Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Sports Med ; 40(1): 43-51, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30481830

RESUMO

To investigate the effect of exercise on adiponectin in young healthy human males, we examined serum total adiponectin and high-molecular-weight (HMW) adiponectin in newly recruited male soldiers who participated in an 8-week basic military training (BMT). A total of 95 males (mean age, 18.79±1.50 years) were sampled from among 1,100 new male army recruits in China. Participants were separated into 3 groups according to their body mass index (BMI): overweight group (BMI: 24.9 kg/m2 to<30 kg/m2; n=26); normal-weight group (BMI: 18.5 kg/m2 to<24.9 kg/m2; n=40); and underweight group (BMI:<18.5 kg/m2; n=29). Anthropometric measurements, fasting serum total adiponectin, HMW adiponectin, and lipid profiles were recorded at baseline and at the end of the 8-week BMT. After the 8-week BMT, the HMW/total adiponectin ratio (HMW/total ratio) and HDL cholesterol improved significantly (p<0.001 and p<0.001, respectively). HMW/total ratio showed significant correlations with HDL cholesterol. Our study suggests that an 8-week BMT can improve the HMW/total ratio in healthy young males regardless of their BMI and anthropometry. Both HMW/total ratio and HDL cholesterol can serve as potential biomarkers for assessing the efficacy of exercise and may have metabolic benefits for preventing obesity and obesity-related disease.


Assuntos
Adiponectina/sangue , Militares , Condicionamento Físico Humano , Adolescente , Antropometria , Biomarcadores/sangue , Índice de Massa Corporal , China , HDL-Colesterol/sangue , Humanos , Lipídeos/sangue , Masculino , Sobrepeso/sangue , Adulto Jovem
2.
FEBS Open Bio ; 5: 182-90, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25834784

RESUMO

CXCR1, a receptor for interleukin-8 (IL-8), plays an important role in defending against pathogen invasion during neutrophil-mediated innate immune response. Human CXCR1 is a G protein-coupled receptor (GPCR) with its characteristic seven transmembrane domains (TMs). Functional and structural analyses of several GPCRs have revealed that conserved residues on TM3 (including the highly conserved Asp-Arg-Tyr (DRY) motif) and TM6 near intracellular loops contain domains critical for G protein coupling as well as GPCR activation. The objective of this study was to elucidate the role of critical amino acid residues on TM3 near intracellular loop 2 (i2) and TM6 near intracellular loop 3 (i3), including S132(3.47) (Baldwin location), D134(3.49), M241(6.34), and F251(6.44), in G protein coupling and CXCR1 activation. The results demonstrate that mutations of D134(3.49) at DRY motif of CXCR1 (D134N and D134V) completely abolished the ligand binding and functional response of the receptor. Additionally, point mutations at positions 241 and 251 between TM6 and i3 loop generated mutant receptors with modest constitutive activity via Gα15 signaling activation. Our results show that D134(3.49) on the highly conserved DRY motif has a distinct role for CXCR1 compared to its homologues (CXCR2 and KSHV-GPCR) in G protein coupling and receptor activation. In addition, M241(6.34) and F251(6.44) along with our previously identified V247(6.40) on TM6 are spatially located in a "hot spot" likely essential for CXCR1 activation. Identification of these amino acid residues may be useful for elucidating mechanism of CXCR1 activation and designing specific antagonists for the treatment of CXCR1-mediated diseases.

3.
Thromb Haemost ; 113(3): 505-12, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25373619

RESUMO

Atherosclerosis is primarily a disorder of lipid metabolism, but there is also a prominent chronic inflammatory component that drives the atherosclerotic lesion progression in the artery wall. During hyperlipidaemic conditions, there is a rapid influx of circulating monocytes into the atherosclerosis-prone areas of the arterial intima. These infiltrated monocytes differentiate into macrophages and take up the atherogenic lipoproteins in the intima of the vessel wall that have been modified within the lesion environment. Interleukin (IL)-10 is a prototypic anti-inflammatory cytokine made primarily by the macrophages and Th2 subtype T lymphocytes. In terms of atherosclerosis its major roles include inhibition of macrophage activation as well as inhibition of matrix metalloproteinase, pro-inflammatory cytokines and cyclooxygenase-2 expression in lipid-loaded and activated macrophage foam cells. Recent discoveries suggest another important role of IL-10 in atherosclerosis: its ability to alter lipid metabolism in macrophages. The current review will highlight the present knowledge on multiple ways in which IL-10 mediates atherosclerosis. As macrophages play a critical role in all stages of atherosclerosis, the review will concentrate on how IL-10 regulates the activities of macrophages that are especially important in the development of atherosclerosis.


Assuntos
Aterosclerose/prevenção & controle , Interleucina-10/metabolismo , Metabolismo dos Lipídeos , Macrófagos/metabolismo , Transdução de Sinais , Animais , Aterosclerose/imunologia , Aterosclerose/metabolismo , Humanos , Hipolipemiantes/uso terapêutico , Mediadores da Inflamação/metabolismo , Interleucina-10/uso terapêutico , Metabolismo dos Lipídeos/efeitos dos fármacos , Macrófagos/imunologia , Fatores de Proteção , Transdução de Sinais/efeitos dos fármacos
4.
PLoS One ; 9(8): e104039, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25090652

RESUMO

Even though combined anti-retroviral therapy (cART) dramatically improves patient survival, they remain at a higher risk of being afflicted with non-infectious complications such as cardiovascular disease (CVD). This increased risk is linked to persistent inflammation and chronic immune activation. In this study, we assessed whether this complication is related to HIV-derived ssRNAs inducing in macrophages increases in TNFα release through TLR8 activation leading to foam cell formation. HIV ssRNAs induced foam cell formation in monocyte-derived macrophages (MDMs) in a dose-dependent manner. This response was reduced when either endocytosis or endosomal acidification was inhibited by dynasore or chloroquine, respectively. Using a flow cytometry FRET assay, we demonstrated that ssRNAs bind to TLR8 in HEK cells. In MDMs, ssRNAs triggered a TLR8-mediated inflammatory response that ultimately lead to foam cell formation. Targeted silencing of the TLR8 and MYD88 genes reduced foam cell formation. Furthermore, foam cell formation induced by these ssRNAs was blocked by an anti-TNFα neutralizing antibody. Taken together in MDMs, HIV ssRNAs are internalized; bind TLR8 in the endosome followed by endosomal acidification. TLR8 signaling then triggers TNFα release and ultimately leads to foam cell formation. As this response was inhibited by a blocking anti-TNFα antibody, drug targeting HIV ssRNA-driven TLR8 activation may serve as a potential therapeutic target to reduce chronic immune activation and inflammation leading to CVD in HIV+ patients.


Assuntos
Células Espumosas/efeitos dos fármacos , HIV/química , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , RNA Viral/farmacologia , Receptor 8 Toll-Like/genética , Anticorpos Neutralizantes/farmacologia , Líquido da Lavagem Broncoalveolar/citologia , Cloroquina/farmacologia , Endocitose/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Células Espumosas/metabolismo , Células Espumosas/patologia , Regulação da Expressão Gênica , Células HEK293 , Humanos , Hidrazonas/farmacologia , Concentração de Íons de Hidrogênio , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patologia , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Cultura Primária de Células , Ligação Proteica/efeitos dos fármacos , Transdução de Sinais , Receptor 8 Toll-Like/agonistas , Receptor 8 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
5.
Adv Pharmacol ; 70: 265-301, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24931199

RESUMO

Chemokines are low-molecular-weight, secreted proteins that act as leukocyte-specific chemoattractants. The chemokine family has more than 40 members. Based on the position of two conserved cysteines in the N-terminal domain, chemokines can be divided into the CXC, C, CC, and CX3C subfamilies. The interaction of chemokines with their receptors mediates signaling pathways that play critical roles in cell migration, differentiation, and proliferation. The receptors for chemokines are G protein-coupled receptors (GPCRs), and thus far, seven CXC receptors have been cloned and are designated CXCR1-7. Constitutively active GPCRs are present in several human immune-mediated diseases and in tumors, and they have provided valuable information in understanding the molecular mechanism of GPCR activation. Several constitutively active CXC chemokine receptors include the V6.40A and V6.40N mutants of CXCR1; the D3.49V variant of CXCR2; the N3.35A, N3.35S, and T2.56P mutants of CXCR3; the N3.35 mutation of CXCR4; and the naturally occurring KSHV-GPCR. Here, we review the regulation of CXC chemokine receptor signaling, with a particular focus on the constitutive activation of these receptors and the implications in physiological conditions and in pathogenesis. Understanding the mechanisms behind the constitutive activation of CXC chemokine receptors may aid in pharmaceutical design and the screening of inverse agonists and allosteric modulators for the treatment of autoimmune diseases and cancers.


Assuntos
Mutação/genética , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Animais , Doença , Humanos , Ligantes , Receptores de Quimiocinas/química , Transdução de Sinais
6.
Am J Pathol ; 183(1): 313-25, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23685110

RESUMO

Chitinase 1 (CHIT1) is secreted by activated macrophages. Chitinase activity is raised in atherosclerotic patient sera and is present in atherosclerotic plaque. However, the role of CHIT1 in atherosclerosis is unknown. Preliminary studies of atherosclerosis in cynomolgous monkeys revealed CHIT1 to be closely correlated with areas of macrophage infiltration. Thus, we investigated the effects of a chitinase inhibitor, allosamidin, on macrophage function in vitro and on atherosclerotic development in vivo. In RAW264.7 cells, allosamidin elevated monocyte chemoattractant protein 1 and tumor necrosis factor alpha expression, and increased activator protein 1 and nuclear factor-κB transcriptional activity. Although inducible nitric oxide synthase, IL-6, and IL-1ß expression were increased, Arg1 expression was decreased by chitinase inhibition, suggesting that suppression of CHIT1 activity polarizes macrophages into a M1 phenotype. Allosamidin decreased scavenger receptor AI, CD36, ABCA1, and ABCG1 expression which led to suppression of cholesterol uptake and apolipoprotein AI-mediated cholesterol efflux in macrophages. These effects were confirmed with CHIT1 siRNA transfection and CHIT1 plasmid transfection experiments in primary macrophages. Apolipoprotein E-deficient hyperlipidemic mice treated for 6 weeks with constant administration of allosamidin and fed an atherogenic diet showed aggravated atherosclerotic lesion formation. These data suggest that CHIT1 exerts protective effects against atherosclerosis by suppressing inflammatory responses and polarizing macrophages toward an M2 phenotype, and promoting lipid uptake and cholesterol efflux in macrophages.


Assuntos
Acetilglucosamina/análogos & derivados , Aterosclerose/enzimologia , Quitinases/antagonistas & inibidores , Inibidores Enzimáticos/efeitos adversos , Macrófagos/enzimologia , Trissacarídeos/efeitos adversos , Acetilglucosamina/efeitos adversos , Animais , Aterosclerose/induzido quimicamente , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Biomarcadores/metabolismo , Linhagem Celular , Quitinases/metabolismo , Citocinas/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Infect Immun ; 81(1): 2-10, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23071135

RESUMO

Mycobacterium tuberculosis disease represents an enormous global health problem, with exceptionally high morbidity and mortality in HIV-seropositive (HIV(+)) persons. Alveolar macrophages from HIV(+) persons demonstrate specific and targeted impairment of critical host cell responses, including impaired M. tuberculosis-mediated tumor necrosis factor (TNF) release and macrophage apoptosis. Vitamin D may promote anti-M. tuberculosis responses through upregulation of macrophage NO, NADPH oxidase, cathelicidin, and autophagy mechanisms, but whether vitamin D promotes anti-M. tuberculosis mechanisms in HIV(+) macrophages is not known. In the current study, human macrophages exposed to M. tuberculosis demonstrated robust release of TNF, IκB degradation, and NF-κB nuclear translocation, and these responses were independent of vitamin D pretreatment. In marked contrast, HIV(+) U1 human macrophages exposed to M. tuberculosis demonstrated very low TNF release and no significant IκB degradation or NF-κB nuclear translocation, whereas vitamin D pretreatment restored these critical responses. The vitamin D-mediated restored responses were dependent in part on macrophage CD14 expression. Importantly, similar response patterns were observed with clinically relevant human alveolar macrophages from healthy individuals and asymptomatic HIV(+) persons at high clinical risk of M. tuberculosis infection. Taken together with the observation that local bronchoalveolar lavage fluid (BALF) levels of vitamin D are severely deficient in HIV(+) persons, the data from this study demonstrate that exogenous vitamin D can selectively rescue impaired critical innate immune responses in vitro in alveolar macrophages from HIV(+) persons at risk for M. tuberculosis disease, supporting a potential role for exogenous vitamin D as a therapeutic adjuvant in M. tuberculosis infection in HIV(+) persons.


Assuntos
Soropositividade para HIV/microbiologia , Macrófagos Alveolares/imunologia , Mycobacterium tuberculosis/imunologia , Receptores Toll-Like/imunologia , Fator de Necrose Tumoral alfa/imunologia , Vitamina D/farmacologia , Líquido da Lavagem Broncoalveolar/imunologia , Linhagem Celular , Soropositividade para HIV/imunologia , Soropositividade para HIV/metabolismo , Humanos , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/imunologia , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virologia , Mycobacterium tuberculosis/metabolismo , NF-kappa B/imunologia , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/imunologia , RNA Mensageiro/metabolismo , Transdução de Sinais/imunologia , Receptores Toll-Like/metabolismo , Tuberculose/metabolismo , Tuberculose/virologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Células U937 , Regulação para Cima/imunologia , Vitamina D/imunologia , Vitamina D/metabolismo
8.
PLoS One ; 7(8): e42765, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22936990

RESUMO

CXCR1, a classic GPCR that binds IL-8, plays a key role in neutrophil activation and migration by activating phospholipase C (PLC)ß through Gα(15) and Gα(i) which generates diacylglycerol and inositol phosphates (IPs). In this study, two conserved amino acid residues of CXCR1 on the transmembrane domain (TM) 3 and TM6, Leu128(3.43) (L128) and Val247(6.40) (V247), respectively, were selectively substituted with other amino acids to investigate the role of these conserved residues in CXCR1 activation. Although two selective mutants on Leu128, Leu128Ala (L128A) and Leu128Arg (L128R), demonstrated high binding affinity to IL-8, they were not capable of coupling to G proteins and consequently lost the functional response of the receptors. By contrast, among the four mutants at residue Val247 (TM6.40), replacing Val247 with Ala (V247A) and Asn (V247N) led to constitutive activation of mutant receptors when cotransfected with Gα(15). The V247N mutant also constitutively activated the Gα(i) protein. These results indicate that L128 on TM3.43 is involved in G protein coupling and receptor activation but is unimportant for ligand binding. On the other hand, V247 on TM6.40 plays a critical role in maintaining the receptor in the inactive state, and the substitution of V247 impaired the receptor constraint and stabilized an active conformation. Functionally, there was an increase in chemotaxis in response to IL-8 in cells expressing V247A and V247N. Our findings indicate that Leu128(3.43) and Val247(6.40) are critical for G protein coupling and activation of signaling effectors, providing a valuable insight into the mechanism of CXCR1 activation.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Leucina/química , Receptores de Interleucina-8A/química , Receptores de Interleucina-8A/metabolismo , Valina/química , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , AMP Cíclico/metabolismo , Citometria de Fluxo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/genética , Humanos , Fosfatos de Inositol/metabolismo , Leucina/genética , Microscopia Confocal , Mutagênese Sítio-Dirigida , Receptores de Interleucina-8A/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Relação Estrutura-Atividade , Valina/genética
9.
Circulation ; 126(18): 2236-47, 2012 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-23011471

RESUMO

BACKGROUND: Macrophages play a central role in the development of atherosclerosis. However, the signaling pathways that regulate their function are not well understood. The Rho-associated coiled-coil-containing kinases (ROCK1 and ROCK2) are serine-threonine protein kinases that are involved in the regulation of the actin cytoskeleton. Recent studies suggest that ROCK1 in macrophages and bone marrow-derived cells mediates atherogenesis. However, a similar role for ROCK2 in the pathogenesis of atherosclerosis has not been determined. METHODS AND RESULTS: The bone marrows from wild-type, ROCK2(+/-), and ROCK2(-/-) mice were transplanted into irradiated recipient low-density lipoprotein receptor(-/-) mice, and atherosclerosis was induced with a 16-week high-cholesterol diet. Compared with wild-type bone marrow-transplanted mice, ROCK2(+/-) bone marrow-transplanted and ROCK2(-/-) bone marrow-transplanted mice showed substantially less lipid accumulation in the aorta (8.46±1.42% and 9.80±2.34% versus 15.64±1.89%; P<0.01 for both) and decreased atherosclerotic lesions in the subaortic sinus (158.1±44.4 and 330.1±109.5×10(3)µm(2) versus 520.2±125.7×10(3)µm(2); P<0.01 for both). These findings correlated with decreased foam cell formation (2.27±0.57 versus 4.10±0.3; P<0.01) and increased cholesterol efflux (17.65±0.6 versus 9.75±0.8; P<0.05) in ROCK2-deficient mice that are mediated, in part, through the peroxisome proliferator-activated receptor-γ/liver X receptor/ATP-binding cassette transporter A1 pathway in macrophages. CONCLUSIONS: ROCK2 contributes to atherosclerosis, in part, by inhibiting peroxisome proliferator-activated receptor-γ-mediated reverse cholesterol transport in macrophages, which contributes to foam cell formation. These findings suggest that inhibition of ROCK2 in macrophages may have therapeutic benefits in preventing the development of atherosclerosis.


Assuntos
Aterosclerose/enzimologia , Colesterol/metabolismo , Macrófagos/enzimologia , Quinases Associadas a rho/fisiologia , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , Doenças da Aorta/enzimologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Doenças da Aorta/prevenção & controle , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Transplante de Medula Óssea , Colesterol na Dieta/farmacocinética , Colesterol na Dieta/toxicidade , Células Espumosas/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Lipoproteínas LDL/farmacologia , Receptores X do Fígado , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Nucleares Órfãos/metabolismo , PPAR gama/metabolismo , Quimera por Radiação , Receptores de LDL/deficiência , Receptores de LDL/genética , Transdução de Sinais/efeitos dos fármacos , Quinases Associadas a rho/deficiência , Quinases Associadas a rho/genética
10.
J Biol Chem ; 287(17): 13778-86, 2012 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-22393042

RESUMO

Human macrophages at mucosal sites are essential targets for acute HIV infection. During the chronic phase of infection, they are persistent reservoirs for the AIDS virus. HIV virions gain entry into macrophages following ligation of surface CD4-CCR5 co-receptors, which leads to the release of two copies of HIV ssRNA. These events lead to reverse transcription and viral replication initiation. Toll-like receptors TLR7 and TLR8 recognize specific intracellular viral ssRNA sequences, but in human alveolar macrophages, their individual roles in TLR-mediated HIV ssRNA recognition are unclear. In the current study, HIV-1 ssRNA induced TNFα release in a dose-dependent manner in adherent human macrophages expressing both intracellular TLR7 and TLR8. This response was reduced by inhibiting either endocytosis (50 µm dynasore) or endosomal acidification (1 µg/ml chloroquine). Either MYD88 or TLR8 gene knockdown with relevant siRNA reduced HIV-1 ssRNA-mediated TNFα release, but silencing TLR7 had no effect on this response. Furthermore, HIV-1 ssRNA induced histone 4 acetylation at the TNFα promoter as well as trimethylation of histone 3 at lysine 4, whereas TLR8 gene knockdown reduced these effects. Taken together in human macrophages, TLR8 binds and internalizes HIV ssRNA, leading to endosomal acidification, chromatin remodeling, and increases in TNFα release. Drugs targeting macrophage TLR8-linked signaling pathways may modulate the innate immune response to acute HIV infection by reducing viral replication.


Assuntos
Epigênese Genética , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/metabolismo , Macrófagos/citologia , RNA/metabolismo , Receptor 7 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Cromatina/química , Montagem e Desmontagem da Cromatina , Imunoprecipitação da Cromatina , Infecções por HIV/genética , Humanos , Macrófagos Alveolares/citologia , Oligonucleotídeos/química , RNA Viral/metabolismo
11.
J Immunol ; 187(11): 6052-8, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22025552

RESUMO

TLR-4-mediated signaling is significantly impaired in macrophages from HIV(+) persons, predominantly owing to altered MyD88-dependent pathway signaling caused in part by constitutive activation of PI3K. In this study we assessed in these macrophages if the blunted increase in TLR-4-mediated TNF-α release induced by lipid A (LA) is associated with PI3K-induced upregulation of mammalian target of rapamycin (mTOR) activity. mTOR inhibition with rapamycin enhanced TLR-4-mediated TNF-α release, but suppressed anti-inflammatory IL-10 release. Targeted gene silencing of mTOR in macrophages resulted in LA-induced TNF-α and IL-10 release patterns similar to those induced by rapamycin. Rapamycin restored MyD88/IL-1R-associated kinase interaction in a dose-dependent manner. Targeted gene silencing of MyD88 (short hairpin RNA) and mTOR (RNA interference) inhibition resulted in TLR-4-mediated 70-kDa ribosomal protein S6 kinase activation and enhanced TNF-α release, whereas IL-10 release was inhibited in both silenced and nonsilenced HIV(+) macrophages. Furthermore, mTOR inhibition augmented LA-induced TNF-α release through enhanced and prolonged phosphorylation of ERK1/2 and JNK1/2 MAPK, which was associated with time-dependent MKP-1 destabilization. Taken together, impaired TLR-4-mediated TNF-α release in HIV(+) macrophages is attributable in part to mTOR activation by constitutive PI3K expression in a MyD88-dependent signaling pathway. These changes result in MAPK phosphatase 1 stabilization, which shortens and blunts MAPK activation. mTOR inhibition may serve as a potential therapeutic target to upregulate macrophage innate immune host defense responsiveness in HIV(+) persons.


Assuntos
Infecções por HIV/metabolismo , Sistema de Sinalização das MAP Quinases/imunologia , Macrófagos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Western Blotting , Ativação Enzimática/imunologia , Ensaio de Imunoadsorção Enzimática , Técnicas de Silenciamento de Genes , Infecções por HIV/imunologia , Humanos , Imunoprecipitação , Macrófagos/imunologia , Macrófagos/virologia , Serina-Treonina Quinases TOR/imunologia , Receptor 4 Toll-Like/imunologia , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/imunologia
12.
FASEB J ; 24(8): 2869-80, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20354139

RESUMO

In atherogenesis, macrophage foam cell formation is modulated by pathways involving both the uptake and efflux of cholesterol. We recently showed that interleukin-10 (IL-10) modulates lipid metabolism by enhancing both uptake and efflux of cholesterol in macrophages. However, the mechanistic details of these properties in vivo have been unclear. Thus, the purpose of this study was to determine whether expression of IL-10 in macrophages would alter susceptibility to atherosclerosis and whether IL-10 exerts its antiatherosclerotic properties by modulating lipid metabolism in macrophages. We utilized a macrophage-specific retroviral vector that allows long-term in vivo expression of IL-10 in macrophages through transplantation of retrovirally transduced bone marrow cells (BMCs). IL-10 expressed by macrophages derived from transduced BMCs inhibited atherosclerosis in LDLR(-/-) mice by reducing cholesteryl ester accumulation in atherosclerotic sites. Experiments with primary macrophages indicated that macrophage source of IL-10 stimulated both the uptake (by up-regulating scavenger receptors) and efflux of cholesterol (by activating the PPARgamma-LXR-ABCA1/ABCG1 pathway), thereby reducing inflammation and apoptosis in atherosclerosis. These findings indicate that BMC-transduced macrophage IL-10 production can act as a strong antiatherogenic agent, and they highlight a novel antiatherosclerotic therapy using a simple, yet effective, stem cell transduction system that facilitates long-term expression of IL-10 in macrophages.


Assuntos
Aterosclerose/tratamento farmacológico , Hiperlipidemias/complicações , Interleucina-10/genética , Macrófagos/metabolismo , Animais , Apoptose/efeitos dos fármacos , Aterosclerose/patologia , Terapia Genética , Inflamação , Interleucina-10/biossíntese , Interleucina-10/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos , Transdução Genética
13.
J Biol Chem ; 284(47): 32950-8, 2009 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-19776020

RESUMO

Foam cell formation is a hallmark event during atherosclerosis. The current paradigm is that lipid uptake by scavenger receptor in macrophages initiates the chronic proinflammatory cascade and necrosis core formation that characterize atherosclerosis. We report here that a cytokine considered to be anti-atherogenic, interleukin-10 (IL10), promotes cholesterol uptake from modified lipoproteins in macrophages and its transformation into foam cells by increasing the expression of scavenger receptor CD36 and scavenger receptor A. Although uptake of modified lipoproteins is considered proatherogenic, we found that IL10 also increases cholesterol efflux from macrophages to protect against toxicity of free cholesterol accumulation in the cell. This process was PPARgamma-dependent and was mediated through up-regulation of ABCA1 (ATP-binding cassette transporter A1) protein expression. Importantly, expression of inflammatory molecules, such as tumor necrosis factor-alpha, intercellular adhesion molecule-1, and MMP9 as well as apoptosis were dramatically suppressed in lipid-laden foam cells treated with IL10. The notion that IL10 can mediate both the uptake of cholesterol from modified lipoproteins and the efflux of stored cholesterol suggests that the process of foam cell formation is not necessarily detrimental as long as mechanisms of cholesterol efflux and transfer to an exogenous acceptor are functioning robustly. Our results present a comprehensive antiatherogenic role of IL10 in macrophages, including enhanced disposal of harmful lipoproteins, inhibition of inflammatory molecules, and reduced apoptosis.


Assuntos
Aterosclerose/metabolismo , Colesterol/metabolismo , Interleucina-10/metabolismo , Macrófagos/metabolismo , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Antígenos CD36/metabolismo , Caspase 3/metabolismo , Linhagem Celular , Citocinas/metabolismo , Citometria de Fluxo , Lipídeos/química , Lipoproteínas/química , Camundongos , Regulação para Cima
14.
J Immunol ; 178(12): 8013-21, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17548638

RESUMO

Previously, we reported that normal colonocytes produce the memory CD4(+) T cell-directed chemokine MIP-3alpha, and that epithelial MIP-3alpha levels are elevated in inflammatory bowel disease. Interestingly, the unique receptor for MIP-3alpha, CCR6, is expressed by a variety of cell types including colonocytes, suggesting that MIP-3alpha may regulate additional biological activities in the intestine. The aim of this study was to determine whether MIP-3alpha can induce intestinal epithelial cell proliferation and to examine the signaling mechanisms that mediate this response. We show that nonstimulated Caco-2 and HT-29 colonic epithelial cells express CCR6, and that stimulation of Caco-2 cells by MIP-3alpha can dose dependently increase cell proliferation as well as activate the epidermal growth factor receptor (EGFR) and ERK1/2 MAPK. MIP-3alpha-mediated ERK1/2 activation in Caco-2 cells appeared to require metalloproteinase-dependent release of the endogenous EGFR ligand amphiregulin and transactivation of the EGFR. Moreover, blockade of amphiregulin bioactivity using a neutralizing polyclonal Ab significantly reduced MIP-3alpha-mediated, but not EGF-mediated Caco-2 cell proliferation. Taken together, our findings indicate that MIP-3alpha can regulate mitogenic signaling in colonic epithelial cells and thus may serve an important homeostatic function in the intestine by regulating tissue turnover and maintenance of the epithelium, in addition to its role in regulating leukocyte recruitment.


Assuntos
Quimiocinas CC/metabolismo , Colo/imunologia , Receptores ErbB/genética , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas Inflamatórias de Macrófagos/metabolismo , Ativação Transcricional , Anfirregulina , Anticorpos Monoclonais , Células CACO-2 , Proliferação de Células , Quimiocina CCL20 , Quimiocinas CC/farmacologia , Colo/efeitos dos fármacos , Família de Proteínas EGF , Glicoproteínas/antagonistas & inibidores , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Proteínas Inflamatórias de Macrófagos/farmacologia , Metaloproteases/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Receptores CCR6 , Receptores de Quimiocinas/metabolismo , Transdução de Sinais
15.
Pediatr Res ; 61(6): 716-21, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17426653

RESUMO

Necrotizing enterocolitis (NEC) is a major cause of morbidity and death in premature infants. NEC is associated with increased levels of pro-inflammatory cytokines in plasma and tissues that are regulated by the transcription factor nuclear factor-kappaB (NF-kappaB). It remains unknown, however, whether NF-kappaB mediates injury in neonatal NEC. We therefore examined the activation status of NF-kappaB perinatally in the small intestine and in a neonatal rat model of NEC. We found that intestinal NF-kappaB is strongly activated at birth and, in dam-fed newborn rats, is down-regulated within a day. In contrast, NF-kappaB remains strongly activated at both d 1 and d 2 in stressed animals, and this is accompanied by a significant decrease in the levels of the endogenous NF-kappaB inhibitor protein IkappaBalpha and IkappaBbeta at d 2. To determine the importance of elevated NF-kappaB activity in intestinal injury in NEC, we administered the NEMO-binding domain (NBD) peptide that selectively inhibits the critical upstream IkappaB kinase (IKK). NBD but not a control peptide decreased mortality and bowel injury in this model, supporting the hypothesis that bowel injury in NEC results from elevated NF-kappaB activity. Our findings therefore lead us to conclude that selective NF-kappaB inhibition represents a promising therapeutic strategy for NEC.


Assuntos
Enterocolite Necrosante/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Peptídeos/uso terapêutico , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Enterocolite Necrosante/patologia , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/patologia , Óxido Nítrico Sintase Tipo II/análise , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Ratos Sprague-Dawley
16.
Immunology ; 118(2): 153-63, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16771850

RESUMO

CXCL2 (macrophage inflammatory protein-2 (MIP-2)), a critical chemokine for neutrophils, has been shown to be produced in the rat intestine in response to platelet-activating factor (PAF) and to mediate intestinal inflammation and injury. The intestinal epithelium, constantly exposed to bacterial products, is the first line of defence against micro-organisms. It has been reported that enterocytes produce proinflammatory mediators, including tumour necrosis factor (TNF) and PAF, and we showed that lipopolysaccharide (LPS) and TNF activate nuclear factor (NF)-kappaB in enterocytes. However, it remains elusive whether enterocytes release CXCL2 in response to LPS and TNF via a NF-kappaB-dependent pathway and whether this involves the endogenous production of TNF and PAF. In this study, we found that TNF and LPS markedly induced CXCL2 gene expression in IEC-6 cells, TNF within 30 min, peaking at 45 min, while LPS more slowly, peaking after 2 hr. TNF- and LPS- induced CXCL2 gene expression and protein release were completely blocked by pyrrolidine dithiocarbamate (PDTC) and helenalin, two potent NF-kappaB inhibitors. NEMO-binding domain peptide, a specific inhibitor of inhibitor protein kappaB kinase (IKK) activation, a major upstream kinase mediating NF-kappaB activation, significantly blocked CXCL2 gene expression and protein release induced by LPS. WEB2170 (PAF antagonist) and anti-TNF antibodies had no effect on LPS-induced CXCL2 expression. In conclusion, CXCL2 gene is expressed in enterocytes in response to both TNF and LPS. LPS-induced CXCL2 expression is dependent on NF-kappaB activation via the IKK pathway. The effect of LPS is independent of endogenous TNF and PAF.


Assuntos
Quimiocinas CXC/imunologia , Enterócitos/imunologia , Lipopolissacarídeos/imunologia , NF-kappa B/imunologia , Animais , Azepinas/farmacologia , Linhagem Celular , Quimiocina CXCL2 , Quimiocinas CXC/biossíntese , Quimiocinas CXC/genética , Relação Dose-Resposta Imunológica , Ensaio de Desvio de Mobilidade Eletroforética , Enterócitos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Proteínas I-kappa B/metabolismo , Lipopolissacarídeos/antagonistas & inibidores , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Fator de Ativação de Plaquetas/imunologia , Pirrolidinas/farmacologia , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transcrição Gênica/efeitos dos fármacos , Triazóis/farmacologia , Fator de Necrose Tumoral alfa/imunologia
17.
Am J Physiol Gastrointest Liver Physiol ; 287(6): G1220-6, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15319184

RESUMO

Platelet-activating factor (PAF) is a potent endogenous mediator of bowel inflammation. It activates neutrophils that are needed to initiate the inflammatory response. Macrophage inflammatory protein-2 (MIP-2), a critical C-X-C chemokine secreted by macrophages and epithelial cells, is a potent chemoattractant for neutrophils. Whereas MIP-2 has been previously shown to mediate the injury in various organs, its role in acute intestinal injury has never been assessed. In this study, we first investigated the effect of PAF on MIP-2 expression in the intestine. Anesthetized young adult male Sprague-Dawley rats were injected intravenously with either PAF (1.5 microg/kg) or saline. Sixty minutes later, ileal MIP-2 gene expression was determined by semiquantitative RT-PCR, and plasma and ileal MIP-2 protein was determined by ELISA. In a second step, we assessed the role of MIP-2 in PAF-induced bowel injury. Rats were pretreated with rabbit anti-rat MIP-2 antibodies or control IgG for 90 min and then injected intravenously with PAF (2.5 microg/kg) for 90 min. We found that, in the rat intestine, 1) MIP-2 mRNA was only minimally expressed constitutively in sham-operated animals; 2) MIP-2 mRNA was significantly upregulated in response to PAF; 3) MIP-2 protein plasma levels and local production of MIP-2 in the ileum were markedly induced by PAF; 4) the administration of anti-rat MIP-2 IgG, but not control rabbit IgG, markedly reduced PAF-induced bowel injury (injury scores of 0.19 +/- 0.09 vs. 1.12 +/- 0.43, P < 0.05), hypotension, and leukopenia but did not reduce PAF-induced hemoconcentration. Thus we conclude that MIP-2 mediates PAF-induced intestinal injury.


Assuntos
Mediadores da Inflamação/farmacologia , Inflamação/patologia , Doenças Inflamatórias Intestinais/induzido quimicamente , Monocinas/biossíntese , Monocinas/genética , Fator de Ativação de Plaquetas/farmacologia , Animais , Anticorpos Bloqueadores/farmacologia , Quimiocina CXCL2 , Quimiocinas/farmacologia , Quimiocinas CXC/biossíntese , Quimiocinas CXC/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Hematócrito , Hipotensão/induzido quimicamente , Hipotensão/fisiopatologia , Íleo/efeitos dos fármacos , Íleo/metabolismo , Íleo/patologia , Inflamação/induzido quimicamente , Mediadores da Inflamação/antagonistas & inibidores , Doenças Inflamatórias Intestinais/patologia , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/genética , Contagem de Leucócitos , Leucopenia/induzido quimicamente , Masculino , Monocinas/antagonistas & inibidores , Infiltração de Neutrófilos/efeitos dos fármacos , Peroxidase/biossíntese , Fator de Ativação de Plaquetas/antagonistas & inibidores , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Immunology ; 106(4): 577-83, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12153521

RESUMO

Bacterial endotoxin (lipopolysaccharide; LPS) and platelet-activating factor (PAF) are important triggers of bowel inflammation and injury. We have previously shown that LPS activates the transcription factor nuclear factor (NF)-kappaB in the intestine, which up-regulates many pro-inflammatory genes. This effect partly depends on neutrophils and endogenous PAF. However, whether LPS and PAF directly activate NF-kappaB in enterocytes remains controversial. In this study, we first investigated the effect of LPS and PAF on NF-kappaB activation in IEC-6 (a non-transformed rat small intestinal crypt cell line) cells, by electrophoresis mobility shift assay and supershift, and found that LPS, but not PAF, activates NF-kappaB mostly as p50-p65 heterodimers. The effect was slower than tumour necrosis factor (TNF). Both LPS and TNF induce the expression of the NF-kappaB-dependent gene inducible nitric oxide synthase (iNOS), which occurs subsequent to NF-kappaB activation. We then examined the effect of LPS and TNF on the inhibitory molecules IkappaBalpha and IkappaBbeta. We found that TNF causes rapid degradation of IkappaBalpha and IkappaBbeta. In contrast, LPS did not change the levels of IkappaBalpha and IkappaBbeta up to 4 hr (by Western blot). However, in the presence of cycloheximide, there was a slow reduction of IkappaBalpha and IkappaBbeta, which disappeared almost completely at 4 hr. These observations suggest that LPS causes slow degradation and synthesis of IkappaBalpha and IkappaBbeta and therefore activates NF-kappaBeta via at least two mechanisms: initially, through an IkappaB-independent mechanism, and later, via an increased turnover of the inhibitor IkappaB. NF-kappaBeta activation precedes the gene expression of iNOS (assayed by reverse transcription-polymerase chain reaction), suggesting that LPS up-regulates iNOS via this transcription factor.


Assuntos
Endotoxinas/imunologia , Enterócitos/metabolismo , Proteínas I-kappa B , NF-kappa B/metabolismo , Fator de Ativação de Plaquetas/imunologia , Animais , Western Blotting , Comunicação Celular/imunologia , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta Imunológica , Sinergismo Farmacológico , Enterócitos/imunologia , Leucócitos Mononucleares/imunologia , Lipopolissacarídeos/imunologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Mol Cell Endocrinol ; 190(1-2): 19-27, 2002 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-11997175

RESUMO

The mammalian gonadotropin-releasing hormone receptor (GnRHR), with 327 amino acids, is among the smallest G protein coupled receptors identified. Absent from this receptor is the cytoplasmic tail, characteristic of other members of this superfamily, which frequently mediates desensitization and down-regulation. The fifteen carboxyl terminal residues in the mammalian GnRHR are absolutely conserved, suggesting important roles for these residues. In the current study, mutations of the mammalian GnRHR were made to study the carboxyl terminus. The receptor mutant GnRHR(Ser(326)Ala) was reduced in ligand affinity (117% reduction compared to wild type (wt)), while receptor numbers and internalization remained unchanged. GnRHR(Ser(326)Tyr) was decreased in effector coupling, while ligand affinity remained unchanged compared to wt. These studies also show that, while mutation of Ser(326) caused a change in ligand binding and effector coupling, truncation at this residue (GnRHR[des(326-327)]) had no measurable effect on GnRHR ligand binding, effector coupling or internalization, functions which appear to require different structural determinants than expression and routing. Removal of all three carboxyl terminal residues (Phe(325), Ser(326) and Leu(327)) or mutation of the receptor (GnRHR[Phe(325)Ala]) caused a complete loss of measurable ligand binding and effector coupling, clearly suggesting an unexplained role for Phe(325).


Assuntos
Endocitose/fisiologia , Receptores LHRH/genética , Receptores LHRH/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Ligantes , Microscopia Confocal , Dados de Sequência Molecular , Mutação , Ligação Proteica , Ratos , Receptores LHRH/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...