativação Transcricional , Proteínas ADAM/biossíntese , Proteínas ADAM/genética , Proteína ADAM17 , Animais , Linhagem Celular Tumoral , Ativação Enzimática/genética , Células Epiteliais/citologia , Células Epiteliais/microbiologia , Receptores ErbB/genética , Regulação Enzimológica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Mucosa Intestinal/citologia , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Crescimento Transformador alfa/genética , Fator de Crescimento Transformador alfa/metabolismo , Regulação para Cima/genética
7.
J Biol Chem ; 287(7): 4581-9, 2012 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-22170060

RESUMO

Macrophage exiting from inflammatory sites is critical to limit the local innate immune response. With tissue insult, resident tissue macrophages rapidly efflux to lymph nodes where they modulate the adaptive immune response, and inflammatory macrophages attracted to the site of injury then exit during the resolution phase. However, the mechanisms that regulate macrophage efflux are poorly understood. This study has investigated soluble forms of integrin ß2 whose levels are elevated in experimental peritonitis at times when macrophages are exiting the peritoneum, suggesting that its proteolytic shedding may be involved in macrophage efflux. Both constitutive and inducible metalloproteinase-dependent shedding of integrin ß2 from mouse macrophages are demonstrated. Soluble integrin ß2 is primarily released as a heterodimeric complex with αM that retains its ability to bind its ligands intracellular adhesion molecule-1, fibrin, and collagen and thus may serve as a soluble antagonist. In a model of accelerated exiting, administration of a metalloproteinase inhibitor prevents macrophage efflux by 50% and impedes loss of macrophage integrin ß2 from the cell surface. Exiting of peritoneal macrophages in mice lacking integrin ß2 is accelerated, and antibody disruption of integrin ß2-substrate interactions can reverse 50% of the metalloprotease inhibitor blockade of macrophage exiting. Thus, our study demonstrates the ability of metalloproteinase-mediated shedding of integrin ß2 to promote macrophage efflux from inflammatory sites, and the release of soluble integrin heterodimers may also limit local inflammation.


Assuntos
Antígenos CD18/metabolismo , Movimento Celular , Macrófagos Peritoneais/metabolismo , Metaloproteases/metabolismo , Peritonite/metabolismo , Multimerização Proteica , Animais , Antígenos CD18/genética , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Fibrina/genética , Fibrina/metabolismo , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Macrófagos Peritoneais/patologia , Metaloproteases/genética , Camundongos , Camundongos Mutantes , Peritonite/genética , Peritonite/patologia , alfa-Macroglobulinas/genética , alfa-Macroglobulinas/metabolismo
8.
J Cell Biochem ; 114(5): 1194-202, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23192608

RESUMO

Osteopontin (OPN) is highly expressed by macrophages and plays a key role in the pathology of several chronic inflammatory diseases including atherosclerosis and the foreign body reaction. However, the molecular mechanism behind OPN regulation of macrophage functions is not well understood. OPN is a secreted molecule and interacts with several integrins via two domains: the RGD sequence binding to α(v) -containing integrins, and the SLAYGLR sequence binding to α(4) ß(1), α(4) ß(7), and α(9) ß(1) integrins. Here we determined the role of OPN in macrophage survival, chemotaxis, and activation state. For survival studies, OPN treated-bone marrow derived macrophages (BMDMs) were challenged with growth factor withdrawal and neutralizing integrin antibodies. We found that survival in BMDMs is mediated primarily through the α(4) integrin. In chemotaxis studies, we observed that migration to OPN was blocked by neutralizing α(4) and α(9) integrin antibodies. Further, OPN did not affect macrophage activation as measured by IL-12 production. Finally, the relative contributions of the RGD and the SLAYGLR functional domains of OPN to leukocyte recruitment were evaluated in an in vivo model. We generated chimeric mice expressing mutated forms of OPN in myeloid-derived leukocytes, and found that the SLAYGLR functional domain of OPN, but not the RGD, mediates macrophage accumulation in response to thioglycollate-elicited peritonitis. Collectively, these data indicate that α(4) and α(9) integrins interacting with OPN via the SLAYGLR domain play a key role in macrophage biology by regulating migration, survival, and accumulation.


Assuntos
Quimiotaxia , Cadeias alfa de Integrinas/metabolismo , Integrina alfa4/metabolismo , Macrófagos/citologia , Osteopontina/metabolismo , Sequência de Aminoácidos , Animais , Células da Medula Óssea/citologia , Sobrevivência Celular , Citocinas/biossíntese , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Osteopontina/química , Peritonite/metabolismo , Peritonite/patologia , Fenótipo , Estrutura Terciária de Proteína , Transdução de Sinais , Relação Estrutura-Atividade , Tioglicolatos
9.
Blood ; 118(3): 786-94, 2011 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-21628404

RESUMO

TNF-α-converting enzyme (TACE, herein denoted as Adam17) proteolytically sheds several cell-surface inflammatory proteins, but the physiologic importance of the cleavage of these substrates from leukocyte subsets during inflammation is incompletely understood. In this study, we show that Adam17-null neutrophils have a 2-fold advantage in their initial recruitment during thioglycollate-induced peritonitis, and they roll slower and adhere more readily in the cremaster model than wild-type neutrophils. Although CD44 and ICAM-1 are both in vitro substrates of Adam17, their surface levels are not altered on Adam17-null neutrophils. In contrast, L-selectin levels are elevated up to 10-fold in Adam17-null circulating neutrophils, and their accelerated peritoneal influx, slower rolling, and increased adhesion in the cremaster muscle are dependent on L-selectin. Analysis of mixed chimeras shows that enhanced L-selectin levels and accelerated influx were both cell-intrinsic properties of neutrophils lacking Adam17. In contrast, Adam17-null monocytes display no acceleration of infiltration into the peritoneum in spite of elevated L-selectin surface levels, and their peritoneal influx was independent of L-selectin. Therefore, our data demonstrate substrate and myeloid cell-type specificity of Adam17-mediated cleavage of its substrates, and show that neutrophils and monocytes use distinct mechanisms for infiltration of tissues.


Assuntos
Proteínas ADAM/imunologia , Proteínas ADAM/metabolismo , Movimento Celular/imunologia , Inflamação/imunologia , Monócitos/imunologia , Neutrófilos/imunologia , Proteínas ADAM/genética , Proteína ADAM17 , Animais , Adesão Celular/imunologia , Quimera , Modelos Animais de Doenças , Fragmentos Fab das Imunoglobulinas/farmacologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Selectina L/imunologia , Selectina L/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Monócitos/citologia , Neutrófilos/citologia , Peritonite/induzido quimicamente , Peritonite/imunologia , Peritonite/metabolismo , Especificidade por Substrato , Tioglicolatos/farmacologia
10.
Am J Pathol ; 178(6): 2931-7, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21550016

RESUMO

The Fas death receptor (CD95) is expressed on macrophages, smooth muscle cells, and T cells within atherosclerotic lesions. Given the dual roles of Fas in both apoptotic and nonapoptotic signaling, the aim of the present study was to test the effect of hematopoietic Fas deficiency on experimental atherosclerosis in low-density lipoprotein receptor-null mice (Ldlr(-/-)). Bone marrow from Fas(-/-) mice was used to reconstitute irradiated Ldlr(-/-) mice as a model for atherosclerosis. After 16 weeks on an 0.5% cholesterol diet, no differences were noted in brachiocephalic artery lesion size, cellularity, or vessel wall apoptosis. However, Ldlr(-/-) mice reconstituted with Fas(-/-) hematopoietic cells had elevated hyperlipidemia [80% increase, relative to wild-type (WT) controls; P < 0.001] and showed marked elevation of plasma levels of CXCL1/KC, CCL2/MCP-1, IL-6, IL-10, IL-12 subunit p70, and soluble Fas ligand (P < 0.01), as well as systemic microvascular inflammation. It was not possible to assess later stages of atherosclerosis because of increased mortality in Fas(-/-) bone marrow recipients. Our data indicate that hematopoietic Fas deficiency does not affect early atherosclerotic lesion development in Ldlr(-/-) mice.


Assuntos
Aterosclerose/patologia , Sistema Hematopoético/metabolismo , Sistema Hematopoético/patologia , Receptor fas/deficiência , Animais , Apoptose , Aterosclerose/complicações , Biomarcadores/metabolismo , Proliferação de Células , Quimiocinas/metabolismo , Quimera , Modelos Animais de Doenças , Hipercolesterolemia/complicações , Hipercolesterolemia/patologia , Inflamação/complicações , Inflamação/patologia , Camundongos , Microvasos/patologia , Receptores de LDL/deficiência , Receptores de LDL/metabolismo , Receptor fas/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 31(11): 2397-401, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22011752

RESUMO

A growth-promoting activity released from activated platelets, the platelet-derived growth factor, was discovered and characterized while the cellular and molecular mechanisms underlying the formation of the lesions of atherosclerosis were being investigated. This review provides a personal account of the different challenges we faced 3 decades ago in this undertaking and describes how our path was influenced by our focus on a disease process and by the evolving general understanding of the molecular effectors of cell proliferation.


Assuntos
Fator de Crescimento Derivado de Plaquetas/história , Fator de Crescimento Derivado de Plaquetas/fisiologia , Doenças Vasculares/fisiopatologia , Proliferação de Células , História do Século XX , Humanos , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Proteínas Tirosina Quinases/fisiologia , Receptores do Fator de Crescimento Derivado de Plaquetas/fisiologia , Doenças Vasculares/patologia
13.
Arterioscler Thromb Vasc Biol ; 31(11): 2473-82, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21868708

RESUMO

OBJECTIVE: Glutamate-cysteine ligase (GCL) is the rate-limiting step in glutathione synthesis. The enzyme is a heterodimer composed of a catalytic subunit, GCLC, and a modifier subunit, GCLM. We generated apolipoprotein E (apoE)-/- mice deficient in GCLM (apoE-/-/Gclm-/-) and transgenic mice that overexpress GCLC specifically in macrophages (apoE-/-/Gclc-Tg) to test the hypothesis that significantly altering the availability of glutathione has a measurable impact on both the initiation and progression of atherosclerosis. METHODS AND RESULTS: Atherosclerotic plaque size and composition were measured in the innominate artery in chow-fed male and female mice at 20, 30, 40, and 50 weeks of age and in the aortic sinus at 40 and 50 weeks of age. The apoE-/-/Gclm-/- mice more rapidly developed complex lesions, whereas the apoE-/-/Gclc-Tg mice had reduced lesion development compared with the littermate apoE-/- control mice. Transplantation of bone marrow from the apoE-/-/Gclm-/- and apoE-/-/Gclc-Tg mice into apoE-/- mice with established lesions also stimulated or inhibited further lesion development at 30 weeks posttransplant. CONCLUSION: Gain and loss of function in the capacity to synthesize glutathione especially in macrophages has reciprocal effects on the initiation and progression of atherosclerosis at multiple sites in apoE-/- mice.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Aterosclerose/patologia , Glutationa/metabolismo , Animais , Apolipoproteínas E/genética , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Glutamato-Cisteína Ligase/deficiência , Glutamato-Cisteína Ligase/genética , Lipídeos/sangue , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Seio Aórtico/metabolismo , Seio Aórtico/patologia
14.
Mol Cell Proteomics ; 8(5): 1044-60, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19116209

RESUMO

Activated macrophages are essential effectors of immunity and a rich source of matrix metalloproteinase-9 (MMP-9; gelatinase B). To search for cellular substrates of the enzyme, we subjected wild-type macrophages and macrophages expressing an autoactivating form of pro-MMP-9 (M9A macrophages) to proteomics analysis. Two-dimensional liquid chromatography together with tandem mass spectrometry identified 467 proteins in medium conditioned by M9A and/or wild-type macrophages. Subtractive proteomics identified 18 candidate MMP-9 substrates. Biochemical studies confirmed that two transmembrane proteins, beta(2) integrin subunit (CD18) and amyloid protein precursor (APP), were enriched in the medium of M9A macrophages. To identify potential cleavage sites, we synthesized an overlapping library of peptides that spanned 60 residues of the ectodomain and transmembrane domain of beta(2) integrin. Active MMP-9 cleaved a single peptide, ECVKGPNVAAIVGGT, at residues corresponding to Ala(705) and Ile(706) of the beta(2) integrin. Peptides corresponding to this cleavage site were detected by tandem mass spectrometric analysis only in medium from M9A macrophages, strongly supporting the proposal that beta(2) integrin is shed by autoactivating MMP-9. Our observations indicate that subtractive proteomics in concert with peptide substrate mapping is a powerful approach for identifying proteolytic substrates and suggest that MMP-9 plays previously unsuspected roles in the regulation and shedding of beta(2) integrin.


Assuntos
Antígenos CD18/metabolismo , Macrófagos/enzimologia , Metaloproteinase 9 da Matriz/metabolismo , Subunidades Proteicas/metabolismo , Sequência de Aminoácidos , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Antígenos CD18/química , Extratos Celulares , Linhagem Celular , Cromatografia Líquida , Meios de Cultivo Condicionados , Ativação Enzimática , Macrófagos/metabolismo , Espectrometria de Massas , Proteínas de Membrana/análise , Camundongos , Dados de Sequência Molecular , Peptídeos/química , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Proteômica , Reprodutibilidade dos Testes , Especificidade por Substrato
16.
J Clin Invest ; 116(1): 59-69, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16374516

RESUMO

The majority of acute clinical manifestations of atherosclerosis are due to the physical rupture of advanced atherosclerotic plaques. It has been hypothesized that macrophages play a key role in inducing plaque rupture by secreting proteases that destroy the extracellular matrix that provides physical strength to the fibrous cap. Despite reports detailing the expression of multiple proteases by macrophages in rupture-prone regions, there is no direct proof that macrophage-mediated matrix degradation can induce plaque rupture. We aimed to test this hypothesis by retrovirally overexpressing the candidate enzyme MMP-9 in macrophages of advanced atherosclerotic lesions of apoE-/- mice. Despite a greater than 10-fold increase in the expression of MMP-9 by macrophages, there was only a minor increase in the incidence of plaque fissuring. Subsequent analysis revealed that macrophages secrete MMP-9 predominantly as a proform, and this form is unable to degrade the matrix component elastin. Expression of an autoactivating form of MMP-9 in macrophages in vitro greatly enhances elastin degradation and induces significant plaque disruption when overexpressed by macrophages in advanced atherosclerotic lesions of apoE-/- mice in vivo. These data show that enhanced macrophage proteolytic activity can induce acute plaque disruption and highlight MMP-9 as a potential therapeutic target for stabilizing rupture-prone plaques.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/genética , Macrófagos/enzimologia , Metaloproteinase 9 da Matriz/genética , Animais , Apolipoproteínas E/genética , Aterosclerose/enzimologia , Aterosclerose/patologia , Transplante de Medula Óssea , Colagenases/metabolismo , Cruzamentos Genéticos , Feminino , Metaloproteinase 12 da Matriz , Metaloproteinase 13 da Matriz , Metaloproteinase 9 da Matriz/metabolismo , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
17.
Circ Res ; 100(11): 1589-96, 2007 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-17478729

RESUMO

Vascular dysfunction is a major complication of metabolic disorders such as diabetes and obesity. The current studies were undertaken to determine whether inflammatory responses are activated in the vasculature of mice with diet-induced obesity, and if so, whether Toll-Like Receptor-4 (TLR4), a key mediator of innate immunity, contributes to these responses. Mice lacking TLR4 (TLR4(-/-)) and wild-type (WT) controls were fed either a low fat (LF) control diet or a diet high in saturated fat (HF) for 8 weeks. In response to HF feeding, both genotypes displayed similar increases of body weight, body fat content, and serum insulin and free fatty acid (FFA) levels compared with mice on a LF diet. In lysates of thoracic aorta from WT mice maintained on a HF diet, markers of vascular inflammation both upstream (IKKbeta activity) and downstream of the transcriptional regulator, NF-kappaB (ICAM protein and IL-6 mRNA expression), were increased and this effect was associated with cellular insulin resistance and impaired insulin stimulation of eNOS. In contrast, vascular inflammation and impaired insulin responsiveness were not evident in aortic samples taken from TLR4(-/-) mice fed the same HF diet, despite comparable increases of body fat mass. Incubation of either aortic explants from WT mice or cultured human microvascular endothelial cells with the saturated FFA, palmitate (100 micromol/L), similarly activated IKKbeta, inhibited insulin signal transduction and blocked insulin-stimulated NO production. Each of these effects was subsequently shown to be dependent on both TLR4 and NF-kappaB activation. These findings identify the TLR4 signaling pathway as a key mediator of the deleterious effects of palmitate on endothelial NO signaling, and are the first to document a key role for TLR4 in the mechanism whereby diet-induced obesity induces vascular inflammation and insulin resistance.


Assuntos
Mediadores da Inflamação/imunologia , Resistência à Insulina/imunologia , Obesidade/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Composição Corporal/genética , Peso Corporal/genética , Células Cultivadas , Gorduras na Dieta , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Ácidos Graxos não Esterificados/sangue , Ácidos Graxos não Esterificados/farmacologia , Humanos , Proteínas I-kappa B/metabolismo , Mediadores da Inflamação/metabolismo , Insulina/sangue , Insulina/farmacologia , Resistência à Insulina/genética , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Inibidor de NF-kappaB alfa , Obesidade/genética , Obesidade/patologia , Técnicas de Cultura de Órgãos , Ácido Palmítico/farmacologia , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/genética
18.
Arterioscler Thromb Vasc Biol ; 27(5): 1043-9, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17303772

RESUMO

OBJECTIVE: Activation of collagen receptors expressed by smooth muscle cells induces matrix metalloproteinase (MMP) expression. The 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) have been shown to interfere with integrin signaling, but their effects on collagen receptor-mediated MMP expression have not been investigated. METHODS AND RESULTS: In the present study, we show that simvastatin (3 micromol/L) reduces MMP1 expression and secretion in human smooth muscle cells cultured on polymerized type I collagen by 39.9+/-11.2% and 36.0+/-2.3%, respectively. Reduced MMP1 protein levels correlate with a similar decrease in MMP1 promoter activity (-33.0+/-8.9%), MMP1 mRNA levels (-37.8+/-10.5%), and attenuation of smooth muscle cell collagen degradation (-34.2+/-6.1%). Mevalonate, and the isoprenoid derivative geranylgeraniol, precursors of geranylgeranylated proteins, completely prevent the inhibitory effect of simvastatin on MMP1. Moreover, the protein geranylgeranyltransferase inhibitor GGTI-286 significantly decreases MMP1 expression. Retroviral overexpression of dominant-negative mutants of geranylgeranylated Rac1 lead to a reduction of MMP1 protein (-50.4+/-5.4%) and mRNA levels (-97.9+/-1.0%), and knockdown of Rac1 by small interfering RNA downregulates MMP1 expression. Finally, simvastatin reduces GTP-bound Rac1 expression levels in smooth muscle cells cultured on polymerized collagen. CONCLUSIONS: These results demonstrate that simvastatin, by inhibiting Rac1 activity, reduces MMP1 expression and collagen degradation in human smooth muscle cells.


Assuntos
Colágeno/farmacologia , Expressão Gênica/efeitos dos fármacos , Metaloproteinase 1 da Matriz/genética , Músculo Liso Vascular/enzimologia , RNA Mensageiro/genética , Sinvastatina/farmacologia , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Células Cultivadas , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Recém-Nascido , Leucina/análogos & derivados , Leucina/farmacologia , Metaloproteinase 1 da Matriz/biossíntese , Inibidores de Metaloproteinases de Matriz , Ácido Mevalônico/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Reação em Cadeia da Polimerase , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/metabolismo
19.
Mol Cell Biol ; 38(17)2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29891514

RESUMO

Macrophages are prominent cells in acute and chronic inflammatory diseases. Recent studies highlight a role for macrophage proliferation post-monocyte recruitment under inflammatory conditions. Using an acute peritonitis model, we identify a significant defect in macrophage proliferation in mice lacking the leukocyte transmembrane protease ADAM17. The defect is associated with decreased levels of macrophage colony-stimulating factor 1 (CSF-1) in the peritoneum and is rescued by intraperitoneal injection of CSF-1. Cell surface CSF-1 (csCSF-1) is one of the substrates of ADAM17. We demonstrate that both infiltrated neutrophils and macrophages are major sources of csCSF-1. Furthermore, acute shedding of csCSF-1 following neutrophil extravasation is associated with elevated expression of iRhom2, a member of the rhomboid-like superfamily, which promotes ADAM17 maturation and trafficking to the neutrophil surface. Accordingly, deletion of hematopoietic iRhom2 is sufficient to prevent csCSF-1 release from neutrophils and macrophages and to prevent macrophage proliferation. In acute inflammation, csCSF-1 release and macrophage proliferation are self-limiting due to transient leukocyte recruitment and temporally restricted csCSF-1 expression. In chronic inflammation, such as atherosclerosis, the ADAM17-mediated lesional macrophage proliferative response is prolonged. Our results demonstrate a novel mechanism whereby ADAM17 promotes macrophage proliferation in states of acute and chronic inflammation.


Assuntos
Proteína ADAM17/metabolismo , Inflamação/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/metabolismo , Neutrófilos/metabolismo , Proteína ADAM17/deficiência , Proteína ADAM17/genética , Doença Aguda , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Proliferação de Células , Doença Crônica , Inflamação/patologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Neutrófilos/patologia , Peritonite/metabolismo , Peritonite/patologia , Receptores de LDL/deficiência , Receptores de LDL/genética , Solubilidade
20.
Endocrinology ; 148(11): 5230-7, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17673516

RESUMO

Systemic inflammatory stimuli cause anorexia and weight loss by disrupting the physiological regulation of energy balance. Mice lacking MyD88, an intracellular mediator of signal transduction activated by Toll-like receptor 4 or IL-1beta receptors, are resistant to anorexia induced by the bacterial endotoxin lipopolysaccharide (LPS), despite a significant circulating cytokine response. Thus, we hypothesized that induction of a peripheral inflammatory response is insufficient to cause LPS-induced anorexia when MyD88 signaling in the central nervous system and other tissues is absent. To test this hypothesis, we used bone marrow transplantation (BMT) to determine if LPS-induced anorexia can be restored to MyD88-deficient mice by reconstituting their bone marrow with wild-type (WT) immune cells. We found that restoring WT circulating immune cells to mice lacking MyD88 conferred only a mild, short-lived anorexia in response to LPS, such that food intake was fully normalized by 20 h post injection (LPS 4.1 +/- 0.5 g vs. vehicle 4.3 +/- 0.3 g), whereas LPS-induced anorexia was profound and sustained in WT controls after either autologous BMT or sham BMT. Similarly, LPS-mediated induction of hypothalamic mRNA encoding IL-1beta and TNFalpha was robust in both WT control groups but was absent in chimeric MyD88 mice, despite comparable peripheral inflammatory responses across the three groups. We conclude that LPS reduces food intake via a mechanism dependent on MyD88 signaling within brain and/or other tissues and that in the absence of this effect, robust stimulation of circulating immune cells cannot induce sustained anorexia.


Assuntos
Anorexia/induzido quimicamente , Hipotálamo/fisiologia , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Transdução de Sinais/fisiologia , Baço/fisiologia , Animais , Composição Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Transplante de Medula Óssea/fisiologia , Células Cultivadas , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Hipotálamo/metabolismo , Sistema Imunitário/metabolismo , Sistema Imunitário/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Baço/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA