Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Placenta ; 39: 87-93, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26992680

RESUMO

OBJECTIVE: Maternal magnesium (Mg) deficiency has been associated with fetal growth restriction. Using a mouse model of maternal Mg deficiency-induced fetal growth restriction, we sought to investigate the effect of Mg deficiency on placental physiology and function. METHODS: In vivo: Pregnant Swiss Webster mice were fed either 100% of the recommended amount of Mg (control) or 10%Mg (Mg-deficient) (8 per group). Dams were euthanized on gestational day 17 and placentas were collected, weighed and assessed for Mg concentrations, as well as nutrient transporter mRNA expression. For nutrient transfer studies, control and Mg-deficient dams (6 per group) were injected with (14)C-amino acids and (3)H-glucose and trans-placental passage was determined. In vitro: BeWo placental cells were grown in media containing 10%Mg to 100%Mg and the effects of Mg status on cell proliferation, oxidative stress and nutrient uptake were measured. Data were analyzed by Student's t-tests comparing controls vs. Mg-deficient animals or cells. For multiple comparisons, data were analyzed by ANOVA followed by Dunnett's post hoc testing. RESULTS: In vivo: Maternal Mg deficiency decreased placental Mg content, placental and fetal weights, ratio of fetal:placental weight (P < 0.05), placental Slc7a5 transporter mRNA expression and transplacental nutrient transport (P < 0.05). In vitro: Mg deficiency reduced BeWo nutrient uptake (P < 0.01) and cell proliferation (P < 0.01), and increased oxidative stress (P < 0.01). CONCLUSION: These findings highlight the adverse effects of maternal Mg deficiency on fetal weight and placental function, including transport and proliferation and may explain the fetal growth restriction observed with moderate Mg deficiency in mice.


Assuntos
Deficiência de Magnésio/complicações , Deficiência de Magnésio/patologia , Placenta/patologia , Placenta/fisiologia , Complicações na Gravidez/patologia , Animais , Células Cultivadas , Feminino , Retardo do Crescimento Fetal/patologia , Retardo do Crescimento Fetal/fisiopatologia , Deficiência de Magnésio/fisiopatologia , Camundongos , Tamanho do Órgão , Gravidez , Complicações na Gravidez/fisiopatologia
2.
Immunol Res ; 63(1-3): 197-208, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26476732

RESUMO

Although classically characterized by chronic airway inflammation with eosinophil infiltration, asthma is a complex and multifactorial condition with numerous clinical phenotypes. Epidemiological studies strongly support the link between obesity and asthma and suggest that obesity precedes and promotes asthma development, increases asthma severity, and reduces steroid responsivity. Using a house dust mite (HDM) model of airway hyperresponsiveness in C57BL/6 mice, we examined the effects of diet-induced obesity on allergic airway inflammation and its treatment with dexamethasone. When compared to lean mice treated with HDM, obese-HDM mice had reduced plasma adiponectin, an anti-inflammatory adipokine, lower eosinophil and higher macrophage infiltration into the lungs and bronchoalveolar lavage (BAL) fluid, increased expression of total, M1, and M2 macrophage markers in the lungs, and enhanced Th2 and non-Th2 cytokine expression in the lungs. While Th2-associated responses in obese-HDM mice were suppressed by systemic dexamethasone, several Th2-independent responses, including total and M1 macrophage markers in the lungs, and lung CXC-motif ligand 1 (CXCL1) levels, were not improved following dexamethasone treatment. Thus, HDM combined with obesity promotes mixed localized inflammatory responses (e.g., M1, M2, Th1, and Th2) and shifts the cellular infiltration from eosinophils to macrophages, which are less sensitive to dexamethasone regulation. Because obese asthmatics exhibit more severe symptoms, lack a predominance of Th2 biomarkers, and are predicted to experience more steroid resistance when compared to lean asthmatics, this model could be used to study blunted steroid responses in obese-HDM mice and to define the macrophages found in the lungs.


Assuntos
Asma/imunologia , Eosinófilos/imunologia , Macrófagos/imunologia , Obesidade/imunologia , Sistema Respiratório/imunologia , Adiponectina/sangue , Animais , Antígenos de Dermatophagoides/imunologia , Asma/complicações , Asma/tratamento farmacológico , Citocinas/metabolismo , Dexametasona/administração & dosagem , Dieta , Progressão da Doença , Eosinófilos/efeitos dos fármacos , Humanos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/complicações , Obesidade/tratamento farmacológico , Pyroglyphidae/imunologia , Células Th2/efeitos dos fármacos , Células Th2/imunologia
3.
Placenta ; 33(5): 392-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22341339

RESUMO

OBJECTIVES: Maternal magnesium sulfate (MgSO4) administration exerts anti-inflammatory and fetal neuroprotective effects. Based on the link between placental inflammation and fetal immune responses, we examined the effect of MgSO4 on LPS-induced inflammation at the maternal-fetal interface. STUDY DESIGN: In vivo model: Pregnant rats (GD19) were injected intraperitoneally with saline, LPS, or MgSO4 plus LPS (n = 6 per group). Rats were euthanized; placentas were assayed for CCL2, IL6, and TNFα and placentas were screened for gene expression. Ex vivo model: Human placental cultures were treated with vehicle, LPS, or MgSO4 plus LPS. Supernatants were assayed for CCL2, IL6, and TNFα. In addition, placental cultures were analyzed for inflammation-related gene expression and NFκ B activation. RESULTS: In vivo model: Maternal LPS administration resulted in pro-inflammatory mediator production within the placenta; maternal MgSO4 treatment significantly attenuated LPS-induced inflammation. Several placental transcripts (APOE, CCL4, CXCL1, and NFκBIZ) differentially expressed following maternal LPS challenge were counter-regulated by MgSO4 treatment. Ex vivo model: LPS promoted human placental inflammation and MgSO4 significantly reduced inflammation induced by LPS. MgSO4 treatment of human placental explants significantly reversed the expression of numerous genes sensitive to LPS regulation and suppressed LPS-induced NFκB activation. CONCLUSIONS: MgSO4 administration inhibited placental inflammation during LPS-mediated maternal infection. Several placental inflammatory genes whose expression was regulated by LPS were reversed by MgSO4 treatment. Our data support the hypothesis that MgSO4 attenuates excessive inflammation at the maternal-fetal interface, which when uncontrolled may compromise neonatal health, including neurologic outcomes.


Assuntos
Corioamnionite/tratamento farmacológico , Sulfato de Magnésio/uso terapêutico , Placenta/efeitos dos fármacos , Complicações Infecciosas na Gravidez/tratamento farmacológico , Tocolíticos/uso terapêutico , Animais , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Lipopolissacarídeos , Sulfato de Magnésio/farmacologia , NF-kappa B/metabolismo , Placenta/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley , Tocolíticos/farmacologia
4.
Kidney Int ; 74(1): 62-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18401335

RESUMO

Inflammation plays a significant role in the pathophysiology of renal ischemia-reperfusion injury. Local inflammation is modulated by the brain via the vagus nerve and nicotinic acetylcholine receptors such that electrical or pharmacologic stimulation of this cholinergic anti-inflammatory pathway results in suppression of proinflammatory cytokine production. We examined the effects of cholinergic stimulation using agonists, nicotine or GTS-21, given before or after bilateral renal ischemia-reperfusion injury in rats. Pretreatment of rats with either agonist significantly attenuated renal dysfunction and tubular necrosis induced by renal ischemia. Similarly, tumor necrosis factor-alpha protein expression and leukocyte infiltration of the kidney were markedly reduced following treatment with cholinergic agonists. We found functional nicotinic acetylcholine receptors were present on rat proximal tubule epithelial cells. Cholinergic stimulation significantly decreased tubular necrosis in vagotomized rats after injury, implying an intact vagus nerve is not required for this renoprotective effect.


Assuntos
Agonistas Colinérgicos/farmacologia , Nefropatias/patologia , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Quimiotaxia de Leucócito , Agonistas Colinérgicos/uso terapêutico , Inflamação/patologia , Nefropatias/tratamento farmacológico , Masculino , Necrose/prevenção & controle , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/patologia , Fator de Necrose Tumoral alfa/análise , Nervo Vago
5.
Genes Immun ; 8(2): 147-53, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17268510

RESUMO

Neutrophils are required for the development of arthritis in rodents, and are the predominant cell in the synovial fluid of active rheumatoid arthritis. We hypothesized that neutrophil migration into the inflammed joint is genetically regulated. In addition, this genetic regulation would be accounted for by one of the arthritis loci that we have previously identified in an intercross between arthritis-susceptible DA and arthritis-resistant ACI rats studied for collagen-induced arthritis. We used the synovial-like air pouch model injected with carrageenan, and tested DA, ACI, and four congenic strains. ACI exudates had a significantly lower number of neutrophils compared with DA. Transfer of DA alleles at Cia7 into the ACI background, as in ACI.DA(Cia7) congenics, was enough to increase exudate neutrophil numbers to levels identical to DA, and this locus accounted for the difference between parental strains. None of the other congenic intervals explained the differences in exudate neutrophil counts. In conclusion, we have identified a novel function for Cia7, and determined that it regulates neutrophil migration into a synovial-like inflammatory site. Our data revealed no intrinsic defect in neutrophil responses to chemotactic agents, and suggest that Cia7 regulates an as yet unidentified factor central to neutrophil recruitment into inflammed tissues.


Assuntos
Artrite Reumatoide/genética , Artrite Reumatoide/imunologia , Movimento Celular/fisiologia , Neutrófilos/fisiologia , Locos de Características Quantitativas/genética , Líquido Sinovial/metabolismo , Animais , Carragenina , Quimiocinas/genética , Quimiotaxia/imunologia , Ensaio de Imunoadsorção Enzimática , Ratos , Ratos Endogâmicos ACI , Análise de Sequência de DNA , Especificidade da Espécie , Líquido Sinovial/imunologia
6.
Neuroscience ; 139(3): 1117-28, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16504406

RESUMO

Macrophage migration inhibitory factor is a potent proinflammatory cytokine; however, its role in spinal cord injury is poorly understood. Therefore, the aim of the present study was to investigate the effects of macrophage migration inhibitory factor on spinal cord neuron survival and viability. Due to the importance of nitric oxide metabolism in these events, part of our study was also focused on the influence of recombinant macrophage migration inhibitory factor on neuronal nitric oxide expression. Exposure of cultured mouse spinal cord neurons to macrophage migration inhibitory factor markedly increased cellular oxidative stress as measured by 2',7'-dichlorofluorescein fluorescence and intracellular calcium levels. In addition, an antagonist of the inositol 1,4,5-triphosphate receptor, 8-(diethylamino)octyl 3,4,5-trimethoxybenzoate, completely blocked the macrophage migration inhibitory factor-induced increase in intracellular calcium levels. Macrophage migration inhibitory factor treatment also decreased cell viability, increased cellular lactate dehydrogenase release, and induced chromatin condensation and aggregation in cultured spinal cord neurons. Finally, exposure to macrophage migration inhibitory factor markedly decreased expression and activity of neuronal nitric oxide, accompanied by a decrease in cellular guanosine 3'5'-cyclic monophosphate levels. The present results indicate that macrophage migration inhibitory factor can induce dysfunction of spinal cord neurons, leading to cell death through oxidative stress and intracellular calcium-dependent pathways.


Assuntos
Fatores Inibidores da Migração de Macrófagos/farmacologia , Neurônios/efeitos dos fármacos , Óxido Nítrico Sintase Tipo I/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Animais , Sinalização do Cálcio/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Células Cultivadas , GMP Cíclico/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Fatores Inibidores da Migração de Macrófagos/metabolismo , Camundongos , Neurônios/metabolismo , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo I/metabolismo , Proteínas Recombinantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/metabolismo
7.
Cell Mol Life Sci ; 60(7): 1342-50, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12943223

RESUMO

Following tissue damage, host wound healing ensues. This process requires an elaborate interplay between numerous cell types which orchestrate a series of regulated and overlapping events. These events include the initiation of an antigen-specific host immune response, blood vessel formation, as well as the production of critical extracellular matrix molecules, cytokines and growth factors which mediate tissue repair and wound closure. Connective tissue fibroblasts are considered essential for successful wound healing; however, their origin remains a mystery. A unique cell population, known as fibrocytes, has been identified and characterized. One of the unique features of these blood-borne cells is their ability to home to sites of tissue damage. This article reviews the identification and characterization of fibrocytes, summarizes the potential role of fibrocytes in the numerous steps of the wound-healing process and highlights the potential role of fibrocytes in fibrotic disease pathogenesis.


Assuntos
Fibroblastos/fisiologia , Cicatrização/fisiologia , Animais , Quimiocinas/fisiologia , Fibrose/patologia , Humanos , Proteínas de Membrana/fisiologia
8.
Proc Natl Acad Sci U S A ; 100(16): 9354-9, 2003 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-12878730

RESUMO

Macrophage migration inhibitory factor (MIF) is a mediator of host immunity and functions as a high, upstream activator of cells within the innate and the adaptive immunological systems. Recent studies have suggested a potentially broader role for MIF in growth regulation because of its ability to antagonize p53-mediated gene activation and apoptosis. To better understand MIF's activity in growth control, we generated and characterized a strain of MIF-knockout (MIF-KO) mice in the inbred, C57BL/6 background. Embryonic fibroblasts from MIF-KO mice exhibit p53-dependent growth alterations, increased p53 transcriptional activity, and resistance to ras-mediated transformation. Concurrent deletion of the p53 gene in vivo reversed the observed phenotype of cells deficient in MIF. In vivo studies showed that fibrosarcomas induced by the carcinogen benzo[alpha]pyrene are smaller in size and have a lower mitotic index in MIF-KO mice relative to their WT counterparts. The data provide direct genetic evidence for a functional link between MIF and the p53 tumor suppressor and indicate an important and previously unappreciated role for MIF in carcinogenesis.


Assuntos
Técnicas de Transferência de Genes , Fatores Inibidores da Migração de Macrófagos/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Alelos , Animais , Benzo(a)pireno , Carcinógenos , Dano ao DNA , Éxons , Fibroblastos/metabolismo , Fibrossarcoma/genética , Fibrossarcoma/patologia , Fatores Inibidores da Migração de Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Genéticos , Mutação , Fenótipo , Retroviridae/genética , Fatores de Tempo , Transcrição Gênica , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo
9.
Nat Immunol ; 2(11): 1061-6, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11668338

RESUMO

The cytokine macrophage-migration inhibitory factor (MIF) is secreted by a number of cell types upon induction by lipopolysaccharide (LPS). Because colitis is dependent on interplay between the mucosal immune system and intestinal bacteria, we investigated the role of MIF in experimental colitis. MIF-deficient mice failed to develop disease, but reconstitution of MIF-deficient mice with wild-type innate immune cells restored colitis. In addition, established colitis could be treated with anti-MIF immunoglobulins. Thus, murine colitis is dependent on continuous MIF production by the innate immune system. Because we found increased plasma MIF concentrations in patients with Crohn's disease, these data suggested that MIF is a new target for intervention in Crohn's disease.


Assuntos
Doenças Autoimunes/sangue , Colite/fisiopatologia , Doença de Crohn/sangue , Fatores Inibidores da Migração de Macrófagos/fisiologia , Transferência Adotiva , Animais , Doenças Autoimunes/imunologia , Transplante de Medula Óssea , Doença Crônica , Colite/imunologia , Colite/microbiologia , Colite/prevenção & controle , Colite/terapia , Doença de Crohn/imunologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Feminino , Humanos , Imunização Passiva , Lipopolissacarídeos/toxicidade , Ativação de Macrófagos/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/sangue , Fatores Inibidores da Migração de Macrófagos/deficiência , Fatores Inibidores da Migração de Macrófagos/genética , Fatores Inibidores da Migração de Macrófagos/farmacologia , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Proteínas Nucleares , Quimera por Radiação , Redução de Peso
10.
FASEB J ; 15(12): 2215-24, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11641248

RESUMO

Angiogenesis is an ordered process requiring the inter-play of numerous cellular and humoral factors. Studies over the past 20 years have identified several growth factors, cytokines, and enzymes that promote blood vessel formation. Most have revealed how individual factors promote an angiogenic phenotype in endothelial cells in vitro or contribute to blood vessel formation in vivo. However, the fundamental question that remains unanswered is how the cellular microenvironment contributes to angiogenesis. Fibrocytes are a recently characterized mesenchymal cell type isolated from peripheral blood that rapidly enter subcutaneously implanted wound chambers and sites of tissue injury. Here we describe the induction of an angiogenic phenotype in microvascular endothelial cells in vitro and promotion of angiogenesis in vivo by cultured fibrocytes. Fibrocytes constitutively secrete extracellular matrix-degrading enzymes, primarily matrix metalloproteinase 9, which promotes endothelial cell invasion. In addition, fibrocytes secrete several proangiogenic factors including VEGF, bFGF, IL-8, PDGF, and hematopoietic growth factors that promote endothelial cell migration, proliferation, and/or tube formation. By contrast, they do not produce representative antiangiogenic factors. Finally, both autologous fibrocytes and fibrocyte-conditioned media were found to induce blood vessel formation in vivo using the Matrigel angiogenesis model.


Assuntos
Endotélio Vascular/fisiologia , Fibroblastos/fisiologia , Neovascularização Fisiológica , Células 3T3 , Animais , Fatores Biológicos/farmacologia , Diferenciação Celular , Divisão Celular , Movimento Celular , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Endotélio Vascular/anatomia & histologia , Endotélio Vascular/citologia , Substâncias de Crescimento/farmacologia , Humanos , Metaloproteinase 9 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/genética , Mesoderma/citologia , Camundongos , Neovascularização Fisiológica/efeitos dos fármacos , Fenótipo , Cicatrização
11.
Insect Mol Biol ; 10(4): 323-31, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11520355

RESUMO

Studying tick feeding and digestion, we discovered in a cDNA library from partially fed Amblyomma americanum ticks the first known arthropod homologue of a human cytokine, the pro-inflammatory Macrophage Migration Inhibitory Factor (MIF). The tick origin of the MIF cDNA clone was confirmed by sequencing a genomic fragment that contained the full-length tick MIF gene with two introns. Antiserum to a tick MIF-specific peptide as well as antiserum to complete tick MIF revealed the expression of tick MIF in the salivary gland and midgut tissues of A. americanum ticks. In an in vitro functional assay, recombinant tick MIF inhibited the migration of human macrophages to the same extent that recombinant human MIF did.


Assuntos
Citocinas/genética , Fatores Inibidores da Migração de Macrófagos/genética , Carrapatos/genética , Sequência de Aminoácidos , Animais , Especificidade de Anticorpos , Proteínas de Artrópodes , Sequência de Bases , Citocinas/classificação , Citocinas/isolamento & purificação , Citocinas/fisiologia , DNA Complementar , Feminino , Expressão Gênica , Humanos , Fatores Inibidores da Migração de Macrófagos/classificação , Fatores Inibidores da Migração de Macrófagos/isolamento & purificação , Fatores Inibidores da Migração de Macrófagos/fisiologia , Macrófagos/fisiologia , Dados de Sequência Molecular , Coelhos , Homologia de Sequência de Aminoácidos , Carrapatos/imunologia , Distribuição Tecidual
12.
Transplantation ; 71(12): 1777-83, 2001 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-11455258

RESUMO

BACKGROUND: Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine that is a potent activator of macrophages and T cells. Previous studies have shown that local MIF production is increased in acute renal allograft rejection, suggesting that it may play an important role in the rejection process. AIMS: To determine if urine and serum MIF concentrations: (1) are increased in acute rejection, and (2) can be used as noninvasive tools to discriminate between acute rejection (AR) and cyclosporine nephrotoxicity (CyA toxicity). METHODS: In a prospective study of nine renal allograft patients (five acute rejection and four stable), serial urine MIF concentrations were measured by ELISA in the first 14 days after transplantation. In a retrospective study, MIF concentrations in urine and serum were measured in 24 patients who were biopsied for acute renal transplant dysfunction (11 AR, 13 CyA toxicity). Urine and serum MIF were also measured in 23 stable renal transplant patients and 10 normals. RESULTS: MIF was readily detected in the urine of normal healthy controls (106+/-61 pg/micromol creatinine). In the prospective study, the urinary MIF concentration was increased substantially on day 1 posttransplantation and subsequently fell in parallel with the serum creatinine. However, urine MIF increased before episodes of biopsy proven acute rejection. The retrospective study showed that urine MIF concentrations in patients with AR were increased 5-fold compared to normal controls (439+/-313 pg/micromol Cr; P<0.01). In contrast, urine MIF concentrations in CyA toxicity were not significantly different to normal controls (145+/-119 pg/micromol Cr; P=NS). A marked increase in MIF immunostaining was seen in biopsies of AR, but not in CyA toxicity. No significant differences were evident in serum MIF levels between normals and any transplant patient group. CONCLUSIONS: These results suggest that measurement of urine MIF concentration may be useful in monitoring renal transplant patients for acute rejection and as a discriminator from cyclosporine nephrotoxicity.


Assuntos
Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/urina , Transplante de Rim , Fatores Inibidores da Migração de Macrófagos/urina , Adulto , Ciclosporina/intoxicação , Diagnóstico Diferencial , Feminino , Humanos , Imunossupressores/intoxicação , Rim/metabolismo , Rim/patologia , Nefropatias/induzido quimicamente , Nefropatias/diagnóstico , Fatores Inibidores da Migração de Macrófagos/sangue , Fatores Inibidores da Migração de Macrófagos/metabolismo , Masculino , Pessoa de Meia-Idade , Concentração Osmolar , Estudos Prospectivos , Estudos Retrospectivos , Transplante Homólogo
13.
Arthritis Rheum ; 44(6): 1273-80, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11407686

RESUMO

OBJECTIVE: Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine with known actions in macrophage and T cell activation. MIF also has the unique capacity to reverse the inhibitory effects of glucocorticoids on these cells. We have recently demonstrated MIF expression in human rheumatoid arthritis (RA) synovium and cultured fibroblast-like synoviocytes (FLS), as well as the ability of FLS-derived MIF to induce monocyte release of tumor necrosis factor alpha. We investigated the effects of MIF on aspects of RA FLS activation, including the induction of phospholipase A2 (PLA2) and cyclooxygenase (COX). METHODS: PLA2 activity was measured by 3H-arachidonic acid released from treated FLS supernatants. COX activity was measured by prostaglandin E2 enzyme-linked immunosorbent assay. Cytosolic PLA2 (cPLA2) and COX-2 messenger RNA (mRNA) were determined using semiquantitative reverse transcriptase-polymerase chain reaction. RESULTS: Constitutive PLA2 activity was detected in RA FLS. Recombinant human MIF up-regulated PLA2 activity (P < 0.01) and cPLA2 mRNA expression, but had no effect on secretory PLA2. Recombinant human MIF up-regulated COX activity (P < 0.05) and COX-2 mRNA, but had no observable effect on COX-1. Interleukin-1beta (IL-1beta) significantly up-regulated PLA2 activity (P < 0.005) and cPLA2 mRNA expression while anti-MIF monoclonal antibody (mAb) significantly inhibited this IL-1beta-induced PLA2 activity (P < 0.02). Anti-MIF mAb significantly reduced IL-1beta-induced COX activity (P < 0.05) and COX-2 mRNA expression. CONCLUSION: MIF exerts a proinflammatory effect on key aspects of RA FLS activation. That anti-MIF mAb inhibited IL-1beta up-regulation of FLS indicates an additional cofactor role for MIF in IL-1beta-induced FLS activation. These data suggest that MIF antagonism has important therapeutic potential in RA.


Assuntos
Artrite Reumatoide/enzimologia , Isoenzimas/metabolismo , Fatores Inibidores da Migração de Macrófagos/farmacologia , Fosfolipases A/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Membrana Sinovial/efeitos dos fármacos , Anticorpos Bloqueadores/farmacologia , Células Cultivadas , Ciclo-Oxigenase 2 , Citosol/enzimologia , Primers do DNA/química , Relação Dose-Resposta a Droga , Indução Enzimática , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Isoenzimas/genética , Fatores Inibidores da Migração de Macrófagos/imunologia , Proteínas de Membrana , Fosfolipases A/genética , Fosfolipases A2 , Prostaglandina-Endoperóxido Sintases/genética , Proteínas Recombinantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Membrana Sinovial/enzimologia , Membrana Sinovial/patologia , Regulação para Cima
14.
J Immunol ; 166(12): 7556-62, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11390511

RESUMO

Fibrocytes are a distinct population of blood-borne cells that display a unique cell surface phenotype (collagen I+/CD11b+/CD13+/CD34+/CD45RO+/MHC class II+/CD86+) and exhibit potent immunostimulatory activities. Circulating fibrocytes rapidly enter sites of tissue injury, suggesting an important role for these cells in wound repair. However, the regulatory processes that govern the differentiation of blood-borne fibrocytes and the mechanisms that underlie the migration of these cells to wound sites are currently not known. We report herein that ex vivo cultured fibrocytes can differentiate from a CD14+-enriched mononuclear cell population and that this process requires contact with T cells. Furthermore, we demonstrate that TGF-beta1 (1-10 ng/ml), an important fibrogenic and growth-regulating cytokine involved in wound healing, increases the differentiation and functional activity of cultured fibrocytes. Because fibrocytes home to sites of tissue injury, we examined the role of chemokine/chemokine receptor interactions in fibrocyte trafficking. We show that secondary lymphoid chemokine, a ligand of the CCR7 chemokine receptor, acts as a potent stimulus for fibrocyte chemotaxis in vitro and for the homing of injected fibrocytes to sites of cutaneous tissue injury in vivo. Finally, we demonstrate that differentiated, cultured fibrocytes express alpha smooth muscle actin and contract collagen gels in vitro, two characteristic features of wound-healing myofibroblasts. These data provide important insight into the control of fibrocyte differentiation and trafficking during tissue repair and significantly expand their potential role during wound healing.


Assuntos
Movimento Celular/fisiologia , Fibroblastos/citologia , Cicatrização/fisiologia , Animais , Células Sanguíneas/citologia , Células Sanguíneas/metabolismo , Células Sanguíneas/patologia , Células Sanguíneas/transplante , Diferenciação Celular/fisiologia , Células Cultivadas , Colágeno/fisiologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibroblastos/transplante , Fibrose , Géis , Humanos , Injeções Intravenosas , Receptores de Lipopolissacarídeos/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Quimiocinas/biossíntese , Transplante de Células-Tronco , Células-Tronco/citologia , Células-Tronco/metabolismo , Células-Tronco/patologia , Fator de Crescimento Transformador beta/fisiologia , Fator de Crescimento Transformador beta1
15.
J Immunol ; 166(2): 747-53, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11145646

RESUMO

Macrophage migration inhibitory factor (MIF) has been shown to be a pivotal cytokine that mediates host inflammatory and immune responses. Recently, immunoneutralization of MIF has been found to inhibit tumor growth in mice; however, the contributing mechanisms underlying this effect have not been well defined. We investigated whether MIF plays a regulatory role in the expression of CTL activity. In a mouse model of the CTL response using the OVA-transfected tumor cell line EL4 (EG.7), we found that cultures of splenocytes obtained from EG.7-primed mice secrete high levels of MIF following Ag stimulation in vitro. Notably, parallel splenocyte cultures treated with neutralizing anti-MIF mAb showed a significant increase in the CTL response directed against EG.7 cells compared with control mAb-treated cultures. This effect was accompanied by elevated expression of IFN-gamma. Histological examination of the EG. 7 tumors from anti-MIF-treated animals showed a prominent increase in both CD4(+) and CD8(+) T cells as well as apoptotic tumor cells, consistent with the observed augmentation of CTL activity in vivo by anti-MIF. This increased CTL activity was associated with enhanced expression of the common gamma(c)-chain of the IL-2R that mediates CD8(+) T cell survival. Finally, CD8(+) T lymphocytes obtained from the spleens of anti-MIF-treated EG.7 tumor-bearing mice, when transferred into recipient tumor-bearing mice, showed increased accumulation in the tumor tissue. These data provide the first evidence of an important role for MIF in the regulation and trafficking of anti-tumor T lymphocytes in vivo.


Assuntos
Citotoxicidade Imunológica/imunologia , Fatores Inibidores da Migração de Macrófagos/fisiologia , Linfócitos T Citotóxicos/imunologia , Adjuvantes Imunológicos/administração & dosagem , Transferência Adotiva , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Movimento Celular/imunologia , Células Cultivadas , Testes Imunológicos de Citotoxicidade , Feminino , Injeções Intraperitoneais , Linfócitos do Interstício Tumoral/imunologia , Fatores Inibidores da Migração de Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Timoma/imunologia , Timoma/patologia , Timoma/prevenção & controle , Células Tumorais Cultivadas/transplante
16.
Infect Immun ; 68(4): 2259-67, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10722628

RESUMO

Human falciparum malaria, caused by Plasmodium falciparum infection, results in 1 to 2 million deaths per year, mostly children under the age of 5 years. The two main causes of death are severe anemia and cerebral malaria. Malarial anemia is characterized by parasite red blood cell (RBC) destruction and suppression of erythropoiesis (the mechanism of which is unknown) in the presence of a robust host erythropoietin response. The production of a host-derived erythropoiesis inhibitor in response to parasite products has been implicated in the pathogenesis of malarial anemia. The identity of this putative host factor is unknown, but antibody neutralization studies have ruled out interleukin-1beta, tumor necrosis factor alpha, and gamma interferon while injection of interleukin-12 protects susceptible mice against lethal P. chabaudi infection. In this study, we report that ingestion of P. chabaudi-infected erythrocytes or malarial pigment (hemozoin) induces the release of macrophage migration inhibitory factor (MIF) from macrophages. MIF, a proinflammatory mediator and counter-regulator of glucocorticoid action, inhibits erythroid (BFU-E), multipotential (CFU-GEMM), and granulocyte-macrophage (CFU-GM) progenitor-derived colony formation. MIF was detected in the sera of P. chabaudi-infected BALB/c mice, and circulating levels correlated with disease severity. Liver MIF immunoreactivity increased concomitant with extensive pigment and parasitized RBC deposition. Finally, MIF was elevated three- to fourfold in the spleen and bone marrow of P. chabaudi-infected mice with active disease, as compared to early disease, or of uninfected controls. In summary, the present results suggest that MIF may be a host-derived factor involved in the pathophysiology of malaria anemia.


Assuntos
Anemia/parasitologia , Eritrócitos/parasitologia , Fatores Inibidores da Migração de Macrófagos/biossíntese , Macrófagos/metabolismo , Malária/parasitologia , Plasmodium chabaudi/imunologia , Anemia/etiologia , Animais , Medula Óssea/metabolismo , Medula Óssea/parasitologia , Células Cultivadas , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Células Precursoras Eritroides/metabolismo , Eritropoese/efeitos dos fármacos , Eritropoese/fisiologia , Eritropoetina/farmacologia , Feminino , Imuno-Histoquímica , Leucopoese , Fígado/metabolismo , Fígado/parasitologia , Fatores Inibidores da Migração de Macrófagos/sangue , Fatores Inibidores da Migração de Macrófagos/fisiologia , Malária/complicações , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Baço/metabolismo , Baço/parasitologia , Fatores de Tempo
17.
Nat Med ; 6(2): 164-70, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10655104

RESUMO

Identification of new therapeutic targets for the management of septic shock remains imperative as all investigational therapies, including anti-tumor necrosis factor (TNF) and anti-interleukin (IL)-1 agents, have uniformly failed to lower the mortality of critically ill patients with severe sepsis. We report here that macrophage migration inhibitory factor (MIF) is a critical mediator of septic shock. High concentrations of MIF were detected in the peritoneal exudate fluid and in the systemic circulation of mice with bacterial peritonitis. Experiments performed in TNFalpha knockout mice allowed a direct evaluation of the part played by MIF in sepsis in the absence of this pivotal cytokine of inflammation. Anti-MIF antibody protected TNFalpha knockout from lethal peritonitis induced by cecal ligation and puncture (CLP), providing evidence of an intrinsic contribution of MIF to the pathogenesis of sepsis. Anti-MIF antibody also protected normal mice from lethal peritonitis induced by both CLP and Escherichia coli, even when treatment was started up to 8 hours after CLP. Conversely, co-injection of recombinant MIF and E. coli markedly increased the lethality of peritonitis. Finally, high concentrations of MIF were detected in the plasma of patients with severe sepsis or septic shock. These studies define a critical part for MIF in the pathogenesis of septic shock and identify a new target for therapeutic intervention.


Assuntos
Infecções Bacterianas/prevenção & controle , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Choque Séptico/prevenção & controle , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Infecções Bacterianas/metabolismo , Feminino , Humanos , Fatores Inibidores da Migração de Macrófagos/imunologia , Fatores Inibidores da Migração de Macrófagos/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Peritonite/metabolismo , Peritonite/prevenção & controle , Choque Séptico/metabolismo
18.
Kidney Int ; 57(2): 499-509, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10652026

RESUMO

BACKGROUND: We have recently demonstrated that macrophage migration inhibitory factor (MIF) plays a pathogenic role in experimental glomerulonephritis (GN). The aim of the current study was to investigate MIF expression in human GN. METHODS: MIF expression was examined by in situ hybridization and immunohistochemistry staining in 65 biopsies from a variety of glomerulonephridities. RESULTS: There is constitutive expression of MIF mRNA and protein in normal human kidney that is largely restricted to tubular epithelial cells and to some glomerular epithelial cells. There was little change in the pattern of MIF expression in nonproliferative forms of GN such as minimal change disease and membranous GN. However, there was a marked increase in both glomerular and tubular MIF expression in proliferative forms of GN, including focal segmental glomerulosclerosis (FGS), lupus nephritis, crescentic GN, and mesangiocapillary proliferative GN. The prominent macrophage and T-cell infiltrate in these diseases were largely restricted to areas with marked up-regulation of MIF expression, contributing to glomerular hypercellularity, glomerular focal segmental lesions, crescent formation, tubulitis, and granulomatous lesions. De novo MIF expression was evident in glomerular endothelial cells and mesangial cells in proliferative forms of GN. In addition, many infiltrating macrophages and T cells showed MIF mRNA and protein expression. Quantitative analysis found that increased glomerular and tubular MIF expression gave a highly significant correlation with macrophage and T-cell accumulation, the severity of histologic lesions, and the loss of creatinine clearance. CONCLUSIONS: Renal MIF expression is markedly up-regulated in proliferative forms of human GN, and this correlates with leukocyte infiltration, histologic damage, and renal function impairment. These results suggest that MIF may be an important mediator of renal injury in progressive forms of human GN. Based on these findings, together with the known pathogenic role of MIF in experimental GN, we propose that MIF is an attractive therapeutic target in the treatment of progressive forms of GN.


Assuntos
Glomerulonefrite Membranoproliferativa/imunologia , Glomerulonefrite Membranosa/imunologia , Fatores Inibidores da Migração de Macrófagos/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Biópsia , Estudos de Coortes , Células Epiteliais/química , Células Epiteliais/imunologia , Feminino , Expressão Gênica/imunologia , Glomerulonefrite Membranoproliferativa/patologia , Glomerulonefrite Membranosa/patologia , Humanos , Hibridização In Situ , Glomérulos Renais/patologia , Glomérulos Renais/fisiologia , Fatores Inibidores da Migração de Macrófagos/análise , Macrófagos/química , Macrófagos/imunologia , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/análise , Valores de Referência , Linfócitos T/química , Linfócitos T/imunologia
20.
J Biol Chem ; 274(25): 18100-6, 1999 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-10364264

RESUMO

Macrophage migration inhibitory factor (MIF) is an important pro-inflammatory mediator with the unique ability to counter-regulate the inhibitory effects of glucocorticoids on immune cell activation. MIF is released from cells in response to glucocorticoids, certain pro-inflammatory stimuli, and mitogens and acts to regulate glucocorticoid action on the ensuing inflammatory response. To gain insight into the molecular mechanism of MIF action, we have examined the role of MIF in the proliferation and intracellular signaling events of the well characterized, NIH/3T3 fibroblast cell line. Both endogenously secreted and exogenously added MIFs stimulate the proliferation of NIH/3T3 cells, and this response is associated with the activation of the p44/p42 extracellular signal-regulated (ERK) mitogen-activated protein kinases (MAP). The MIF-induced activation of these kinases was sustained for a period of at least 24 h and was dependent upon protein kinase A activity. We further show that MIF regulates cytosolic phospholipase A2 activity via a protein kinase A and ERK dependent pathway and that the glucocorticoid suppression of cytokine-induced cytoplasmic phospholipase A2 activity and arachidonic acid release can be reversed by the addition of recombinant MIF. These studies indicate that the sustained activation of p44/p42 MAP kinase and subsequent arachidonate release by cytoplasmic phospholipase A2 are important features of the immunoregulatory and intracellular signaling events initiated by MIF and provide the first insight into the mechanisms that underlie the pro-proliferative and inflammatory properties of this mediator.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Sulfonamidas , Células 3T3 , Animais , Ácido Araquidônico/metabolismo , Divisão Celular/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citosol/enzimologia , Dexametasona/farmacologia , Ativação Enzimática/efeitos dos fármacos , Isoquinolinas/farmacologia , Fatores Inibidores da Migração de Macrófagos/farmacologia , Camundongos , Fosforilação , Proteínas Recombinantes/farmacologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA