Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Infect Immun ; 75(8): 3979-88, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17517872

ABSTRACT

The pathogenicity of Mycobacterium ulcerans, the agent of Buruli ulcer, depends on the cytotoxic exotoxin mycolactone. Little is known about the immune response to this pathogen. Following the demonstration of an intracellular growth phase in the life cycle of M. ulcerans, we investigated the production of tumor necrosis factor (TNF) induced by intramacrophage bacilli of diverse toxigenesis/virulence, as well as the biological relevance of TNF during M. ulcerans experimental infections. Our data show that murine bone marrow-derived macrophages infected with mycolactone-negative strains of M. ulcerans (nonvirulent) produce high amounts of TNF, while macrophages infected with mycolactone-positive strains of intermediate or high virulence produce intermediate or low amounts of TNF, respectively. These results are in accordance with the finding that TNF receptor P55-deficient (TNF-P55 KO) mice are not more susceptible than wild-type mice to infection by the highly virulent strains but are more susceptible to nonvirulent and intermediately virulent strains, demonstrating that TNF is required to control the proliferation of these strains in animals experimentally infected by M. ulcerans. We also show that mycolactone produced by intramacrophage M. ulcerans bacilli inhibits, in a dose-dependent manner, but does not abrogate, the production of macrophage inflammatory protein 2, which is consistent with the persistent inflammatory responses observed in experimentally infected mice.


Subject(s)
Bacterial Toxins/immunology , Macrophages/microbiology , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium ulcerans/immunology , Mycobacterium ulcerans/pathogenicity , Tumor Necrosis Factor-alpha/immunology , Animals , Cells, Cultured , Chemokine CXCL2 , Disease Models, Animal , Female , Foot/pathology , Macrolides , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monokines/antagonists & inhibitors , Monokines/biosynthesis , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium ulcerans/growth & development , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor Decoy Receptors/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Virulence
2.
World J Gastroenterol ; 12(6): 858-67, 2006 Feb 14.
Article in English | MEDLINE | ID: mdl-16521212

ABSTRACT

AIM: To study the role of macrophage inflammatory protein (MIP)-2 in liver resection-induced acceleration of tumor growth in a mouse model of hepatic metastasis. METHODS: After a 50% hepatectomy, 1x10(5) CT26.WT cells were implanted into the left liver lobe of syngeneic balb/c mice (PHx). Additional animals were treated with a monoclonal antibody (MAB452) neutralizing MIP-2 (PHx+mAB). Non-resected and non-mAB-treated mice (Con) served as controls. After 7 d, tumor angiogenesis and microcirculation as well as cell proliferation, tumor growth, and CXCR-2 expression were analyzed using intravital fluorescence microscopy, histology, immunohistochemistry, and flow cytometry. RESULTS: Partial hepatectomy increased (P<0.05) the expression of the MIP-2 receptor CXCR-2 on tumor cells when compared with non-resected controls, and markedly accelerated (P<0.05) angiogenesis and metastatic tumor growth. Neutralization of MIP-2 by MAB452 treatment significantly (P<0.05) depressed CXCR-2 expression. Further, the blockade of MIP-2 reduced the angiogenic response (P<0.05) and inhibited tumor growth (P<0.05). Of interest, liver resection-induced hepatocyte proliferation was not effected by anti-MIP-2 treatment. CONCLUSION: MIP-2 significantly contributes to liver resection-induced acceleration of colorectal CT26.WT hepatic metastasis growth.


Subject(s)
Liver Neoplasms/pathology , Liver Neoplasms/secondary , Liver Neoplasms/surgery , Monokines/physiology , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Adenocarcinoma/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Cell Division , Chemokine CXCL2 , Colorectal Neoplasms/pathology , Disease Models, Animal , Disease Progression , Flow Cytometry , Liver Neoplasms/therapy , Mice , Mice, Inbred BALB C , Microcirculation/pathology , Monokines/antagonists & inhibitors , Monokines/therapeutic use , Neovascularization, Pathologic/physiopathology
3.
J Leukoc Biol ; 78(3): 696-704, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15944212

ABSTRACT

Angiotensin-II (Ang-II) has inflammatory activity and is involved in different diseases associated with the cardiovascular system. This study has evaluated the effect of boldine (B), and two phenanthrene alkaloids semisynthesized by us, secoboldine (SB) and boldine methine (BM), on Ang-II-induced neutrophil recruitment. Intraperitoneal administration of 1 nM Ang-II induced significant neutrophil accumulation, which was maximal at 4-8 h. BM inhibited neutrophil infiltration into the peritoneal cavity at 4 h and 8 h by 73% and 77%, respectively, SB at 8 h by 55%, and B had no effect on this response. Although BM inhibited the release of cytokine-inducible neutrophil chemoattractant/keratinocyte-derived chemokine, macrophage inflammatory protein-2 (MIP-2), and platelet-activating factor (PAF) elicited by Ang-II, SB only reduced the release of MIP-2 after 4 h of its administration. Sixty-minute superfusion of the rat mesentery with 1 nM Ang-II induced a significant increase in the leukocyte-endothelial cell interactions and P-selectin up-regulation, which were inhibited by 1 microM BM and SB. The generation of reactive oxygen species (ROS) in endothelial cells stimulated with Ang-II was inhibited significantly by the three alkaloids tested. BM also diminished Ang-II-induced interleukin-8 release from endothelial cells and blocked the PAF receptor on human neutrophils (concentration of the compound needed to produce 50% inhibition value: 28.2 microM). Therefore, BM is a potent inhibitor of Ang-II-induced neutrophil accumulation in vivo. This effect appears to be mediated through inhibition of CXC chemokine and PAF release, ROS scavenging activity, and blockade of the PAF receptor. Thus, it may have potential therapeutic interest for the control of neutrophil recruitment that occurs in inflammation associated with elevated levels of Ang-II.


Subject(s)
Angiotensin II/administration & dosage , Aporphines/pharmacology , Neutrophils/drug effects , Phenanthrenes/pharmacology , Angiotensin II/antagonists & inhibitors , Animals , Chemokine CXCL2 , Chemokines/antagonists & inhibitors , Chemokines/biosynthesis , Chemokines/immunology , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/biosynthesis , Endothelial Cells/drug effects , Endothelial Cells/immunology , Humans , Infusions, Parenteral , Intercellular Signaling Peptides and Proteins/biosynthesis , Interleukin-8/biosynthesis , Interleukin-8/immunology , Keratinocytes/immunology , Male , Molecular Structure , Monokines/antagonists & inhibitors , Monokines/biosynthesis , Monokines/immunology , Neutrophils/immunology , P-Selectin/drug effects , P-Selectin/immunology , Platelet Activating Factor/antagonists & inhibitors , Platelet Activating Factor/biosynthesis , Platelet Activating Factor/immunology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/immunology
4.
Cytokine ; 31(3): 169-79, 2005 Aug 07.
Article in English | MEDLINE | ID: mdl-15961314

ABSTRACT

Neutrophil associated lung injury is identified with a variety of local and systemic priming insults. In vitro studies have shown that TNF-alpha mediated suppression of neutrophil apoptosis is due to the secretion of interleukin-8 (IL-8), a human chemokine shown to alter neutrophil chemotaxis. Our initial in vitro antibody neutralization studies with neutrophil chemotactic proteins, keratinocyte-derived chemokine (KC) and macrophage inflammatory protein-2alpha (MIP-2alpha), mouse IL-8 homologues, indicate that MIP-2alpha but not KC appears to mediate TNF-alpha suppression of mouse neutrophil apoptosis. Therefore, we hypothesized that in vivo neutralization of KC or MIP-2alpha during an initial priming insult would produce differential effects on the extent of lung injury by restoring normal neutrophil apoptotic function. To assess this, mice were hemorrhaged followed with septic challenge at 24 h. Antibody against KC or MIP-2alpha or a nonspecific IgG was given during resuscitation immediately following hemorrhage. Anti-MIP-2alpha treatment resulted in a significant reduction in lung tissue IL-6 and myeloperoxidase levels. Percentage of neutrophil apoptosis increased significantly in the anti-KC group. Tissue and plasma KC and MIP-2alpha were reduced in their respective treatment groups. These data suggest that KC and MIP-2alpha differ in their mediation of neutrophil function (apoptosis and chemotaxis) and contribution to the pathogenesis of lung injury following hemorrhage subsequent to sepsis.


Subject(s)
Cytokines/physiology , Hemorrhage/complications , Lung/metabolism , Lung/physiopathology , Monokines/physiology , Neutrophils/pathology , Acute Disease , Animals , Antibodies/pharmacology , Apoptosis/physiology , Cells, Cultured , Chemokine CXCL1 , Chemokine CXCL2 , Chemokines , Chemokines, CXC , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Disease Models, Animal , Hemorrhage/physiopathology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Lung/pathology , Male , Mice , Mice, Inbred C3H , Monokines/antagonists & inhibitors , Monokines/biosynthesis , Monokines/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Sepsis/metabolism , Sepsis/pathology , Sepsis/physiopathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology
5.
J Cell Physiol ; 204(3): 889-95, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15828023

ABSTRACT

The immune system of vertebrates detects bacterial DNA as a "danger signal" based on the presence of unmethylated CpG motifs. We examined whether oligodeoxynucleotides (ODNs) with CpG motifs (CpG-ODNs) also induce mobilization of hematopoietic progenitor cells (HPCs). Mice challenged with CpG-ODNs showed an increase in peripheral blood colony-forming units (CFU) with a peak at day 4 after treatment, associated with an increase, starting 30 min after CpG treatment, in serum levels of mouse keratinocyte-derived chemokine (mKC), a functional homolog of human interleukin (IL) 8; production of granulocyte-colony-stimulating factor (CSF) was also detected. Mobilization and mKC induction were sequence-specific and dose-dependent occurring even with low doses of CpG-ODNs. Interestingly, intestinal cells were involved in mKC production. HPC mobilization by CpG-ODNs was dependent on peripheral blood mononuclear cells since mobilization was reduced in neutrophil-depleted mice. Moreover, CpG-ODN treatment significantly increased G-CSF mobilizing capacity. Finally, pretreatment with an anti-mKC neutralizing antibody significantly reduced CpG-induced mobilization, further supporting a role for mKC. Thus, bacterial DNA is a "danger signal" not only for immune cells but also for hematopoietic cells, communicating the need for increased hematopoiesis during infections and for the renewal of the immune system. The HPC mobilization activity of CpG-ODNs will need to be considered in the design of treatment regimens for cancer clinical trials using CpG-ODNs in association with chemotherapy.


Subject(s)
Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Interleukin-8/biosynthesis , Oligodeoxyribonucleotides/pharmacology , Animals , Cell Movement/drug effects , Chemokine CXCL2 , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/metabolism , Interleukin-8/antagonists & inhibitors , Interleukin-8/blood , Keratinocytes/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/physiology , Mice , Mice, Inbred C57BL , Monokines/antagonists & inhibitors , Monokines/metabolism
6.
Int J Immunopathol Pharmacol ; 18(1): 85-94, 2005.
Article in English | MEDLINE | ID: mdl-15698514

ABSTRACT

Mimosine is a plant amino-acid which has been reported to block DNA replication in mammalian cells and to arrest cell reversibly towards the end of the G1 phase or at the beginning of the S phase. In this study, 42 mice were infected with T. spiralis a nematode parasite, and treated with the anti-inflammatory compound L-mimosine, to determine if any alteration in the chronic inflammatory state occurred, by investigating the hosts immunological response. MCP-1, a C-C chemokine and MIP-2, a C-X-C chemokine were tested and calculated in the sera of infected animals, after 1, 10, 20, 30, 40, 50 and 60 days post infection, by ELISA method. The diaphragm and the masseters of the infected mice, were tested for inflammatory response. Here we found, that MCP-1 was partially inhibited by L-mimosine, while MIP-2 was totally inhibited. Moreover in sections of the diaphragm and masseters, the infiltration of inflammatory cells, such as macrophages, lymphocytes and eosinophils were more intense in untreated animals compared to those treated with L-mimosine. These findings show, that L-mimosine may have an inhibitory effect on MCP-1 and MIP-2 serum levels in Trichinellosis and may influence the recruitment of inflammatory cells and the intensity of the inflammatory reaction in this parasitic disease.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Mimosine/pharmacology , Monokines/antagonists & inhibitors , Receptors, Chemokine/antagonists & inhibitors , Trichinellosis/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Chemokine CXCL2 , Cysts/parasitology , Diaphragm/pathology , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Mimosine/administration & dosage , Monokines/metabolism , Receptors, CCR2 , Receptors, Chemokine/metabolism , Trichinella spiralis , Trichinellosis/parasitology , Trichinellosis/pathology
7.
Am J Physiol Gastrointest Liver Physiol ; 287(6): G1220-6, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15319184

ABSTRACT

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.


Subject(s)
Inflammation Mediators/pharmacology , Inflammation/pathology , Inflammatory Bowel Diseases/chemically induced , Monokines/biosynthesis , Monokines/genetics , Platelet Activating Factor/pharmacology , Animals , Antibodies, Blocking/pharmacology , Chemokine CXCL2 , Chemokines/pharmacology , Chemokines, CXC/biosynthesis , Chemokines, CXC/genetics , Gene Expression Regulation/drug effects , Hematocrit , Hypotension/chemically induced , Hypotension/physiopathology , Ileum/drug effects , Ileum/metabolism , Ileum/pathology , Inflammation/chemically induced , Inflammation Mediators/antagonists & inhibitors , Inflammatory Bowel Diseases/pathology , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Leukocyte Count , Leukopenia/chemically induced , Male , Monokines/antagonists & inhibitors , Neutrophil Infiltration/drug effects , Peroxidase/biosynthesis , Platelet Activating Factor/antagonists & inhibitors , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
8.
J Pept Res ; 63(1): 36-47, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14984572

ABSTRACT

The CCL15 is a human CC chemokine that activates the receptors, CCR1 and CCR3. Unlike other chemokines, it contains an unusually long N-terminal domain of 31 amino acids preceding the first cysteine residue and a third disulfide bond. To elucidate the functional role of distinct structural determinants, a series of sequential amino-terminal truncated and point-mutated CCL15 derivatives as well as mutants lacking the third disulfide bond and the carboxy-terminal alpha-helix were synthesized using 9-fluorenylmethoxycarbonyl (Fmoc) chemistry. We demonstrate that a truncation of 24 amino acid residues (delta24-CCL15) converts the slightly active 92-residue delta0-CCL15 into a potent agonist of CC chemokine receptor 1 (CCR1) and a weak agonist of CCR3 in cell-based assays. The biological activity decreases from delta24-CCL15 to delta29-CCL15, and re-increases from delta29-CCL15 to delta30-CCL15. Thus, an exocyclic N-terminal region of only one amino acid residue is sufficient for efficient CCR1 activation. As none of the peptides investigated except for delta24-CCL15 activates CCR3, we suggest that CCR1 is the major receptor for CCL15 in vivo. Further we demonstrate that the third disulfide bond of CCL15 and an exchange of tyrosine in position 70 by a leucine residue, which is conserved in CXC chemokines, do not alter the interaction with CCR1. In contrast, a CCL15 derivative lacking the carboxy-terminal alpha-helix exhibits a complete loss of tertiary structure and hence loss of CCR1 agonistic and binding activity. This study demonstrates that specific protein residues in chemokines, which contribute to receptor-ligand interaction, vary significantly between chemokines and cannot be extrapolated using data from functionally related chemokines.


Subject(s)
Monokines/chemistry , Peptides/chemical synthesis , Peptides/pharmacology , Receptors, Chemokine/agonists , Amino Acid Sequence , Animals , Binding, Competitive , CHO Cells , Calcium/metabolism , Cell Line, Tumor , Chemokines, CC , Chemotaxis, Leukocyte/drug effects , Cricetinae , Cricetulus , Heparin/metabolism , Humans , Ligands , Macrophage Inflammatory Proteins , Molecular Sequence Data , Monocytes/immunology , Monokines/antagonists & inhibitors , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemistry , Protein Structure, Tertiary , Radioligand Assay , Receptors, CCR1 , Receptors, Chemokine/chemistry , Receptors, Chemokine/metabolism , Structure-Activity Relationship
9.
J Immunol ; 170(9): 4665-72, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12707345

ABSTRACT

We have previously demonstrated that administration of the recently described cytokine IL-17 in rat airways in vivo recruits and activates neutrophils locally. In the current study, we examined whether endogenous IL-17 is involved in mediating neutrophil recruitment caused by endotoxin exposure in mouse airways. Our in vivo data show that local endotoxin exposure causes the release of free, soluble IL-17 protein 6 h later. Systemic pretreatment with a neutralizing anti-IL-17 Ab almost completely inhibits neutrophil recruitment 24 h, but not 6 h, after endotoxin exposure in the airways. Pretreatment with neutralizing anti-IL-6 and anti-macrophage inflammatory protein (MIP)-2 Abs inhibits neutrophil recruitment caused by local endotoxin exposure and IL-17, respectively. Our in vitro data show that endotoxin exposure stimulates the release of soluble IL-17 protein in T lymphocytes harvested from lung and spleen, respectively, and that this cytokine release requires coculture with airway macrophages. Intracellular IL-17 protein is detected in T lymphocytes from spleen but not in airway macrophages after coculture and stimulation of these two cell types. Finally, anti-IL-17 does not alter endotoxin-induced release of IL-6 and MIP-2 from T lymphocytes and airway macrophages in coculture. In conclusion, our results indicate that endotoxin exposure causes the release of IL-17 from T lymphocytes and that this cytokine release requires the presence of macrophages. Once released, endogenous IL-17 acts in part by inducing local release of neutrophil-mobilizing cytokines such as IL-6 and MIP-2, from nonlymphocyte, nonmacrophage cells, and this contributes to recruitment of neutrophils in the airways. These IL-17-related mechanisms constitute potential targets for pharmacotherapy against exaggerated neutrophil recruitment in airway disease.


Subject(s)
Inflammation Mediators/physiology , Interleukin-17/physiology , Lipopolysaccharides/administration & dosage , Lung/immunology , Neutrophil Infiltration/immunology , Administration, Intranasal , Animals , Antibodies, Monoclonal/administration & dosage , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Chemokine CXCL2 , Dose-Response Relationship, Immunologic , Inflammation/blood , Inflammation/immunology , Inflammation Mediators/analysis , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/immunology , Injections, Intraperitoneal , Interleukin-17/analysis , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Monokines/antagonists & inhibitors , Monokines/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Solubility , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
10.
Circulation ; 103(18): 2296-302, 2001 May 08.
Article in English | MEDLINE | ID: mdl-11342480

ABSTRACT

BACKGROUND: Mechanisms by which neutrophils are attracted to the myocardium in ischemia/reperfusion are not fully defined. Lipopolysaccharide-induced CXC chemokine (LIX), cytokine-induced neutrophil chemoattractant (KC), and macrophage inflammatory protein-2 (MIP-2) are rodent chemokines with potent neutrophil-chemotactic activity. The goals of the present study were to evaluate the roles of these chemokines in a rat model of ischemia/reperfusion and to examine the mechanisms of chemokine induction by oxidative stress and cytokines in cultured cardiomyocytes. METHODS AND RESULTS: Male Wistar-Kyoto rats underwent 45 minutes of ligation of the left anterior descending coronary artery, followed by reperfusion for various periods. Compared with sham-operated controls, myocardium from reperfused animals had higher levels of free radicals, increased neutrophil infiltration evidenced histologically and by elevated myeloperoxidase activity, and increased nuclear factor (NF)-kappaB DNA binding activity. Ischemia-reperfusion also induced the expression of interleukin-1beta, tumor necrosis factor (TNF)-alpha, LIX, KC, and MIP-2 mRNA and protein. LIX expression was localized to resident myocardial cells, whereas KC and MIP-2 were expressed only in infiltrating inflammatory cells. Neutralization of LIX inhibited 79% of neutrophil infiltration into previously ischemic myocardium. In contrast, neutralization of KC and MIP-2 reduced neutrophil infiltration by only 28% and 37%, respectively. In cultured cardiomyocytes, LIX expression was induced by oxidative stress or TNF-alpha and was blocked by the NF-kappaB inhibitor pyrrolidinedithiocarbamate. CONCLUSIONS: LIX is expressed by resident myocardial cells during ischemia-reperfusion and is induced in cultured cardiomyocytes by oxidative stress or TNF-alpha via NF-kappaB activation. Although KC and MIP-2 are expressed by inflammatory cells infiltrating the myocardium during reperfusion after ischemia, neutrophil recruitment to reperfused rat myocardium is mainly due to cardiomyocyte expression of LIX.


Subject(s)
Chemokines, CXC/metabolism , Intercellular Signaling Peptides and Proteins , Myocardium/metabolism , NF-kappa B/metabolism , Neutrophil Infiltration , Reperfusion Injury/metabolism , Animals , Antibodies/pharmacology , Cells, Cultured , Chemokine CXCL2 , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/genetics , Chemotactic Factors/antagonists & inhibitors , Chemotactic Factors/genetics , Chemotactic Factors/metabolism , Disease Models, Animal , Free Radicals/metabolism , Gene Expression/drug effects , Growth Substances/genetics , Growth Substances/metabolism , Hydrogen Peroxide/pharmacology , Immunohistochemistry , Interleukin-1/genetics , Interleukin-1/metabolism , Male , Monokines/antagonists & inhibitors , Monokines/genetics , Monokines/metabolism , Myocardial Ischemia/immunology , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardium/immunology , Myocardium/pathology , Neutrophil Infiltration/immunology , RNA, Messenger/metabolism , Rats , Rats, Inbred WKY , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
11.
Am J Respir Crit Care Med ; 162(6): 2316-23, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11112157

ABSTRACT

Hyperoxia may contribute to lung disease in newborns through effects on alveolar neutrophils which predominate in respiratory distress syndrome and other acute lung injuries. Neutrophil chemokines such as interleukin-8 (IL-8) regulate chemoattraction, and are elevated in tracheal aspirates of newborns who develop bronchopulmonary dysplasia (BPD). Blockade of neutrophil chemokines may reduce hyperoxia-induced inflammatory lung injury and BPD. We therefore tested the hypothesis that hyperoxia contributes to elevations of rat neutrophil chemokines, cytokine-induced neutrophil chemoattractant-1 (CINC-1), and macrophage inflammatory protein-2 (MIP-2) in newborn rat lung. Newborn rats were exposed to air or 95% O(2) for 8 d. CINC-1 and MIP-2 were measured in whole lung homogenates by ELISA. Newborn 95% O(2)-exposed animals were given anti-CINC-1 or anti-MIP-2, 1, 5, or 10 microg on Days 3 and 4 of 95% O(2) exposure. Bronchoalveolar lavage (BAL) was performed after perfusion on day 6 to evaluate airway neutrophils, and myeloperoxidase (MPO) was measured in perfused whole lung. Lungs were examined histologically and immunohistochemically for effects of 95% O(2) +/- antichemokine. CINC-1 and MIP-2 increased nearly tenfold by Day 8 95% O(2) treatment versus air control. CINC-1 and MIP-2 immunolabeling was increased in alveolar macrophages and alveolar epithelium in 95% O(2). Anti-CINC-1 and anti-MIP-2 treatment at every dose reduced neutrophil number > 90% in BAL. Anti-CINC-1 10 microg reduced tissue MPO by 50%. Antichemokine treatment on days 3 and 4 prevented alveolar septal thickening and reduced chemokine immunolabeling on Day 6. Hyperoxia-induced neutrophil influx is mediated in part by CINC-1 and MIP-2 in newborn rats and can be partially prevented by treatment with anti-CINC-1 and anti-MIP-2.


Subject(s)
Chemokines, CXC/antagonists & inhibitors , Chemotactic Factors/antagonists & inhibitors , Hyperoxia/drug therapy , Intercellular Signaling Peptides and Proteins , Monokines/antagonists & inhibitors , Pneumonia/prevention & control , Acute Disease , Analysis of Variance , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/prevention & control , Chemokine CXCL1 , Chemokine CXCL2 , Chemokines, CXC/metabolism , Chemotactic Factors/metabolism , Dose-Response Relationship, Drug , Growth Substances/metabolism , Humans , Hyperoxia/complications , Hyperoxia/metabolism , Immunohistochemistry , Infant, Newborn , Lung/chemistry , Lung/metabolism , Lung/pathology , Monokines/metabolism , Neutrophils/metabolism , Pneumonia/etiology , Pneumonia/metabolism , Random Allocation , Rats , Time Factors
12.
Sheng Li Xue Bao ; 52(1): 50-4, 2000 Feb.
Article in Chinese | MEDLINE | ID: mdl-11971171

ABSTRACT

Two-microelectrode voltage clamp (TEV) method was used to study the blocking effects of extracellular Ba(2+) on the inward rectifier potassium channel (IRK1) expressed in Xenopus oocytes. Blockage of Ba(2+) on IRK1 (1 ms after voltage applied) is Ba(2+) concentration (0,1,3,10 or 100 micromol/L) dependent with 10 or 90 mmol/L potassium and also voltage-dependent. Ba(2+) almost has no effect on the open/close of IRK1. IRK1 is not permeable to Ba(2+). Three exponential fitting analysis indicates that Ba(2+) and K(+) compete the same binding site in IRK1 when external Ba(2+) concentration is lower (1 or 3 micromol/L). The time constant of IRK1 does not increase, but the concentration dependency of the weights of the fittings increases with the increase of external Ba(2+) concentration. As a result, the inactivation becomes faster and faster as the external Ba(2+) concentration increases. Moreover, since the time constant of the channel decreases and the weights of the fittings concentration dependently increase with the increase of external Ba(2+) concentration (10 or 100 micromol/L), the inactivation becomes faster and faster. It is demonstrated that Ba(2+) can contact with deeper binding sites in IRK1 as external Ba(2+) concentration increases. It is suggested that two different mechanisms may underlie the external Ba(2+) blocking effect. External Mn(2+) or Mg(2+) can compete with external Ba(2+) at the IRK1 binding site at an external Ba(2+) concentration of 30 mol/L and K(+) concentration of 90 mmol/L. Inactivation becomes slower and slower and Ba(2+) is repelled from the IRK1 binding site when Mg(2+) or Mn(2+) con-centration is further increased. Mg(2+), but not Mn(2+), can contact with deeper binding sites of IRK1 to block the channel, suggesting that multiple-ion blockage may exist in IRK1.


Subject(s)
Barium/pharmacology , Extracellular Space/metabolism , Monokines/antagonists & inhibitors , Potassium Channels/metabolism , Animals , Chemokine CXCL2 , Microelectrodes , Oocytes/metabolism , Rats , Xenopus
13.
Inflammation ; 23(4): 371-85, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10443799

ABSTRACT

Acute thermal trauma is well known to produce evidence of a "systemic inflammatory response" in vivo, as manifested by evidence of complement activation, appearance in plasma of a variety of inflammatory factors, and development of multi-organ injury. The current studies were focused on acute thermal injury of rat skin and factors responsible for accompanying activation of blood neutrophils. Acute thermal injury of rat skin resulted in a time-dependent loss of L-selectin and up-regulation of Mac-1 (CD11b/CD18) on blood neutrophils, with no changes in LFA-1 (CD11a/CD18). The loss of L-selectin was prevented by blockade of C5a but not by blockade of the alpha-chemokine, macrophage inflammatory protein-2 (MIP-2). C5a, the alpha chemokines, MIP-2 and keratinocyte-derived cytokine (KC), and platelet activating factor (PAF) contributed to up-regulation of blood neutrophil Mac-1. Blocking interventions against these mediators also blunted the degree of neutropenia developing after thermal trauma. These data suggest that activation of blood neutrophils after thermal trauma is related to the role of several chemotactic mediators. These studies may provide clues regarding factors responsible for development of the "systemic inflammatory response syndrome" after thermal injury in the experimental model employed.


Subject(s)
Burns/immunology , Chemotactic Factors/physiology , Neutrophil Activation/immunology , Receptors, Cell Surface , Receptors, G-Protein-Coupled , Animals , Antibodies, Blocking/pharmacology , Burns/metabolism , Burns/therapy , CD11 Antigens/biosynthesis , CD18 Antigens/biosynthesis , Cell Adhesion Molecules/biosynthesis , Chemokine CXCL2 , Chemokines , Complement C5a/antagonists & inhibitors , Complement C5a/immunology , Complement C5a/physiology , Cytokines/antagonists & inhibitors , Cytokines/physiology , Immune Sera/pharmacology , L-Selectin/biosynthesis , Leukocyte Count , Male , Monokines/antagonists & inhibitors , Monokines/immunology , Monokines/physiology , Neutrophils/immunology , Neutrophils/metabolism , Platelet Membrane Glycoproteins/antagonists & inhibitors , Rats , Rats, Long-Evans , Up-Regulation/immunology
14.
Infect Immun ; 67(5): 2590-601, 1999 May.
Article in English | MEDLINE | ID: mdl-10225925

ABSTRACT

Chemokines are low-molecular-weight chemotactic cytokines that have been shown to play a central role in the perivascular transmigration and accumulation of specific subsets of leukocytes at sites of tissue damage. Using in situ hybridization (ISH), we investigated the mRNA induction of macrophage inflammatory protein 2 (MIP-2), MIP-1alpha, monocyte chemoattractant protein 1 (MCP-1), and RANTES. Challenge of infant rats' brains with Haemophilus influenzae type b intraperitoneally resulted in the time-dependent expression of MIP-2, MIP-1alpha, MCP-1, and RANTES, which was maximal 24 to 48 h postinoculation. Immunohistochemistry showed significant increases in neutrophils and macrophages infiltrating the meninges, the ventricular system, and the periventricular area. The kinetics of MIP-2, MIP-1alpha, MCP-1, and RANTES mRNA expression paralleled those of the recruitment of inflammatory cells and disease severity. Administration of anti-MIP-2 or anti-MIP-1alpha antibodies (Abs) resulted in significant reduction of neutrophils. Administration of anti-MCP-1 Abs significantly decreased macrophage infiltration. Combined studies of ISH and immunohistochemistry showed that MIP-2- and MIP-1alpha-positive cells were neutrophils and macrophages. MCP-1-positive cells were neutrophils, macrophages, and astrocytes. Expression of RANTES was localized predominantly to resident astrocytes and microglia. The present study indicates that blocking of MIP-2 or MIP-1alpha bioactivity in vivo results in decreased neutrophil influx. These data are also the first demonstration that the C-C chemokine MIP-1alpha is involved in neutrophil recruitment in vivo.


Subject(s)
Chemotactic Factors/antagonists & inhibitors , Chemotactic Factors/immunology , Macrophage Inflammatory Proteins/antagonists & inhibitors , Macrophage Inflammatory Proteins/immunology , Meningitis, Bacterial/immunology , Monokines/antagonists & inhibitors , Monokines/immunology , Neutrophils/immunology , Animals , Base Sequence , Central Nervous System/immunology , Central Nervous System/pathology , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CCL3 , Chemokine CCL4 , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CXCL2 , Chemotaxis, Leukocyte , DNA Probes/genetics , Haemophilus Infections/immunology , Haemophilus Infections/pathology , Haemophilus influenzae type b/immunology , Haemophilus influenzae type b/pathogenicity , Immunohistochemistry , In Situ Hybridization , Macrophages/immunology , Macrophages/pathology , Meningitis, Bacterial/pathology , Neutralization Tests , Neutrophils/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rabbits , Rats , Rats, Sprague-Dawley
15.
Clin Infect Dis ; 28(3): 520-6, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10194070

ABSTRACT

Specific microorganisms initiate the immunoinflammatory processes that destroy tissue in periodontitis. Recent work has demonstrated, in addition to bacterial control, that modulation of the host immunoinflammatory response is also capable of controlling periodontitis. Matrix metalloproteinases (MMPs) destroy collagen and other matrix components, and the osteoclastic bone remodeling determines the periodontal bone response to a bacterial challenge. Other components of the biology, including cytokines and prostanoids, regulate MMPs and bone remodeling and are also involved in regulating the production of defensive elements, such as antibody. Agents directed at blocking MMPs or osteoclastic activity are effective in reducing periodontitis. Agents that inhibit prostaglandin E2 and selective blockage of specific cytokines have also been effective. Improved knowledge of bacterium-host interactions and of the processes leading to tissue destruction will help to identify targets for host modulation to reduce periodontitis in selected situations.


Subject(s)
Periodontitis/immunology , Periodontitis/therapy , Alveolar Bone Loss/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Bone Remodeling/drug effects , Diphosphonates/therapeutic use , Humans , Immunologic Factors/therapeutic use , Mandibular Diseases/prevention & control , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/metabolism , Monokines/antagonists & inhibitors , Periodontitis/prevention & control
16.
J Immunol ; 162(4): 2217-26, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-9973497

ABSTRACT

Protection against infections with the intracellular bacterium Chlamydia spp. requires Th1-polarized CD4+ T cell immunity. In BALB/c mouse lung infections, immediate innate and nascent Chlamydia-specific immune responses following intranasal inoculation of Chlamydia psittaci strain B577 were modulated by 7-day i.p. administration of murine rIL-12, the initiation cytokine for Th1 immunity. Treatment with IL-12 reduced the severity of chlamydial pneumonia, abolished mortality (37.5% in untreated mice), and significantly reduced numbers of chlamydial organisms in lungs. On day 4 after inoculation, the neutrophil:macrophage ratio in bronchointerstitial pneumonias was 1.96 in untreated mice and 0.51 in IL-12-treated mice. This immediate, IL-12-mediated shift in innate inflammatory phenotype was correlated with a significant reduction of lung concentrations of the neutrophil chemoattractant macrophage inflammatory protein (MIP)-2 (putative murine homologue of human IL-8), monocyte chemotactic protein-1, and TNF-alpha; and a reduction in MIP-1alpha and IFN-gamma, at high-dose infection only, and IL-12-independent IL-10 levels. Chlamydia-specific Ab titers and Ig isotype ratios indicated an IL-12-dependent Th1 shift. Recall responses of IL-12-primed mice to secondary chlamydial lung infection eliminated chlamydiae more effectively and generated a lung cytokine profile conducive to perpetuation of the Th1 memory population. These data support the hypothesis that genetic differences in endogenous IL-12 production and response pathways could determine disease outcomes characterized by poor chlamydial clearance and a purulent inflammatory infiltrate vs effective elimination of chlamydiae in a macrophage-dominated response.


Subject(s)
Cell Movement/immunology , Chlamydia Infections/prevention & control , Interleukin-12/administration & dosage , Monokines/metabolism , Neutrophils/immunology , Pneumonia, Bacterial/prevention & control , Administration, Intranasal , Animals , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/metabolism , Chemokine CXCL2 , Chlamydia Infections/immunology , Chlamydia Infections/mortality , Chlamydia Infections/pathology , Chlamydophila psittaci/immunology , Female , Immunization, Secondary , Immunoglobulin G/biosynthesis , Interleukin-12/therapeutic use , Leukocyte Count , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred BALB C , Monokines/antagonists & inhibitors , Neutrophils/microbiology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/mortality , Pneumonia, Bacterial/pathology , Survival Analysis , Th1 Cells/immunology , Th1 Cells/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
17.
J Immunol ; 162(4): 2321-5, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-9973510

ABSTRACT

Under a variety of conditions, alveolar macrophages can generate early response cytokines (TNF-alpha, IL-1), complement components, and chemotactic cytokines (chemokines). In the current studies, we determined the requirements for TNF-alpha and the complement activation product C5a in chemokine production in vitro and in vivo. Two rat CXC chemokines (macrophage inflammatory protein (MIP)-2 and cytokine-induced neutrophil chemoattractant (CINC)) as well as three rat CC chemokines (MIP-1alpha, MIP-1beta, and monocyte chemoattractant protein (MCP)-1) were investigated. Chemokine generation in vitro was studied in rat alveolar macrophages stimulated with IgG immune complexes in the absence or presence of Abs to TNF-alpha or C5a. The rat lung injury model induced by IgG immune complex deposition was employed for in vivo studies. Abs to TNF-alpha or C5a were administered intratracheally or i.v., and effects on chemokine levels in bronchoalveolar lavage fluids were quantitated by ELISA. Both in vitro and in vivo studies demonstrated the requirements for TNF-alpha and C5a for full generation of CXC and CC chemokines. In vitro and in vivo blockade of TNF-alpha or C5a resulted in significantly reduced production of chemokines. Supernatant fluids from in vitro-stimulated macrophages revealed by Western blot analysis the presence of C5a/C5adesArg, indicating intrinsic generation of C5a/C5adesArg by alveolar macrophages and explaining the higher efficiency of intratracheal vs i.v. blockade of C5a in reducing chemokine production. These results underscore the central role of both TNF-alpha and C5a, which appear to function as autocrine activators to promote CXC and CC chemokine generation by alveolar macrophages.


Subject(s)
Chemokines, CXC/biosynthesis , Complement C5a/physiology , Intercellular Signaling Peptides and Proteins , Tumor Necrosis Factor-alpha/physiology , Animals , Antibodies, Blocking/administration & dosage , Antibodies, Blocking/pharmacology , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Cell-Free System/chemistry , Cell-Free System/immunology , Cells, Cultured , Chemokine CXCL2 , Chemokines, CXC/antagonists & inhibitors , Chemotactic Factors/biosynthesis , Complement C5a/immunology , Growth Substances/biosynthesis , Injections, Intravenous , Intubation, Intratracheal , Lung/immunology , Lung/metabolism , Macrophages, Alveolar/chemistry , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Monokines/antagonists & inhibitors , Monokines/biosynthesis , Rats , Rats, Long-Evans , Tumor Necrosis Factor-alpha/immunology
18.
Am J Physiol ; 273(2 Pt 1): G333-41, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9277411

ABSTRACT

Interleukin-11 (IL-11) is a stromal cell-derived cytokine with several biological activities against hematopoietic cells. Recent results indicated that IL-11 reduced mucosal damage in animal models of colitis. This study aimed to explore the action of recombinant human IL-11 (rhIL-11) on the intestinal effects of Clostridium difficile toxin A, an inflammatory enterotoxin, and cholera toxin, a noninflammatory enterotoxin in rat ileum. We administered rhIL-11 subcutaneously to rats before injection of toxin A into ileal loops and measured fluid secretion, epithelial permeability to mannitol, histopathological damage, and release of rat mast cell protease II (RMCP II) from intestinal mast cells and of tumor necrosis factor-alpha (TNF-alpha) and macrophage inflammatory protein-2 (MIP-2) from lamina propria macrophages. rhIL-11 (50-1,000 micrograms/kg) inhibited toxin A but not cholera toxin-mediated secretion and permeability in a dose-dependent fashion and reduced toxin A-induced epithelial cell damage. Rats treated with rhIL-11 also showed reduced RMCP II, TNF-alpha, and MIP-2 release in response to toxin A. Exposure of rat peripheral monocytes in vitro to rhIL-11 (1 microgram/ml) inhibited lipopolysaccharide and toxin A-mediated increases in TNF-alpha mRNA and protein levels. We conclude that rhIL-11 blocks the intestinal effects of C. difficile toxin A, possibly by inhibiting release of inflammatory mediators from mucosal mast cells and intestinal macrophages.


Subject(s)
Bacterial Toxins , Enterotoxins/antagonists & inhibitors , Enterotoxins/pharmacology , Ileum/drug effects , Interleukin-11/pharmacology , Animals , Chemokine CXCL2 , Chymases , Humans , Ileum/pathology , Macrophages/metabolism , Male , Monocytes/metabolism , Monokines/antagonists & inhibitors , Rats , Rats, Wistar , Recombinant Proteins , Serine Endopeptidases/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors
19.
Cytokine ; 9(4): 242-8, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9112332

ABSTRACT

We investigated time-related productions of certain cytokines in lipopolysaccharide (LPS)-stimulated RAW264.7 cells, a murine macrophage cell line, by enzyme-linked immunosorbent assay. Macrophage inflammatory protein-2 (MIP-2) levels became detectable after 2 h and markedly increased over the first 8 h. Thereafter, this level remained at the same level between 10 and 16 h, and then increased again until 24 h, showing a tendency of biphasic pattern. Tumour necrosis factor (TNF)-alpha was detectable at 2 h and then increased sharply until 6 h at which it attained its peak. A low but recognizable level of interleukin (IL)-1alpha/beta was also detectable. When the inhibitory effect of ferulic acid (FA), an active component of the Rhizoma of Cimicifuga sp. used frequently as anti-inflammatory drug in Japanese Oriental medicines, was compared with that of dexamethasone (DX) on MIP-2 and TNF-alpha productions in response to LPS, both FA and DX could reduce the production of these cytokines in a dose-dependent manner. Concerning TNF-alpha, however, the inhibitory effect of FA was very weak compared with that of DX. In addition, FA as well as DX reduced MIP-2 production induced by TNF-alpha. These data suggest that MIP-2 might be induced by a direct effect of LPS and in part indirect one via initial induction of other cytokines such as TNF-alpha, leading a tendency of biphasic pattern. Comparing DX, FA is considered to be a novel and unique drug inhibiting MIP-2 production more selectively.


Subject(s)
Coumaric Acids/pharmacology , Macrophages/metabolism , Monokines/antagonists & inhibitors , Monokines/biosynthesis , Animals , Cell Line , Chemokine CXCL2 , Dexamethasone/pharmacology , Kinetics , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice , Monokines/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology
20.
Eur J Neurosci ; 8(2): 424-8, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8714712

ABSTRACT

Macrophage inflammatory protein-1 (MIP-1) evokes an intense fever, independent of a prostaglandin mechanism, and is now thought to play an important role in the defence response to bacterial pyrogens. The purpose of this study was 2-fold: (i) to determine whether the potent doublet of this cytokine, MIP-1beta, is actually produced in the brain in response to a pyrogenic dose of a lipopolysaccharide of Escherichia coli and (ii) to determine the anatomical site of synthesis of this cytokine in the brain. Following the intense fever produced by intraperitoneal administration of lipopolysaccharide in the unrestrained rat, MIP-1beta immunoreactivity was identified post mortem in two regions of the brain implicated in fever: the organum vasculosum laminae terminalis (OVLT) and the anterior hypothalamic, preoptic area (AH/POA). Microinjection of goat anti-mouse MIP-1beta antibody (anti-MIP-1beta) directly int the AH/POA markedly suppressed fever in rats in response to lipopolysaccharide. Further anti-MIP-1beta administered 180 min after the injection of lipopolysaccharide acted as an antipyretic and reversed the fever induced by the endotoxin. anti-MIP-1beta or control immunoglobulin G antibody microinjected into the hypothalamus immediately before the intraperitoneal injection of the control saline did not alter the temperature of the rats. Taken together, the present results demonstrate that MIP-1beta is produced in the brain in response to a bacterial endotoxin. These observations, in the light of earlier data on fever induced by MIP-1beta, further support the hypothesis that endogenously synthesized MIP-1beta acts as an intermediary factor in the evocation of fever by acting on the thermosensitive cells of the brain.


Subject(s)
Brain/metabolism , Endotoxins/toxicity , Escherichia coli/chemistry , Fever/metabolism , Lipopolysaccharides/toxicity , Monokines/biosynthesis , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Chemokine CCL4 , Endotoxins/administration & dosage , Fever/chemically induced , Fever/therapy , Hypothalamus, Anterior/metabolism , Injections, Intraperitoneal , Lipopolysaccharides/administration & dosage , Macrophage Inflammatory Proteins , Male , Microinjections , Monokines/antagonists & inhibitors , Monokines/genetics , Monokines/immunology , Preoptic Area/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...