Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
J Pharmacol Sci ; 140(1): 94-101, 2019 May.
Article in English | MEDLINE | ID: mdl-31105025

ABSTRACT

High mobility group box-1 (HMGB1), a representative damage associated-molecular pattern (DAMP), has been reported to be involved in many inflammatory diseases. Several drugs are thought to have potential to control the translocation and secretion of HMGB1, or to neutralize extracellular HMGB1 by binding to it. One of these drugs, anti-HMGB1 monoclonal antibody (mAb), is highly specific for HMGB1 and has been shown to be effective for the treatment of a wide range of CNS diseases when modeled in animals, including stroke, traumatic brain injury, Parkinson's disease, epilepsy and Alzheimer's disease. Thus, anti-HMGB1 mAb not only is useful for target validation but also has extensive potential for the treatment of the above-mentioned diseases. In this review, we summarize existing knowledge on the effects of anti-HMGB1 mAb on CNS and PNS diseases, the common features of translocation and secretion of HMGB1 and the functional roles of HMGB1 in these diseases. The existing literature suggests that anti-HMGB1 mAb therapy would be effective for a wide range of CNS and PNS diseases.


Subject(s)
Alzheimer Disease/drug therapy , Antibodies, Monoclonal/therapeutic use , Brain Injuries, Traumatic/drug therapy , HMGB1 Protein/immunology , Parkinson Disease/drug therapy , Stroke/drug therapy , Epilepsy/drug therapy , HMGB1 Protein/metabolism , HMGB1 Protein/physiology , Humans , Molecular Targeted Therapy , Neuralgia/drug therapy
2.
World Neurosurg ; 122: e864-e871, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30391757

ABSTRACT

BACKGROUND: High mobility group box 1 (HMGB1) protein plays a key role in triggering inflammatory responses in many diseases. Our previous study showed that HMGB1 is found upstream of secondary damage in traumatic brain injury (TBI). We found that anti-HMGB1 monoclonal antibody (mAb) effectively decreased acute brain damage, including the disruption of the blood-brain barrier, brain edema, and neurologic dysfunction. This effect of anti-HMGB1 mAb lasts for at least 1 week. In this study, we explored subacute effects of anti-HMGB1 mAb after TBI. METHODS: TBI was induced in rats by fluid percussion. Anti-HMGB1 mAb or control mAb was given intravenously after TBI. Histochemical staining, plasma levels of HMGB1, motor activity and memory, and video electroencephalography monitoring were evaluated 2 weeks after fluid percussion injury. RESULTS: Anti-HMGB1 mAb remarkably attenuated accumulation of activated microglia in the rat cortex in the ipsilateral hemisphere after TBI. Anti-HMGB1 mAb also prevented neuronal death in the hippocampus in the ipsilateral hemisphere after TBI. Treatment of rats with anti-HMGB1 mAb inhibited HMGB1 translocation and suppressed impairment of motor function. The beneficial effects of anti-HMGB1 mAb on motor and cognitive function persisted for 14 days after injury. Treatment with anti-HMGB1 mAb also had positive effects on electroencephalography activity. CONCLUSIONS: The beneficial effects of anti-HMGB1 mAb continued during the subacute postinjury phase, suggesting that anti-HMGB1 mAb may prevent cognitive dysfunction after TBI.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Brain Injuries, Traumatic/drug therapy , Cognitive Dysfunction/drug therapy , HMGB1 Protein/antagonists & inhibitors , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacology , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/pathology , Cognitive Dysfunction/blood , Cognitive Dysfunction/pathology , HMGB1 Protein/blood , Male , Protein Binding/physiology , Rats , Rats, Wistar
3.
Stem Cells ; 36(5): 737-750, 2018 05.
Article in English | MEDLINE | ID: mdl-29517828

ABSTRACT

Together with residual host neurons, transplanted neural stem cell (NSC)-derived neurons play a critical role in reconstructing disrupted neural circuits after spinal cord injury (SCI). Since a large number of tracts are disrupted and the majority of host neurons die around the lesion site as the damage spreads, minimizing this spreading and preserving the lesion site are important for attaining further improvements in reconstruction. High mobility group box-1 (HMGB1) is a damage-associated molecular pattern protein that triggers sterile inflammation after tissue injury. In the ischemic and injured brain, neutralization of HMGB1 with a specific antibody reportedly stabilizes the blood-brain barrier, suppresses inflammatory cytokine expression, and improves functional recovery. Using a SCI model mouse, we here developed a combinatorial treatment for SCI: administering anti-HMGB1 antibody prior to transplantation of NSCs derived from human induced pluripotent stem cells (hiPSC-NSCs) yielded a dramatic improvement in locomotion recovery after SCI. Even anti-HMGB1 antibody treatment alone alleviated blood-spinal cord barrier disruption and edema formation, and increased the number of neurites from spared axons and the survival of host neurons, resulting in functional recovery. However, this recovery was greatly enhanced by the subsequent hiPSC-NSC transplantation, reaching an extent that has never before been reported. We also found that this improved recovery was directly associated with connections established between surviving host neurons and transplant-derived neurons. Taken together, our results highlight combinatorial treatment with anti-HMGB1 antibody and hiPSC-NSC transplantation as a promising novel therapy for SCI. Stem Cells 2018;36:737-750.


Subject(s)
Cell Differentiation/physiology , Neural Stem Cells/cytology , Recovery of Function/physiology , Spinal Cord Injuries/therapy , Animals , Cells, Cultured , Disease Models, Animal , HMGB1 Protein/immunology , Humans , Mice, Inbred NOD , Mice, SCID , Stem Cell Transplantation/methods
4.
Neuropharmacology ; 85: 18-26, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24859607

ABSTRACT

Glycyrrhizin (GL) is a major constituent of licorice root and has been suggested to inhibit the release of high mobility group box-1 (HMGB1), a protein considered representative of damage-associated molecular patterns. We found that GL bound HMGB1 but not RAGE with a moderate equilibrium dissociation constant value based on surface plasmon resonance analysis. This complex formation prevented HMGB1 from binding to RAGE in vitro. The effects of glycyrrhizin on traumatic brain injury (TBI) induced by fluid percussion were examined in rats or mice in the present study. GL was administered intravenously after TBI. Treatment of rats with GL dose-dependently suppressed the increase in BBB permeability and impairment of motor functions, in association with the inhibition of HMGB1 translocation in neurons in injured sites. The beneficial effects of GL on motor and cognitive functions persisted for 7 days after injury. The expression of TNF-α, IL-1ß and IL-6 in injured sites was completely inhibited by GL treatment. In RAGE-/- mice, the effects of GL were not observed. These results suggested that GL may be a novel therapeutic agent for TBI through its interference with HMGB1 and RAGE interaction.


Subject(s)
Brain Injuries/drug therapy , Glycyrrhizic Acid/pharmacology , HMGB1 Protein/metabolism , Neuroprotective Agents/pharmacology , Receptors, Immunologic/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Brain/drug effects , Brain/pathology , Brain/physiopathology , Brain Edema/drug therapy , Brain Edema/pathology , Brain Edema/physiopathology , Brain Injuries/pathology , Brain Injuries/physiopathology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cognition/drug effects , Cognition/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Random Allocation , Rats, Wistar , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics
5.
Ann Neurol ; 72(3): 373-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22915134

ABSTRACT

OBJECTIVE: High mobility group box-1 (HMGB1) plays an important role in triggering inflammatory responses in many types of diseases. In this study, we examined the involvement of HMGB1 in traumatic brain injury (TBI) and evaluated the ability of intravenously administered neutralizing anti-HMGB1 monoclonal antibody (mAb) to attenuate brain injury. METHODS: Traumatic brain injury was induced in rats or mice by fluid percussion. Anti-HMGB1 mAb or control mAb was administered intravenously after TBI. RESULTS: Anti-HMGB1 mAb remarkably inhibited fluid percussion-induced brain edema in rats, as detected by T2-weighted magnetic resonance imaging; this was associated with inhibition of HMGB1 translocation, protection of blood-brain barrier (BBB) integrity, suppression of inflammatory molecule expression, and improvement of motor function. In contrast, intravenous injection of recombinant HMGB1 dose-dependently produced the opposite effects. Experiments using receptor for advanced glycation end product (RAGE)(-/-) , toll-like receptor-4 (TLR4)(-/-) , and TLR2(-/-) mice suggested the involvement of RAGE as the predominant receptor for HMGB1. INTERPRETATION: Anti-HMGB1 mAb may provide a novel and effective therapy for TBI by protecting against BBB disruption and reducing the inflammatory responses induced by HMGB1.


Subject(s)
Brain Injuries/drug therapy , HMGB1 Protein/immunology , Immunoglobulin G/therapeutic use , Analysis of Variance , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Brain Edema/etiology , Brain Edema/pathology , Brain Edema/prevention & control , Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Death/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Evans Blue , Functional Laterality , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glycation End Products, Advanced/genetics , Glyceraldehyde 3-Phosphate/metabolism , HMGB1 Protein/metabolism , HMGB1 Protein/pharmacology , Hypoxia-Inducible Factor 1/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/metabolism , Motor Activity/drug effects , Neurons/drug effects , Neurons/pathology , Neurons/ultrastructure , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Wistar , Rotarod Performance Test , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 4/deficiency
6.
Acta Med Okayama ; 65(6): 369-77, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22189477

ABSTRACT

We investigated the expression and localization of high-mobility group box chromosomal protein-1 (HMGB-1) in human osteoarthritic (OA) cartilage in relation to the histopathological grade of cartilage destruction, and examined the role of HMGB-1 in the regulation of proinflammatory cytokine expression in chondrocytes. An immunohistochemical study demonstrated that total HMGB-1-positive cell ratios increase as the Osteoarthritis Research Society International (OARSI) histological grade increased. The population of cytoplasmic HMGB-1-positive chondrocytes was especially increased in the deep layers of higher-grade cartilage. The ratios and localization of receptors for advanced glycation end products (RAGE) expression by chondrocytes in Grade 2, 3, and 4 were significantly higher than those in Grade 1. In vitro stimulation with IL-1ß, but not TNFα, significantly upregulated the expression of HMGB-1 mRNA by human OA chondrocytes. Both IL-1ß and TNFα promoted the translocation of HMGB-1 from nuclei to cytoplasm. IL-1ß and TNFα secretions were stimulated at higher levels of HMGB-1. The results of our study suggest the involvement of HMGB-1 in the pathogenesis of cartilage destruction in OA.


Subject(s)
Cartilage, Articular/physiology , Gene Expression , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Osteoarthritis/genetics , Osteoarthritis/metabolism , Aged , Aged, 80 and over , Animals , Cartilage, Articular/cytology , Cartilage, Articular/pathology , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/physiology , Humans , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Middle Aged , Osteoarthritis/pathology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
7.
Stroke ; 42(5): 1420-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21474801

ABSTRACT

BACKGROUND AND PURPOSE: High mobility group box-1 (HMGB1) exhibits inflammatory cytokine-like activity in the extracellular space. We previously demonstrated that intravenous injection of anti-HMGB1 monoclonal antibody (mAb) remarkably ameliorated brain infarction induced by middle cerebral artery occlusion in rats. In the present study, we focused on the protective effects of the mAb on the marked translocation of HMGB1 in the brain, the disruption of the blood-brain barrier (BBB), and the resultant brain edema. METHODS: Middle cerebral artery occlusion in the rat was used as the ischemia model. Rats were treated with anti-HMGB1 mAb or control IgG intravenously. BBB permeability was measured by MRI. Ultrastructure of the BBB unit was observed by transmission electron microscope. The in vitro BBB system was used to study the direct effects of HMGB1 in BBB components. RESULTS: HMGB1 was time-dependently translocated and released from neurons in the ischemic rat brain. The mAb reduced the edematous area on T2-weighted MRI. Transmission electron microscope observation revealed that the mAb strongly inhibited astrocyte end feet swelling, the end feet detachment from the basement membrane, and the opening of the tight junction between endothelial cells. In the in vitro reconstituted BBB system, recombinant HMGB1 increased the permeability of the BBB with morphological changes in endothelial cells and pericytes, which were inhibited by the mAb. Moreover, the anti-HMGB1 mAb facilitated the clearance of serum HMGB1. CONCLUSIONS: These results indicated that the anti-HMGB1 mAb could be an effective therapy for brain ischemia by inhibiting the development of brain edema through the protection of the BBB and the efficient clearance of circulating HMGB1.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Blood-Brain Barrier/physiology , Brain Ischemia/therapy , HMGB1 Protein/immunology , Animals , Antibodies, Monoclonal/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/ultrastructure , Brain Edema/physiopathology , Brain Edema/prevention & control , Brain Ischemia/metabolism , Brain Ischemia/pathology , Disease Models, Animal , HMGB1 Protein/metabolism , Immunoglobulins, Intravenous , Infarction, Middle Cerebral Artery , Magnetic Resonance Imaging , Male , Microscopy, Electron, Transmission , Rats , Rats, Wistar , Time Factors , Treatment Outcome
8.
Arterioscler Thromb Vasc Biol ; 31(2): 313-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21088249

ABSTRACT

OBJECTIVE: High-mobility group box protein 1 (HMGB1) is a DNA-binding protein and cytokine highly expressed in atherosclerotic lesions, but its pathophysiological role in atherosclerosis is unknown. We investigated its role in the development of atherosclerosis in ApoE-/- mice. METHODS AND RESULTS: Apolipoprotein E-deficient (ApoE-/-) mice fed a high-fat diet were administered a monoclonal anti-HMGB1 neutralizing antibody, and the effects on lesion size, immune cell accumulation, and proinflammatory mediators were assessed using Oil Red O, immunohistochemistry, and real-time polymerase chain reaction. As with human atherosclerotic lesions, lesions in ApoE-/- mice expressed HMGB1. Treatment with the neutralizing antibody attenuated atherosclerosis by 55%. Macrophage accumulation was reduced by 43%, and vascular cell adhesion molecule-1 and monocyte chemoattractant protein-1 expression was attenuated by 48% and 72%, respectively. CD11c+ dendritic cells were reduced by 65%, and the mature (CD83+) population was reduced by 60%. Treatment also reduced CD4+ cells by nearly 50%. mRNAs in lesions encoding tumor necrosis factor-α and interleukin-1ß tended to be reduced. Mechanistically, HMGB1 stimulated macrophage migration in vitro and in vivo; in vivo, it markedly augmented the accumulation of F4/80+Gr-1(Ly-6C)+ macrophages and also increased F4/80+CD11b+ macrophage numbers. CONCLUSIONS: HMGB1 exerts proatherogenic effects augmenting lesion development by stimulating macrophage migration, modulating proinflammatory mediators, and encouraging the accumulation of immune and smooth muscle cells.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Apolipoproteins E/deficiency , Atherosclerosis/chemically induced , Atherosclerosis/prevention & control , Dietary Fats/adverse effects , HMGB1 Protein/immunology , Animals , Antibodies, Neutralizing/pharmacology , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Cell Movement/drug effects , Chemokine CCL2/metabolism , Disease Models, Animal , HMGB1 Protein/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
10.
Acta Med Okayama ; 63(5): 249-62, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19893601

ABSTRACT

Angiogenesis involves complex processes mediated by several factors and is associated with inflammation and wound healing. High mobility group box 1 (HMGB1) is released from necrotic cells as well as macrophages and plays proinflammatory roles. In the present study, we examined whether HMGB1 would exhibit angiogenic activity in a matrigel plug assay in mice. HMGB1 in combination with heparin strongly induced angiogenesis, whereas neither HMGB1 nor heparin alone showed such angiogenic activity. The heparin-dependent induction of angiogenesis by HMGB1 was accompanied by increases in the expression of tumor necrosis factor-alpha (TNF-alpha) and vascular endothelial growth factor-A120 (VEGF-A120). It is likely that the dependence of the angiogenic activity of HMGB1 on heparin was due to the efficiency of the diffusion of the HMGB1-heparin complex from matrigel to the surrounding areas. VEGF-A165 possessing a heparin-binding domain showed a pattern of heparin-dependent angiogenic activity similar to that of HMGB1. The presence of heparin also inhibited the degradation of HMGB1 by plasmin in vitro. Taken together, these results suggested that HMGB1 in complex with heparin possesses remarkable angiogenic activity, probably through the induction of TNF-alpha and VEGF-A120.


Subject(s)
HMGB1 Protein/pharmacology , Heparin/pharmacology , Implants, Experimental , Neovascularization, Physiologic , Animals , Collagen , Cytokines/genetics , Cytokines/metabolism , Drug Combinations , Fibrinolysin/metabolism , Laminin , Male , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Proteoglycans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
11.
Eur J Pharmacol ; 623(1-3): 89-95, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19786011

ABSTRACT

Histidine-rich glycoprotein (HRG) is a heparin-binding glycoprotein present in plasma at 100microg/ml. A recent study revealed that HRG suppressed heparin-dependent basic fibroblast growth factor (bFGF)-induced angiogenesis. Additionally, we reported that high mobility group box 1 (HMGB1) in complex with heparin induces angiogenesis; therefore, we examined the effect of HRG on heparin-dependent HMGB1-induced angiogenesis in the present study. HRG completely inhibited angiogenesis induced by HMGB1 in complex with heparin. HRG inhibited the diffusion of a complex of HMGB1 with heparin from matrigel into surrounding tissue. HRG also competed with HMGB1 for heparin binding in vitro. Moreover, HRG inhibited heparin-dependent vascular endothelial growth factor-A(165) (VEGF-A(165))-induced angiogenesis. These results strongly suggested that HRG might be an inhibitor of angiogenesis induced by growth factors with heparin binding activity and that HRG may be a potential drug for angiogenic diseases, including tumor growth.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Angiogenic Proteins/antagonists & inhibitors , HMGB1 Protein/antagonists & inhibitors , Heparin/pharmacology , Neovascularization, Pathologic , Proteins/pharmacology , Angiogenic Proteins/pharmacology , Animals , Binding, Competitive , Biocompatible Materials , Collagen , Drug Combinations , Gene Expression Regulation/drug effects , HMGB1 Protein/metabolism , HMGB1 Protein/pharmacology , Heparin/metabolism , Implants, Experimental , Laminin , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/chemically induced , Protein Binding , Proteins/metabolism , Proteoglycans , RNA, Messenger/metabolism , Time Factors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
12.
Acta Med Okayama ; 63(4): 203-11, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19727205

ABSTRACT

Interaction between the receptor for advanced glycation end products (RAGE) and its ligands has been implicated in the pathogenesis of various inflammatory disorders. In this study, we establish an in vitro binding assay in which recombinant human high-mobility group box 1 (rhHMGB1) or recombinant human S100A12 (rhS100A12) immobilized on the microplate binds to recombinant soluble RAGE (rsRAGE). The rsRAGE binding to both rhHMGB1 and rhS100A12 was saturable and dependent on the immobilized ligands. The binding of rsRAGE to rhS100A12 depended on Ca2+ and Zn2+, whereas that to rhHMGB1 was not. Scatchard plot analysis showed that rsRAGE had higher affinity for rhHMGB1 than for rhS100A12. rsRAGE was demonstrated to bind to heparin, and rhS100A12, in the presence of Ca2+, was also found to bind to heparin. We examined the effects of heparin preparations with different molecular sizes - unfractionated native heparin (UFH), low molecular weight heparin (LMWH) 5000Da, and LMWH 3000Da - on the binding of rsRAGE to rhHMGB1 and rhS100A12. All 3 preparations concentration-dependently inhibited the binding of rsRAGE to rhHMGB1 to a greater extent than did rhS100A12. These results suggested that heparin's anti-inflammatory effects can be partly explained by its blocking of the interaction between HMGB1 or S100A12 and RAGE. On the other hand, heparin would be a promising effective remedy against RAGE-related inflammatory disorders.


Subject(s)
HMGB1 Protein/metabolism , Heparin/pharmacology , Receptors, Immunologic/metabolism , S100 Proteins/metabolism , Anti-Inflammatory Agents/pharmacology , Calcium/pharmacology , Chromatography, Affinity , Humans , Receptor for Advanced Glycation End Products , S100A12 Protein , Zinc/pharmacology
13.
Acta Med Okayama ; 63(1): 65-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19247424

ABSTRACT

Lipopolysaccharide (LPS) is one of the major causes of septic shock. The polymyxin B-immobilized filter column (PMX) was developed for the adsorption of endotoxin by direct hemoperfusion and has been used for the treatment of LPS-induced septic shock. In this study, we demonstrated that PMX also specifically bound monocytes from the peripheral blood leukocytes of septic patients by mean of an analysis of bound cells using immunocytochemical and electron microscopic techniques. The specific removal of monocytes from septic patients may produce beneficial effects by reducing the interaction between monocytes and functionally associated cells including vascular endothelial cells.


Subject(s)
Cell Separation/methods , Monocytes/cytology , Polymyxin B/chemistry , Sepsis/therapy , Adsorption , Aged , CD11b Antigen/blood , Female , Filtration , Humans , L-Selectin/blood , Male , Middle Aged , Sepsis/blood
14.
Yakugaku Zasshi ; 129(1): 25-31, 2009 Jan.
Article in Japanese | MEDLINE | ID: mdl-19122433

ABSTRACT

Ischemic brain infarction is high among th causes of death in Japan, and the medical and social burden by severe sequela is also extremely serious. In this symposium, we show that treatment with anti-high mobility group box 1 (HMGB1) monoclonal antibody (mAb) remarkably ameliorated brain infarction induced by 2-hour occlusion of the middle cerebral artery in rats, even when the mAb was administered after the start of reperfusion. Whereas HMGB1 is usually localized in nucleus, after stimulation it is secreted into extracellular space by an unknown non-classical pathway, and exhibits an inflammatory cytokine-like activity. Treatment with mAb reduced infarct size, and the accompanying neurological deficits in locomotor function were significantly improved. In addition, some biochemical markers such as permeability of the blood-brain barrier, the expression of tumor necrosis factor-alfa, inducible nitric oxide synthase and matrix metalloproteinase-9 were altered by mAb injection. These findings indicate the usefulness of HMGB1 as a novel therapeutic to target ischemic stroke.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cerebral Infarction/drug therapy , Cerebral Infarction/etiology , HMGB1 Protein/immunology , Animals , Blood-Brain Barrier/metabolism , HMGB1 Protein/physiology , Humans , Matrix Metalloproteinase 9/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats , Tumor Necrosis Factor-alpha/metabolism
15.
Basic Clin Pharmacol Toxicol ; 102(4): 394-8, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18312491

ABSTRACT

Inflammatory reactions play an important role in ischaemia/reperfusion injury in various organs. Since histamine H(4) action has been shown to prevent the development of ischaemia/reperfusion liver injury, we examined the effects of dimaprit, a histamine H(2)/H(4) receptor agonist, on ischaemia-induced cytokine release and liver damage. Male Wistar rats (300 g) were subjected to warm ischaemia for 30 min. by occlusion of the left portal vein and hepatic artery under halothane anaesthesia. Saline or dimaprit (20 mg/kg, subcutaneously) was injected immediately after reperfusion of blood flow. Transient ischaemia provoked severe liver damage 24 hr after reperfusion, and the plasma concentrations of alanine transaminase and aspartate transaminase were 4600 IU/l and 13,200 IU/l, respectively. The values in the dimaprit group were 55% and 46% of those in control animals, respectively. Dimaprit also reduced the infarct size to 50%. Liver ischaemia markedly increased interleukin-12 levels 2-24 hr after reperfusion. The dimaprit treatment depressed the values to 40-64% of those in the corresponding control group 4-24 hr after reperfusion. Since interleukin-12 facilitates cell-mediated cytotoxicity, the protective effect of dimaprit may be attributed to regulation of cytokine release during reperfusion.


Subject(s)
Cytokines/metabolism , Dimaprit/pharmacology , Histamine Agonists/pharmacology , Ischemia/drug therapy , Liver/drug effects , Reperfusion Injury/prevention & control , Adenosine Triphosphate/metabolism , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Dimaprit/therapeutic use , Disease Models, Animal , Down-Regulation , Hepatic Artery/surgery , Histamine/blood , Histamine Agonists/therapeutic use , Interleukin-12/blood , Ischemia/complications , Ischemia/metabolism , Ischemia/pathology , Ligation , Liver/blood supply , Liver/enzymology , Liver/pathology , Male , Portal Vein/surgery , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine/metabolism , Receptors, Histamine H2/drug effects , Receptors, Histamine H2/metabolism , Receptors, Histamine H4 , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Severity of Illness Index , Time Factors
16.
Yakugaku Zasshi ; 128(2): 247-53, 2008 Feb.
Article in Japanese | MEDLINE | ID: mdl-18239372

ABSTRACT

Histamine is well known for its roles in allergic diseases and anaphylaxis through H(1)-receptor stimulation. The H(1)-receptor stimulation by histamine results in an increase in vascular permeability, vasodilatation, and stimulation of nerve terminals in primary sensory neurons, thereby accelerating the inflammatory responses. On the other hand, histamine has been demonstrated to be involved in the regulation of innate and acquired immune responses through H(2)-receptors. In a previous study with human peripheral blood mononuclear cells, we observed that histamine exerts various regulatory effects on monocyte/macrophage function. In this review, we discuss how inducible histamine protects mice from lethal hepatitis, induced by heat-killed P.acnes (1 mg, i.v.) followed by challenge with a low dose of lipopolysaccharide (1 microg), by reducing the excessive cytokine response in the liver. In addition, from in vivo studies with histidine decarboxylase knockout and H(1)-, H(2)-receptor knockout mice, the protective effect of histamine against fulminant hepatitis is shown to be elicited through H(2)-receptor stimulation.


Subject(s)
Hepatitis/prevention & control , Histamine/physiology , Receptors, Histamine H2/immunology , Animals , Disease Models, Animal , Hepatitis/microbiology , Humans , Lipopolysaccharides , Mice , Propionibacterium acnes
17.
FASEB J ; 21(14): 3904-16, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17628015

ABSTRACT

The high mobility group box-1 (HMGB1), originally identified as an architectural nuclear protein, exhibits an inflammatory cytokine-like activity in the extracellular space. Here we show that treatment with neutralizing anti-HMGB1 monoclonal antibody (mAb; 200 microg, twice) remarkably ameliorated brain infarction induced by 2-h occlusion of the middle cerebral artery in rats, even when the mAb was administered after the start of reperfusion. Consistent with the 90% reduction in infarct size, the accompanying neurological deficits in locomotor function were significantly improved. Anti-HMGB1 mAb inhibited the increased permeability of the blood-brain barrier, the activation of microglia, the expression of TNF-alpha and iNOS, and suppressed the activity of MMP-9, whereas it had little effect on blood flow. Intracerebroventricular injection of HMGB1 increased the severity of infarction. Immunohistochemical study revealed that HMGB1 immunoreactivity in the cell nuclei decreased or disappeared in the affected areas, suggesting the release of HMGB1 into the extracellular space. These results indicate that HMGB1 plays a critical role in the development of brain infarction through the amplification of plural inflammatory responses in the ischemic region and could be an outstandingly suitable target for the treatment. Intravenous injection of neutralizing anti-HMGB1 mAb provides a novel therapeutic strategy for ischemic stroke.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Brain Infarction/immunology , Brain Infarction/therapy , HMGB1 Protein/immunology , Ischemic Attack, Transient/immunology , Ischemic Attack, Transient/therapy , Animals , Brain Infarction/pathology , Cells, Cultured , Drug Delivery Systems , Encephalitis/immunology , Encephalitis/pathology , Encephalitis/therapy , HMGB1 Protein/metabolism , Ischemic Attack, Transient/pathology , Male , Rats , Rats, Sprague-Dawley , Rats, Wistar
18.
Eur J Pharmacol ; 559(1): 69-74, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17254563

ABSTRACT

Cell-to-cell interaction through binding intercellular adhesion molecule (ICAM)-1, B7.1, B7.2 and CD40 on monocytes and their ligands on T-cells plays roles in cytokine production and T-cell proliferation. Interleukin (IL)-18, which is elevated in the plasma during acute rejection after organ transplantation, induces the expression of ICAM-1, B7.1, B7.2 and CD40, production of interferon (IFN)-gamma and IL-12 and proliferation of lymphocytes during human mixed lymphocyte reaction. Nicotine is known to inhibit the production of pro-inflammatory cytokines from macrophages through the stimulation of nicotinic acetylcholine receptor alpha7 subunit. In the present study, we examined the effect of increasing concentrations ranging from 0.1 to 100 microM of nicotine on the expression of ICAM-1, B7.1, B7.2 and CD40, production of IFN-gamma and IL-12 and proliferation of lymphocytes during mixed lymphocyte reaction treated with IL-18 at 100 ng/ml for 48 h. Nicotine inhibited the expression of adhesion molecules, cytokine production and lymphocyte proliferation. The IC50 values of nicotine for inhibition of the IL-18-enhanced ICAM-1 expression, IFN-gamma production and proliferation were 1, 1 and 2 microM, respectively. A non-selective and a selective antagonist for nicotinic acetylcholine receptor alpha7 subunit, mecamylamine and alpha-bungarotoxin abolished the effects of nicotine. The actions of nicotine might depend on stimulation of nicotinic acetylcholine receptor alpha7 subunit. Nicotine induced prostaglandin E(2) production during mixed lymphocyte reaction. The inhibitors of cyclooxygenase (COX)-2 and protein kinase A (PKA) at 100 microM inhibited the actions of nicotine, suggesting that the endogenous prostaglandin E(2) might be, at least, partially involved the actions of nicotine.


Subject(s)
Immunosuppressive Agents , Lymphocyte Culture Test, Mixed , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Cell Adhesion Molecules/biosynthesis , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclooxygenase 2 Inhibitors/pharmacology , Cytokines/biosynthesis , Dinoprostone/biosynthesis , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interleukin-18/pharmacology , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/physiology , alpha7 Nicotinic Acetylcholine Receptor
19.
Eur J Pharmacol ; 558(1-3): 179-84, 2007 Mar 08.
Article in English | MEDLINE | ID: mdl-17222819

ABSTRACT

We examined whether treatment with amodiaquine, a potent inhibitor of histamine N-methyltransferase protects mice from Propionibacterium acnes (P. acnes)-primed and lipopolysaccharide (LPS)-induced hepatitis. The subcutaneous injection of amodiaquine (2 and 5 mg/kg) significantly increased the histamine levels in the liver in comparison to saline treated mice. Pretreatment with amodiaquine also improved the survival rate of the hepatitis mice, and this improvement was partially associated with the decrease in serum levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT). Amodiaquine partially suppressed increases of tumor necrosis factor (TNF)-alpha in the serum and TNF-alpha mRNA expression in the liver, whereas the expression of interleukin (IL)-18, interferon (IFN)-gamma and IL-12 in the liver was not changed by amodiaquine treatment. In conclusion, the present findings suggested that the elevation of endogenous histamine by amodiaquine may thus play a protective role through the regulation of TNF-alpha production in endotoxin-induced hepatic injury mice.


Subject(s)
Amodiaquine/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Enzyme Inhibitors/pharmacology , Histamine N-Methyltransferase/antagonists & inhibitors , Lipopolysaccharides/toxicity , Propionibacterium acnes/drug effects , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Histamine/metabolism , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...