Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Exp Med ; 178(6): 2193-200, 1993 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-7504060

RESUMO

Vascular endothelium activated by endotoxin (lipopolysaccharide [LPS]) and cytokines plays an important role in organ inflammation and blood leukocyte recruitment observed during sepsis. Endothelial cells can be activated by LPS directly, after its interaction with LPS-binding protein and soluble CD14 in plasma. LPS-LPS-binding protein complexes in blood also interact with monocytes and neutrophils bearing glycosyl-phosphatidylinositol (GPI) anchored membrane CD14 (mCD14), promoting the release of cytokines such as tumor necrosis factor and interleukin 1 (IL-1). These molecules, in turn, have the capacity to activate endothelial cells providing an indirect pathway for LPS-dependent endothelial cell activation. In this work, we address the relative importance of the direct and the indirect pathway of in vitro LPS-induced human umbilical vein endothelial cell (HUVEC) activation. Substituting whole blood for plasma resulted in a 1,000-fold enhancement of HUVEC sensitivity to LPS. Both blood- and plasma-dependent enhanced activation of HUVEC were blocked with an anti-CD14 monoclonal antibody. Blood from patients with paroxysmal nocturnal hemoglobinuria, whose cells lack mCD14 and other GPI anchored proteins, was unable to enhance LPS activation of HUVEC above the level observed with plasma alone. IL-10, an inhibitor of monocyte release of cytokines, decreased the blood-dependent enhancement of HUVEC activation by LPS. Blood adapted to small doses of LPS was also less efficient than nonadapted blood in producing this enhancement. Addition of purified mononuclear cells to HUVEC or the transfer of plasma from whole blood incubated with LPS to HUVEC, duplicated the enhancement effect observed when whole blood was incubated with HUVEC. Taken together, these data suggest that the indirect pathway of LPS activation of endothelial cell is mediated by monocytes and mCD14 through the secretion of a soluble mediator(s). The indirect pathway is far more efficient than the direct, plasma-dependent pathway.


Assuntos
Antígenos CD/fisiologia , Antígenos de Diferenciação Mielomonocítica/fisiologia , Adesão Celular , Endotélio Vascular/fisiologia , Lipopolissacarídeos/farmacologia , Monócitos/fisiologia , Antígenos CD/química , Antígenos de Diferenciação Mielomonocítica/química , Moléculas de Adesão Celular/metabolismo , Endotélio Vascular/citologia , Hemoglobinúria Paroxística/fisiopatologia , Humanos , Técnicas In Vitro , Interleucina-10/farmacologia , Receptores de Lipopolissacarídeos , Solubilidade , Regulação para Cima , Molécula 1 de Adesão de Célula Vascular
2.
J Exp Med ; 164(3): 777-93, 1986 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-2427635

RESUMO

This report describes the purification of an acute phase reactant from acute phase rabbit serum, which endows normal serum with the properties of acute phase serum, insofar as LPS is concerned. The acute phase reactant is referred to as LPS-binding protein, or LBP. LBP was purified approximately 2,000-fold by chromatography of acute phase serum on Bio-Rex 70 and Mono-Q resins. The resulting preparation consisted of two glycoproteins having molecular weights of 60,500 and 58,000; the two were obtained in a variable ratio, usually near 10:1, respectively. After separation by SDS-PAGE, the N-terminal 36 amino acid sequences of the two proteins were identical. From the N-terminal sequence, as well as other properties of LBP, LBP appears to be unrelated to any known acute phase reactants. The direct interaction of LPS and LBP was inferred from two types of evidence: first, immunoprecipitation of [3H]LPS from APRS by anti-LBP sera; and second, by the 125I-labeling of LBP when APRS-containing 125I-labeled 2-(p-azidosalicylamido)ethyl 1,3'-dithiopropionyl-LPS was photolysed. The data presented here support the concept that the 60-kD glycoprotein we have termed LBP is a newly recognized acute phase reactant that may modulate the biochemical and biologic properties of LPS in vivo.


Assuntos
Proteínas Sanguíneas/isolamento & purificação , Proteínas de Transporte/isolamento & purificação , Lipopolissacarídeos/metabolismo , Proteínas de Fase Aguda , Sequência de Aminoácidos , Animais , Proteínas Sanguíneas/análise , Proteínas Sanguíneas/imunologia , Proteínas de Transporte/análise , Proteínas de Transporte/imunologia , Cromatografia , Soros Imunes/imunologia , Coelhos , Ratos
3.
J Exp Med ; 181(3): 857-66, 1995 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-7532683

RESUMO

A murine model system was used to study the distribution and regulation of CD14 gene expression in vivo. Western blot analysis failed to detect CD14 in plasma from untreated CB6 (BALB/c x C57Bl6) mice, but showed markedly increased levels of CD14 in plasma from mice treated with lipopolysaccharide (LPS). Plasma levels of CD14 increased in a time- and dose-dependent manner, reaching a maximum between 8 and 16 h. Northern blot analysis of total RNA extracted from mouse tissues revealed low, but significant, levels of CD14 mRNA in many tissues of untreated animals with the highest levels in uterus, adipose tissue, and lung. After intraperitoneal injection of LPS, induction of CD14 gene expression was detected in all organs examined with the extent of induction varying between organs. Induction of CD14 mRNA was both time and dose dependent. Maximum induction in the heart and lung was observed 2-4 h after injection of LPS, while liver and kidney showed maximal induction between 8 and 16 h. In situ hybridization showed that CD14 mRNA was expressed in myeloid cells in many tissues, and that expression in these cells was upregulated by LPS. Unexpectedly, CD14 mRNA was also detected in other cells within tissues, including epithelial cells, and expression in these cell types also was upregulated by LPS. Immunochemical analysis revealed that CD14 antigen colocalized to the cytoplasm of cells expressing CD14 mRNA. These studies demonstrate that CD14 gene expression is not restricted to myeloid cells, and that the level of expression of CD14 is influenced by exposure to LPS.


Assuntos
Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Animais , Antígenos CD/biossíntese , Antígenos CD/sangue , Antígenos de Diferenciação Mielomonocítica/biossíntese , Antígenos de Diferenciação Mielomonocítica/sangue , Receptores de Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , RNA Mensageiro/análise
4.
J Exp Med ; 176(2): 485-94, 1992 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-1380063

RESUMO

Lipopolysaccharides (LPS) that lack acyloxyacyl groups can antagonize responses to LPS in human cells. Although the site and mechanism of inhibition are not known, it has been proposed that these inhibitory molecules compete with LPS for a common cellular target such as a cell-surface binding receptor. In the present study, we used an in vitro model system to test this hypothesis and to evaluate the role of CD14 in cellular responses to LPS. Cells of the THP-1 human monocyte-macrophage cell line were exposed to 1,25 dihydroxyvitamin D3 to induce adherence to plastic and expression of CD14, a binding receptor for LPS complexed with LPS-binding protein (LBP). The uptake of picograms of [3H]LPS (agonist) and enzymatically deacylated LPS [3H]dLPS (antagonist) was measured by exposing the cells to the radiolabeled ligands for short incubation periods. The amounts of cell-associated LPS and dLPS were then correlated with cellular responses by measuring the induction of nuclear NF-kappa B binding activity and the production of cell-associated interleukin (IL)-1 beta. We found that similar amounts of [3H]LPS or [3H]dLPS were taken up by the cells. The rate of cellular accumulation of the ligands was greatly enhanced by LBP and blocked by a monoclonal antibody to CD14 (mAb 60b), yet no cellular responses were induced by dLPS or dLPS-LBP complexes. In contrast, LPS stimulated marked increases of NF-kappa B binding activity and IL-1 beta. These responses were enhanced by LBP and inhibited by mAb 60b. dLPS and its synthetic lipid A counterpart, LA-14-PP (also known as lipid Ia, lipid IVa, or compound 406) strongly inhibited LPS-induced NF-kappa B and IL-1 beta, yet neither antagonist inhibited the uptake of LPS via CD14. dLPS did not inhibit NF-kappa B responses to tumor necrosis factor (TNF) alpha or phorbol ester. Our results indicate that (a) both stimulatory and nonstimulatory ligands can bind to CD14 in the presence of LBP; (b) the mechanism of inhibition by dLPS is LPS-specific, yet does not involve blockade of LPS binding to CD14; and (c) in keeping with previous results of others, large concentrations of LPS can stimulate the cells in the absence of detectable binding to CD14. The findings indicate that the site of dLPS inhibition is distal to CD14 binding in the LPS signal pathway in THP-1 cells, and suggest that molecules other than CD14 are important in LPS signaling.


Assuntos
Proteínas de Fase Aguda , Antígenos CD/fisiologia , Antígenos de Diferenciação Mielomonocítica/fisiologia , Lipopolissacarídeos/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana , Anticorpos Monoclonais/imunologia , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Sequência de Bases , Proteínas de Transporte/metabolismo , Adesão Celular , Linhagem Celular , Colecalciferol/fisiologia , DNA , Humanos , Interleucina-1/metabolismo , Cinética , Receptores de Lipopolissacarídeos , Macrófagos/citologia , Dados de Sequência Molecular , NF-kappa B/metabolismo , Plásticos
5.
J Exp Med ; 170(4): 1231-41, 1989 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-2477488

RESUMO

Lipopolysaccharide binding protein (LBP) is an acute-phase reactant that binds bacterial LPS. We show that LBP binds to the surface of live Salmonella and to LPS coated erythrocytes (ELPS), and strongly enhances the attachment of these particles to macrophages. LBP bridges LPS-coated particles to macrophages (MO) by first binding to the LPS, then binding to MO. Pretreatment of ELPS with LBP enabled binding to MO, but pretreatment of MO had no effect. Moreover, MO did not recognize erythrocytes coated with LBP unless LPS was also added, thus suggesting that interaction of LBP with LPS results in a conformational change in LBP that allows recognition by MO. Binding of LBP-coated particles appears to be mediated by a receptor found on blood monocytes and MO but not on other leukocytes or umbilical vein endothelium. The receptor is mobile in the plane of the membrane since binding activity on MO was downmodulated upon spreading of cells on surfaces coated with LBP-LPS complexes. The receptor appears to be distinct from other opsonic receptors since downmodulation of CR1, CR3, Fc gamma RI, Fc gamma RII, and Fc gamma RIII with mAbs did not affect binding of LBP-coated particles, and leukocytes from CD18-deficient patients bound LBP-coated particles normally. Coating of erythrocytes with LBP-LPS complexes strongly enhanced phagocytosis observed in the presence of suboptimal amounts of anti-erythrocyte IgG. However, binding mediated by LBP-LPS complexes alone caused neither phagocytosis of the LBP-coated erythrocytes nor initiation of an oxidative burst. The results of our studies define LBP as an opsonin. During the acute phase, LBP can be expected to bind gram-negative bacteria and bacterial fragments and promote the interaction of coated bacteria with phagocytes.


Assuntos
Proteínas de Fase Aguda/metabolismo , Proteínas de Transporte/metabolismo , Lipopolissacarídeos/metabolismo , Macrófagos/fisiologia , Glicoproteínas de Membrana , Proteínas Opsonizantes , Receptores de Superfície Celular/fisiologia , Membrana Eritrocítica , Técnicas In Vitro , Substâncias Macromoleculares , Fluidez de Membrana , Proteínas de Membrana/metabolismo , Consumo de Oxigênio , Fagocitose , Receptores de Complemento/fisiologia , Receptores de Complemento 3b , Receptores Fc/fisiologia
6.
J Exp Med ; 175(6): 1697-705, 1992 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-1375269

RESUMO

Bacterial endotoxin (lipopolysaccharide [LPS]) causes fatal shock in humans and experimental animals. The shock is mediated by cytokines released by direct LPS stimulation of cells of monocytic origin (monocyte/macrophage [MO]). Recent studies have supported the concept that the plasma protein, LPS binding protein (LBP), plays an important role in controlling MO responses to LPS. Specifically, evidence has been presented to suggest that CD14, a membrane protein present in MO, serves as a receptor for complexes of LPS and the plasma protein LPS binding protein (LBP). In this function CD14 mediates attachment of LPS-bearing particles opsonized with LBP and appears to play an important role in regulating cytokine production induced by complexes of LPS and LBP. The CD14-, murine pre-B cell line 70Z/3 responds to LPS by synthesis of kappa light chains and consequent expression of surface IgM. To better understand the role of CD14 in controlling cellular responses to LPS, we investigated the effect of transfection of CD14 into 70Z/3 cells on LPS responsiveness. We report here that transfection of human or rabbit CD14 cDNA into 70Z/3 cells results in membrane expression of a glycosyl-phosphatidylinositol-anchored CD14. When LPS is complexed with LBP, CD14-bearing 70Z/3 cells bind more LPS than do the parental or 70Z/3 cells transfected with vector only. Remarkably, the expression of CD14 lowers the amount of LPS required to stimulate surface IgM expression by up to 10,000-fold when LPS dose-response curves in the CD14-, parental and CD14-bearing, transfected 70Z/3 cells are compared. In contrast, the response of CD14-bearing 70Z/3 cells and the parental 70Z/3 cell line (CD14-) to interferon gamma is indistinguishable. LPS stimulation of the parental and CD14-bearing 70Z/3 cells results in activation of NF-kB. These data provide evidence to support the concept that the LPS receptor in cells that constitutively express CD14 may be a multiprotein complex containing CD14 and membrane protein(s) common to a diverse group of LPS-responsive cells.


Assuntos
Proteínas de Fase Aguda , Antígenos CD/fisiologia , Antígenos de Diferenciação Mielomonocítica/fisiologia , Proteínas de Transporte/metabolismo , Lipopolissacarídeos/metabolismo , Glicoproteínas de Membrana , Receptores Imunológicos/metabolismo , Animais , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Linfócitos B , Sequência de Bases , Linhagem Celular , Imunoglobulina M/biossíntese , Cadeias kappa de Imunoglobulina/biossíntese , Cinética , Receptores de Lipopolissacarídeos , Lipopolissacarídeos/toxicidade , Camundongos , Dados de Sequência Molecular , Monócitos/fisiologia , NF-kappa B/metabolismo , Oligodesoxirribonucleotídeos , Transfecção
7.
J Exp Med ; 142(6): 1570-90, 1975 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-1104750

RESUMO

We have studied the role of complement in lipopolysaccharide (LPS)-induced hypotension and disseminated intravascular coagulation (DIC) by comparing the effects of injection of three preparations of LPS from E. Coli 0111:B4, S. minnesota Re595, and S. marcescens. Injections of nonlethal doses of these LPS preparations into normal rabbits produced decreases in mean arterial blood pressure during a 5-h period. When rabbits treated with cobra venom factor (CoF) to deplete C3 were injected with the various LPS preparations, mean arterial pressures fell at a rate and extent essentially identical to that observed in normal rabbits. Rabbits genetically deficient in C6 also demonstrated LPS-induced hypotensive changes. Only minimal, or no changes in plasma C3 levels or serum CH50 values were detected in normal rabbits after LPS injection. Hypotensive changes were also induced in rabbits when complement was rapidly activated by intravenous injection of CoF. In contrast to the hypotension induced by LPS, the fall in arterial pressure associated with the consumption of complement was short lived and required the rapid consumption of considerable amounts of C3. The occurrence of DIC noted in normal rabbits injected with each preparation of LPS was not inhibited in either rabbits treated with cobra factor or in C6-deficient rabbits. The DIC was most pronounced after injection of Re595 and S. marcescens LPS. Injection of the various LPS preparations produced a rapid disappearance of circulating neutrophils and mononuclear cells, which occurred with the same kinetics and to the same extent in normal, CoF-treated, and C6-deficient rabbits. Injection of either Re595 LPS or S. marcescens LPS produced a biphasic disappearance of circulating 51Cr-platelets. In contrast, injection of 0111:B4 LPS affected only slightly the rate of disappearance of 51Cr-platelets. Depletion of C3 by cobra factor treatment had no effect on the disappearance of platelets in animals injected with 0111:B4. In marked contrast cobra factor treatment greatly reduced the initial rapid disappearance of platelets in rabbits injected with either Re595 or S. marcescens LPS, but had no effect in the secondary disappearance phase.


Assuntos
Proteínas do Sistema Complemento , Coagulação Intravascular Disseminada/imunologia , Hipotensão/imunologia , Lipopolissacarídeos , Animais , Plaquetas , Pressão Sanguínea , Feminino , Masculino , Polissacarídeos Bacterianos , Coelhos , Salmonella , Serratia marcescens , Venenos de Serpentes , Especificidade da Espécie
8.
J Exp Med ; 147(4): 973-83, 1978 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-206648

RESUMO

An acute phase of experimental autoimmune myasthenia gravis (EAMG) occurs transiently early in the immune response of Lewis rats to nicotinic acetylcholine receptors (AChR) when Bordetella pertussis is used as adjuvant. It is characterized by a destructive cellular attack directed at the postsynaptic membranes of muscle. Acute EAMG can be passively transferred to normal rats by IgG from serum of rats with chronic EAMG. In the present study, acute EAMG, induced either by passive transfer of syngeneic antibodies or by active immmunization, was inhibited in rats depleted of complement by treatment with cobra venom factor (CoF). Furthermore, passive transfer of antibodies in excess of the muscle's content of AChR was without any measurable effect in rats treated with CoF. Although 60% of the muscle's AChR was complexed with antibody, there was no reduction in the muscle's content of AChR, and neuromuscular transmission was not compromised as judged electromyographically by curare sensitivity. These data imply that redistribution, accelerated degradation, and impairment of the ionophore function of AChR, effects of antibodies described in vitro on extrajunctional AChR, do not play a significant role in vivo in impairing neuromuscular transmission in an intact neuromuscular junction. Complement appears to be a critical mediator of anti-AChR antibodies' pathogenicity in vivo.


Assuntos
Doenças Autoimunes/etiologia , Proteínas do Sistema Complemento/fisiologia , Miastenia Gravis/etiologia , Animais , Anticorpos , Complemento C3/fisiologia , Venenos Elapídicos/farmacologia , Imunização Passiva , Miastenia Gravis/imunologia , Junção Neuromuscular/imunologia , Ratos , Receptores Nicotínicos/imunologia , Membranas Sinápticas/imunologia , Transmissão Sináptica
9.
Science ; 265(5173): 808-11, 1994 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-7914033

RESUMO

Mammalian cells respond to endotoxic lipopolysaccharide (LPS) by activation of protein kinase cascades that lead to new gene expression. A protein kinase, p38, that was tyrosine phosphorylated in response to LPS, was cloned. The p38 enzyme and the product of the Saccharomyces cerevisiae HOG1 gene, which are both members of the mitogen-activated protein (MAP) kinase family, have sequences at and adjacent to critical phosphorylation sites that distinguish these proteins from most other MAP kinase family members. Both HOG1 and p38 are tyrosine phosphorylated after extracellular changes in osmolarity. These findings link a signaling pathway in mammalian cells with a pathway in yeast that is responsive to physiological stress.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/química , Endotoxinas/farmacologia , Proteínas Quinases Ativadas por Mitógeno , Proteínas de Saccharomyces cerevisiae , Equilíbrio Hidroeletrolítico/fisiologia , Sequência de Aminoácidos , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Linhagem Celular , Teste de Complementação Genética , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/enzimologia , Camundongos , Camundongos Endogâmicos C3H , Dados de Sequência Molecular , Pressão Osmótica , Paclitaxel/farmacologia , Saccharomyces cerevisiae/genética , Homologia de Sequência de Aminoácidos , Proteínas Quinases p38 Ativadas por Mitógeno
10.
Science ; 267(5198): 682-5, 1995 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-7839144

RESUMO

Mammalian mitogen-activated protein (MAP) kinases include extracellular signal-regulated protein kinase (ERK), c-Jun amino-terminal kinase (JNK), and p38 subgroups. These MAP kinase isoforms are activated by dual phosphorylation on threonine and tyrosine. Two human MAP kinase kinases (MKK3 and MKK4) were cloned that phosphorylate and activate p38 MAP kinase. These MKK isoforms did not activate the ERK subgroup of MAP kinases, but MKK4 did activate JNK. These data demonstrate that the activators of p38 (MKK3 and MKK4), JNK (MKK4), and ERK (MEK1 and MEK2) define independent MAP kinase signal transduction pathways.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , MAP Quinase Quinase 4 , Quinases de Proteína Quinase Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Clonagem Molecular , Ativação Enzimática , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , MAP Quinase Quinase 3 , Proteína Quinase 1 Ativada por Mitógeno , Dados de Sequência Molecular , Fosforilação , Proteínas Serina-Treonina Quinases/química , Proteínas Tirosina Quinases/química , Especificidade por Substrato , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno
11.
Science ; 249(4975): 1431-3, 1990 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-1698311

RESUMO

Leukocytes respond to lipopolysaccharide (LPS) at nanogram per milliliter concentrations with secretion of cytokines such as tumor necrosis factor-alpha (TNF-alpha). Excess secretion of TNF-alpha causes endotoxic shock, an often fatal complication of infection. LPS in the bloodstream rapidly binds to the serum protein, lipopolysaccharide binding protein (LBP), and cellular responses to physiological levels of LPS are dependent on LBP. CD14, a differentiation antigen of monocytes, was found to bind complexes of LPS and LBP, and blockade of CD14 with monoclonal antibodies prevented synthesis of TNF-alpha by whole blood incubated with LPS. Thus, LPS may induce responses by interacting with a soluble binding protein in serum that then binds the cell surface protein CD14.


Assuntos
Proteínas de Fase Aguda , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Proteínas de Transporte/imunologia , Lipopolissacarídeos/imunologia , Glicoproteínas de Membrana , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD18 , Eritrócitos/imunologia , Leucócitos/imunologia , Receptores de Lipopolissacarídeos , Macrófagos/imunologia , Receptores de Adesão de Leucócito/imunologia , Ovinos , Fator de Necrose Tumoral alfa/biossíntese
12.
Science ; 249(4975): 1429-31, 1990 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-2402637

RESUMO

The primary structure of lipopolysaccharide binding protein (LBP), a trace plasma protein that binds to the lipid A moiety of bacterial lipopolysaccharides (LPSs), was deduced by sequencing cloned complementary DNA. LBP shares sequence identity with another LPS binding protein found in granulocytes, bactericidal/permeability-increasing protein, and with cholesterol ester transport protein of the plasma. LBP may control the response to LPS under physiologic conditions by forming high-affinity complexes with LPS that bind to monocytes and macrophages, which then secrete tumor necrosis factor. The identification of this pathway for LPS-induced monocyte stimulation may aid in the development of treatments for diseases in which Gram-negative sepsis or endotoxemia are involved.


Assuntos
Proteínas de Fase Aguda , Proteínas Sanguíneas/genética , Proteínas de Transporte/genética , Lipopolissacarídeos/metabolismo , Glicoproteínas de Membrana , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas de Transporte/metabolismo , Biblioteca Gênica , Humanos , Cinética , Lipídeo A/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Dados de Sequência Molecular , Sondas de Oligonucleotídeos , Coelhos , Homologia de Sequência do Ácido Nucleico , Ovinos , Staphylococcus aureus , Fator de Necrose Tumoral alfa/biossíntese
13.
Cell Death Differ ; 14(4): 830-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17186025

RESUMO

Nod1 is a member of the NLR/Nod/CATERPILLER family. It acts as a sensor for intracellular bacteria by recognizing specific glycopeptides derived from peptidoglycan. Nod1 activation mediates distinct cellular responses including activation of MAP kinases, IL-8 release, apoptosis and suppression of several estrogen-dependent responses in MCF-7 cells. Here we have extended these studies by identifying key regulatory steps in Nod1-dependent signaling pathways. We provide multiple lines of data showing that Nod1-dependent apoptosis is a caspase 8-mediated event and that apoptosis requires RIP2. In contrast, several lines of evidence show that Nod1-dependent JNK activation and IL-8 production did not require the presence of caspase 8 but required activation of TAK1 as well as RIP2. Thus, we have identified several key control points that lie downstream of Nod1. This work provides the basis for further studies of the biological significance and regulation of the Nod1 pathway.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Apoptose/fisiologia , Neoplasias da Mama/genética , Caspase 8/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Interleucina-8/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Mutagênese Sítio-Dirigida , Mapeamento de Interação de Proteínas , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/metabolismo , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas
14.
J Clin Invest ; 62(6): 1313-24, 1978 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-372234

RESUMO

Normal rabbit serum reduces the buoyant density of lipopolysaccharide (LPS) from Escherichia coli 0111:B4 (d = 1.44 g/cm3) and Salmonella minnesota R595 (d = 1.38 g/cm3) to a value less than g/cm3. This density shift is associated with the inhibition of a number of endotoxic activities of the LPS; namely, the pyrogenic activity, the ability to produce an immediate neutropenia in rabbits, lethality in adrenalectomized mice, and anticomplementary activity. A qualitatively similar change in buoyant density was observed to occur after intravenous injection of the LPS into rabbits. Preliminary evidence suggests that the density shift does not occur as a result of the degradation of the glycolipid backbone of the LPS. These data suggest that the interactions of LPS with plasma (or serum) components leading to reduction in buoyant density may account for a major pathway of LPS detoxification.


Assuntos
Biotransformação , Sangue , Endotoxinas , Inativação Metabólica , Lipopolissacarídeos , Animais , Proteínas do Sistema Complemento , Escherichia coli , Lipopolissacarídeos/sangue , Lipopolissacarídeos/isolamento & purificação , Lipopolissacarídeos/farmacologia , Coelhos , Salmonella , Ultracentrifugação
15.
J Clin Invest ; 81(6): 1925-37, 1988 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-3384955

RESUMO

Macrophages are induced by LPS to release a number of products that determine the host response during gram negative sepsis. To examine the role of one such substance, tumor necrosis factor (TNF), in mediating LPS-induced injury, we employed a rabbit model of endotoxic shock to (a) determine the kinetics and extent of release of TNF into plasma after injection of LPS, and (b) to evaluate the protective effect of in vivo neutralization of LPS-induced TNF by prior infusion of anti-TNF antibody. TNF was maximally induced 45-100 min after injection of 10 micrograms i.v. parent Salmonella minnesota Re595 LPS or 250 micrograms Re595 LPS-HDL complexes. Maximal induction of TNF by LPS was associated with development of hypotension, focal hepatic necrosis, intravascular fibrin deposition and lethality. Based on (a) the peak levels of TNF observed in serum, 2.5 X 10(3) U/ml, (b) the specific activity of purified rabbit macrophage-derived TNF, 1 X 10(8) U/mg, and (c) the biphasic disappearance of intravenously injected purified TNF (t1/2 = 0.5 min, 11 min) we constructed a kinetic model showing that at least 130 micrograms of TNF (1.3 X 10(7) U) was released into plasma 30-200 min postinjection of LPS. Prior infusion of anti-TNF antibody (30-45 min before LPS injection) resulted in neutralization of the LPS-induced serum TNF activity and provided significant protection from the development of hypotension, fibrin deposition, and lethality. Thus, these results provide further evidence that TNF plays a central role mediating the pathophysiologic changes that occur during gram negative endotoxic shock.


Assuntos
Bactérias Gram-Negativas , Lipopolissacarídeos/toxicidade , Choque Séptico/metabolismo , Fator de Necrose Tumoral alfa/sangue , Animais , Modelos Animais de Doenças , Meia-Vida , Injeções Intravenosas , Cinética , Lipopolissacarídeos/administração & dosagem , Macrófagos/metabolismo , Masculino , Coelhos , Fator de Necrose Tumoral alfa/metabolismo
16.
J Clin Invest ; 64(5): 1516-24, 1979 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-227936

RESUMO

The addition of bacterial lipopolysaccharide (LPS) from Escherichia coli 0111:B4 or Salmonella minnesota R595 to plasma (or serum) resulted in a marked reduction of the hydrated buoyant density of the parent LPS (0111:B4 [d = 1.44 g/cm3] and R595 [d = 1.38 g/cm3]), to d less than 1.2 g/cm3. This reduction in buoyant density to less than 1.2 g/cm3 of the LPS required plasma (or serum) lipid. Delipidation of plasma (or serum) by extraction with n-butanol/diisopropyl ether (40/60, vol:vol) prevented the conversion of the parent LPS to a form with d less than 1.2 g/cm3. Reversal of the effect of delipidation was accomplished by the addition of physiologic concentrations of high density lipoprotein (HDL). In contrast, as much as two times normal serum concentration of low density or very low density lipoprotein were ineffective. The ability of normal plasma (or serum) to inhibit the pyrogenic activity of LPS, lost after delipidation, was also restored after the addition of HDL. Preliminary results suggested that prior modifications of the LPS, probably disaggregation, may be required before interaction with HDL.


Assuntos
Antitoxinas/imunologia , Atividade Bactericida do Sangue , Endotoxinas/antagonistas & inibidores , Escherichia coli , Lipopolissacarídeos/antagonistas & inibidores , Lipoproteínas HDL/imunologia , Polissacarídeos Bacterianos/antagonistas & inibidores , Salmonella , Animais , Butanóis , Coagulação Intravascular Disseminada/etiologia , Coagulação Intravascular Disseminada/prevenção & controle , Éteres , Febre/etiologia , Febre/prevenção & controle , Humanos , Lipoproteínas HDL/sangue , Lipoproteínas LDL/imunologia , Masculino , Coelhos , Sepse/etiologia , Sepse/prevenção & controle
17.
J Clin Invest ; 67(3): 827-37, 1981 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7204557

RESUMO

The addition of bacterial lipopolysaccharide (LPS) from Salmonella minnesota R595 to rabbit plasma results in a marked reduction of the hydrated buoyant density of the parent R595 LPS, from 1.38 to less than 1.2 g/cm3. Using immunopurified anti-R595 LPS antibody covalently linked to Sepharose 4B, we were able to separate the altered R595 LPS (d less than 1.2 g/cm3) from the remainder of the plasma proteins by elution of the bound material with 2.5 M KSCN. The KSCN eluate was shown to have a d less than 1.2 g/cm3 and to contain both R595 LPS as well as protein and lipid characteristic of high density lipoprotein (HDL). The major protein in the KSCN eluate is a single polypeptide chain with an apparent molecular weight of 26,000 in sodium dodecyl sulfate and an amino acid composition essentially identical to that of apoprotein AI, the major protein of rabbit HDL. The lipid composition of the KSCN eluate is similar to that of HDL, although marked differences in the cholesterol ester/cholesterol ration and the phosphatidyl choline/phosphatidyl ethanolamine ratio were observed when the KSCN eluate and rabbit HDL were compared. The formation of this R595 LPS-protein-lipid complex in plasma accounts for the marked reduction in buoyant density found when LPS is added to plasma.


Assuntos
Lipopolissacarídeos/sangue , Lipoproteínas HDL/fisiologia , Aminoácidos/análise , Animais , Fenômenos Químicos , Físico-Química , Eletroforese em Gel de Poliacrilamida , Lipídeos/análise , Peso Molecular , Ligação Proteica , Coelhos , Salmonella
18.
J Clin Invest ; 92(4): 2053-9, 1993 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7691891

RESUMO

Exposure of rabbit peritoneal exudate macrophages (PEM) or whole blood to picomolar concentrations of LPS induces adaptation or hyporesponsiveness to LPS. Because of the importance of plasma LPS-binding protein (LBP) and the macrophage cell membrane protein CD14 in recognition of LPS, we examined the effect of LBP on LPS-induced adaptation in PEM. PEM exposed to LPS in the presence of LBP for 8 h were markedly less responsive to subsequent stimulation by LPS than monocytes/macrophages (M phi) adapted in the absence of LBP. LPS-induced expression of TNF was sharply reduced in LBP-LPS-adapted PEM, but in contrast these cells remained fully responsive to Staphylococcus aureus peptidoglycan. We considered that specific hyporesponsiveness in LPS-adapted M phi or in blood monocytes could be due to decreased expression of CD14 or diminished binding of LBP-LPS complexes to CD14. However, flow cytometry analysis revealed only minimal reduction of CD14 expression or CD14-dependent binding of a fluorescent LPS derivative when normo- and hyporesponsive cells were compared. These results show that complexes of LPS and LBP are more effective than LPS alone in inducing adaptation to LPS, and LPS-induced hyporesponsiveness probably results from changes in cellular elements distinct from CD14 that are involved in either LPS recognition or LPS-specific signal transduction.


Assuntos
Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Proteínas de Transporte/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Glicoproteínas de Membrana , Aclimatação , Proteínas de Fase Aguda/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Clonais , Cinética , Receptores de Lipopolissacarídeos , Lipopolissacarídeos/metabolismo , Macrófagos Peritoneais/metabolismo , Masculino , Coelhos , Proteínas Recombinantes/farmacologia , Salmonella , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/farmacologia
19.
J Clin Invest ; 98(7): 1533-8, 1996 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-8833900

RESUMO

Lipopolysaccharide (LPS), residing in the outer membrane of all gram-negative bacteria, is considered a major initiating factor of the gram-negative septic shock syndrome in humans. LPS forms a complex with the LPS binding protein (LBP) in plasma, and LPS-LBP complexes engage a specific receptor, CD14, on the surface of myeloid cells, leading to the production of potent proinflammatory cytokines. The major goal of this study was to test the importance of the CD14 pathway in vivo in a primate model that is similar to human septic shock. Primates were pretreated with one of two different inhibitory anti-CD14 mAbs, then challenged with intravenous endotoxin (375 microg/kg/h) for 8 h. The anti-CD14 treatment regimens were successful in preventing profound hypotension, reducing plasma cytokine levels (TNF-alpha, IL-1beta, IL-6, and IL-8), and inhibiting the alteration in lung epithelial permeability that occurred in animals treated with LPS and an isotype-matched control antibody. These results demonstrate for the first time the importance of the CD14 pathway in a primate model that is similar to human septic shock. Inhibition of the CD14 pathway represents a novel therapeutic approach to treating this life-threatening condition.


Assuntos
Anticorpos/uso terapêutico , Receptores de Lipopolissacarídeos/imunologia , Síndrome do Desconforto Respiratório/prevenção & controle , Choque Séptico/prevenção & controle , Doença Aguda , Animais , Pressão Sanguínea , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Débito Cardíaco , Modelos Animais de Doenças , Interleucinas/análise , Macaca fascicularis , Permeabilidade/efeitos dos fármacos , Albumina Sérica/análise , Fator de Necrose Tumoral alfa/análise
20.
J Clin Invest ; 102(9): 1645-52, 1998 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9802878

RESUMO

Recognition of bacterial endotoxin (LPS) elicits multiple host responses, including activation of cells of the innate immune system. LPS exposure occurs repeatedly during septicemia, making strict regulation of gene expression necessary. Such regulation might prevent, for example, the continuous production of proinflammatory cytokines such as tumor necrosis factor (TNF), which could lead to severe vascular collapse. Tolerance to LPS is characterized by a diminished production of TNF during prolonged exposure to LPS, and is therefore likely to represent an essential control mechanism during sepsis. In the present study, which uses mice with genetic deletions of the proteins of NF-kappaB complex, we provide data demonstrating that increased expression of the p50 subunit of NF-kappaB directly results in the downregulation of LPS-induced TNF production. This contention is supported by the following observations: (1) tolerance to LPS is not induced in macrophages from p50-/- mice; (2) long-term pretreatment with LPS does not block synthesis of TNF mRNA in p50-/- macrophages (in contrast to wild-type macrophages); (3) ectopic overexpression of p50 reduces transcriptional activation of the murine TNF promoter; and (4) analysis of the four kappaB sites from the murine TNF promoter demonstrates that binding of p50 homodimers to the positively acting kappaB3 element is associated with development of the LPS-tolerant phenotype. Thus, p50 expression plays a key role in the development of LPS tolerance.


Assuntos
Imunidade Inata/fisiologia , Macrófagos Peritoneais/imunologia , NF-kappa B/imunologia , Animais , Células Cultivadas , Regulação para Baixo , Tolerância Imunológica , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Camundongos , Camundongos Knockout , Subunidade p50 de NF-kappa B , Regiões Promotoras Genéticas , RNA Mensageiro , Fator de Necrose Tumoral alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA