Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
J Neuroimmunol ; 203(1): 94-103, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18692252

ABSTRACT

Complement activation plays a central role in autoimmune demyelination. To explore the possible effects of C5 on post-inflammatory tissue repair, we investigated the transcriptional profile induced by C5 in chronic experimental allergic encephalomyelitis (EAE) using oligonucleotide arrays. We used C5-deficient (C5-d) and C5-sufficient (C5-s) mice to compare the gene expression profile and we found that 390 genes were differentially regulated in C5-s mice as compared to C5-d mice during chronic EAE. Among them, a group of genes belonging to the family of insulin-like growth factor binding proteins (IGFBP) and transforming growth factor (TGF)-beta3 were found most significantly differentially regulated by C5. The dysregulation of these genes suggests that these proteins might be responsible for the gliosis and lack of remyelination seen in C5-d mice with chronic EAE.


Subject(s)
Complement C5/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Gene Expression Regulation/immunology , Insulin-Like Growth Factor Binding Proteins/genetics , Animals , Animals, Outbred Strains , Blotting, Western , Chronic Disease , Complement C5/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Profiling , Gliosis/immunology , Gliosis/pathology , Immunohistochemistry , Insulin-Like Growth Factor Binding Proteins/metabolism , Mice , Mice, Congenic , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology
2.
J Neuroimmunol ; 180(1-2): 9-16, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16905199

ABSTRACT

Multiple sclerosis and its animal model experimental allergic encephalomyelitis are inflammatory demyelinating diseases of the central nervous system mediated by activated lymphocytes, macrophages/microglia and the complement system. Complement activation and the C5b-9 terminal complex contribute to the pathogenesis of these diseases through its role to promote demyelination. C5b-9 was also shown to protect oligodendrocytes from apoptosis both in vitro and in vivo. Our findings indicate that activation of complement and C5b-9 assembly plays a pro-inflammatory role in the acute phase, but may also be neuroprotective.


Subject(s)
Complement System Proteins/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Multiple Sclerosis/immunology , Nerve Fibers, Myelinated/immunology , Animals , Apoptosis/immunology , Complement Membrane Attack Complex/immunology , Cytoprotection , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Humans , Inflammation/physiopathology , Multiple Sclerosis/physiopathology , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Oligodendroglia/immunology
3.
J Rehabil Res Dev ; 43(1): 123-32, 2006.
Article in English | MEDLINE | ID: mdl-16847778

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system. It is mediated by activated lymphocytes, macrophages, microglia, and complement. In MS, myelin-forming oligodendrocytes (OLGs) are the targets of inflammatory and immune attacks. OLG death by apoptosis or necrosis causes the cell loss seen in MS plaques. Studies of experimental allergic encephalomyelitis (EAE) in caspase 11-deficient mice show that caspase-mediated death of OLGs is critical to demyelination. Complement activation may affect MS pathogenesis through activated terminal complex C5b-9, which promotes demyelination, and through sublytic C5b-9, which protects OLGs from apoptosis. By inducing EAE in C5-deficient mice, we showed that complement C5 promotes axon preservation and new myelin formation, which protect OLGs from apoptosis. These findings indicate that activated complement C5b-9 plays a proinflammatory role in acute MS but may also protect OLGs from death in chronic MS.


Subject(s)
Apoptosis/physiology , Complement Activation/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Multiple Sclerosis/physiopathology , Neuroprotective Agents/pharmacology , Oligodendroglia/cytology , Animals , Apoptosis/drug effects , Caspases/deficiency , Cell Death/drug effects , Cell Death/physiology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Fas Ligand Protein/drug effects , Fas Ligand Protein/metabolism , Humans , Mice , Oligodendroglia/drug effects , Risk Factors , Sensitivity and Specificity
4.
J Biol Chem ; 281(28): 19009-18, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16670089

ABSTRACT

Migration and proliferation of aortic endothelial cells (AEC) are critical processes involved in angiogenesis, atherosclerosis, and postangioplasty restenosis. Activation of complement and assembly of the C5b-9 complement complex have been implicated in the pre-lesional stage of atherogenesis and progression of the atherosclerotic lesion. We have shown that C5b-9 induces proliferation and activates phosphatidylinositol 3-kinase (PI3K), but it is unknown whether this can lead to activation of Akt in AEC, a major downstream target of PI3K, or if C5b-9 can induce the migration of AEC, a critical step in angiogenesis. In this study, we show that C5b-9 induces AEC proliferation and migration and also activates the PI3K/Akt pathway. C5b-9 activates Akt as shown by in vitro kinase assay and phosphorylation of Ser-473. C5b-9-induced cell cycle activation was inhibited by pretreatment with LY294002 (PI3K inhibitor), SH-5 (Akt inhibitor), or transfection with Akt siRNA. These data suggests that the PI3K/Akt pathway is required for C5b-9-induced cell cycle activation. FOXO1, a member of forkhead transcription factor family, was phosphorylated at Ser-256 and inactivated after C5b-9 stimulation as shown by a decrease in DNA binding and cytoplasmic relocalization. Cytoplasmic relocalization was significantly reduced after pretreatment with LY294002, SH-5, or transfection with Akt siRNA. Silencing FOXO1 expression using siRNA stimulated AEC proliferation and regulated angiogenic factor release. Our data indicate that C5b-9 regulation of the cell cycle activation in AEC through Akt pathway is dependent on inactivation of FOXO1.


Subject(s)
Aorta/cytology , Complement Membrane Attack Complex/physiology , Endothelium, Vascular/cytology , Forkhead Transcription Factors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Angioplasty , Atherosclerosis/pathology , Cell Movement , Cell Proliferation , Disease Progression , Endothelial Cells/metabolism , Forkhead Box Protein O1 , Humans , Neovascularization, Pathologic
5.
J Immunol ; 176(5): 3173-80, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16493077

ABSTRACT

Activation of the terminal complement cascade involving C5 to C9 proteins has a beneficial role for oligodendrocytes (OLG) in experimental allergic encephalomyelitis, an animal model of multiple sclerosis, by protecting them from apoptotic cell death. We have previously shown that sublytic C5b-9 complexes, through posttranslational regulation of Bad, inhibit the mitochondrial pathway of apoptosis induced by serum deprivation. In the present study, we examined the possible involvement of the caspase-8 and Fas pathway in OLG apoptosis and the role of C5b-9 in this process. In a serum-free defined medium, OLG undergo apoptosis and differentiation concomitantly. Under this condition, we found that caspase-8 processing was increased in association with Bid cleavage and markedly reduced expression of cellular FLIP long isoform protein. The caspase-8 inhibitor Z-IETD-FMK inhibited cell death associated with differentiation in a dose-dependent manner. Exposure to C5b-9 induced an inhibition of caspase-8 activation, Bid cleavage, and a significant increase in expression of cellular FLIP long isoform. These C5b-9 effects were reversed by PI3K inhibitor LY294002. C5b-9 also down-regulated the expression of FasL and the Fas-induced apoptosis. These data suggest that C5b-9 through PI3K signaling can rescue OLG from Fas-mediated apoptosis by regulating caspase-8 processing.


Subject(s)
Apoptosis/immunology , Caspase Inhibitors , Caspases/metabolism , Complement Membrane Attack Complex/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Oligodendroglia/immunology , Protein Processing, Post-Translational/immunology , Up-Regulation/immunology , Animals , Animals, Newborn , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , BH3 Interacting Domain Death Agonist Protein/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspase 8 , Cells, Cultured , Death Domain Receptor Signaling Adaptor Proteins , Down-Regulation/physiology , Fas Ligand Protein , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Oligodendroglia/cytology , Oligodendroglia/enzymology , Phosphoinositide-3 Kinase Inhibitors , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Tumor Necrosis Factors/biosynthesis , Tumor Necrosis Factors/genetics
6.
Exp Mol Pathol ; 78(2): 116-22, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15713436

ABSTRACT

Tumors often exhibit deregulation of the cell cycle and overexpression of cyclins and cyclin-dependent kinases (CDKs). Response gene to complement (RGC)-32 is a substrate and regulator of CDC2 and its overexpression induces cell cycle activation. We investigated RGC-32 mRNA and protein expression in tumors with special emphasis in colon carcinoma. By using an expression array technique we found that 19% of tumor tissues showed increased RGC-32 mRNA expression over the levels of corresponding normal tissues. On the other hand, an increased RGC-32 protein was found in 70% of colon adenocarcinoma samples tested. In colon carcinomas, two major patterns of RGC-32 immunoreactivity were seen: staining of malignant epithelial cells only in some tumors and RGC-32 reactivity of both malignant epithelia as well as cells in the interstitium in others. Colonic epithelium obtained from normal individuals was consistently negative for RGC-32 protein. Overexpression of RGC-32 protein was found in other tumors including prostate, bladder, breast, lung, and other digestive tract tumors. RGC-32 expression was present in the same malignant epithelial cells that also expressed the proliferation marker Ki-67. Our data suggest that RGC-32 overexpression might be part of the deregulation of the cell cycle that is required for the growth of tumor cells.


Subject(s)
Biomarkers, Tumor/analysis , Cell Cycle Proteins/biosynthesis , Colonic Neoplasms/metabolism , Muscle Proteins/biosynthesis , Neoplasms/metabolism , Nerve Tissue Proteins/biosynthesis , Adenocarcinoma/metabolism , Blotting, Northern , Blotting, Western , Carcinoma/metabolism , Cell Line, Tumor , Humans , Immunohistochemistry , Immunoprecipitation , Ki-67 Antigen/metabolism , Polymerase Chain Reaction , RNA, Messenger/analysis , Up-Regulation
7.
J Immunol ; 172(9): 5702-6, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15100315

ABSTRACT

Complement activation is involved in the initiation of Ab-mediated inflammatory demyelination in experimental autoimmune encephalomyelitis (EAE). At a sublytic dose, the C5b-9 membrane attack complex protects oligodendrocytes (OLG) from apoptosis. Using C5-deficient (C5-d) mice, we previously showed a dual role for C5: enhancement of inflammatory demyelination in acute EAE, and promotion of remyelination during recovery. In this study, we investigated the role of C5 in apoptosis in myelin-induced EAE. In acute EAE, C5-d and C5-sufficient (C5-s) mice had similar numbers of total apoptotic cells, whereas C5-s had significantly fewer than C5-d during recovery. In addition, although both groups of mice displayed TUNEL(+) OLG, there were significantly fewer in C5-s than in C5-d during both acute EAE and recovery. Gene array and immunostaining of apoptosis-related genes showed that Fas ligand expression was higher in C5-s. In C5-s mice, Fas(+) cells were also higher than in C5-d mice in acute EAE; however, these cells were significantly reduced during recovery. Together, these findings are consistent with the role of C5, possibly by forming the membrane attack complex, in limiting OLG apoptosis in EAE, thus promoting remyelination during recovery.


Subject(s)
Apoptosis/immunology , Complement C5/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Acute Disease , Animals , Apoptosis/genetics , Cell Count , Complement Activation/genetics , Complement C5/deficiency , Complement C5/genetics , Complement C5/metabolism , Complement Membrane Attack Complex/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Fas Ligand Protein , Female , Ligands , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath/immunology , Myelin Sheath/pathology , Oligodendroglia/immunology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology , fas Receptor/biosynthesis , fas Receptor/metabolism
8.
J Neuroimmunol ; 142(1-2): 58-66, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14512164

ABSTRACT

Sublytic C5b-9 alters the molecular phenotype of myotubes by inhibiting muscle-specific gene expression. Here, we showed that C5b-9 induced c-fos mRNA and transcription. Using c-fos promoter-CAT constructs and electrophoretic mobility shift assay (EMSA), the minimal c-fos promoter activity was shown to increase within 30-min exposure to serum C5b-9, which also induced the binding of serum response factor (SRF), along with ternary complex factor (TCF) Elk1 and Sap1a to the serum response element. C5b-9 activated ERK1, which in turn activated Elk1 in myotubes. We propose that c-fos gene transcription associated with myotube dedifferentiation is induced by C5b-9 through ERK1-mediated assembly of serum response factor-ternary complex.


Subject(s)
Complement Membrane Attack Complex/physiology , Gene Expression Regulation/immunology , Genes, fos/immunology , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Transcriptional Activation/immunology , Adult , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , DNA/metabolism , DNA-Binding Proteins/metabolism , Enzyme Activation/genetics , Enzyme Activation/immunology , Humans , Lymphoid Enhancer-Binding Factor 1 , Mice , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Protein Binding/genetics , Protein Binding/immunology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Response Elements/immunology , Serum Response Factor/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Transcription Factors/metabolism , ets-Domain Protein Elk-1
9.
Am J Pathol ; 163(3): 1069-80, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12937147

ABSTRACT

Activation of the classical complement system is known to play a central role in autoimmune demyelination. We have analyzed the role of complement component C5 in experimental autoimmune encephalomyelitis (EAE) using C5-deficient (C5-d) and C5-sufficient (C5-s) mice. Both groups of mice displayed early onset EAE, a short recovery phase, and similar stable chronic courses. However, in contrast to the clinical similarities, marked differences were apparent by histopathology. During acute EAE in C5-d, a delay in inflammatory cell infiltration and tissue damage was observed along with restricted lesion areas, which in C5-s mice were more extensive and diffuse. More striking were the differences in chronic lesions. In C5-d mice, inflammatory demyelination and Wallerian degeneration were followed by axonal depletion and severe gliosis, while in C5-s, the same initial signs were followed by axonal sparing and extensive remyelination. In C5-d, immunohistochemistry and Western blotting showed an increase in glial fibrillary acidic protein and a decrease in neurofilament protein, proteolipid protein, and several pro-inflammatory markers. These results in the EAE model indicate that absence of C5 resulted in fiber loss and extensive scarring, whereas presence of C5-favored axonal survival and more efficient remyelination.


Subject(s)
Complement C5/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Gliosis/prevention & control , Myelin Sheath , Regeneration , Animals , Animals, Outbred Strains , Astrocytes/pathology , Biomarkers , Brain/pathology , Brain/ultrastructure , Cell Adhesion Molecules/metabolism , Complement C5/deficiency , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gliosis/pathology , Hemolysis , Mice , Nerve Fibers/pathology , RNA, Messenger/metabolism , Spinal Cord/pathology , Spinal Cord/ultrastructure , Wound Healing
10.
J Neurotrauma ; 20(2): 207-19, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12675973

ABSTRACT

We showed previously that, after spinal cord injury (SCI), tumor necrosis factor-alpha (TNF-alpha) may serve as an external signal, initiating apoptosis in neurons and oligodendrocytes. To further characterize the apoptotic cascade initiated by TNF-alpha after SCI, we examined the expression of TNF-alpha, inducible nitric oxide (NO) synthase (iNOS), and the level of NO after SCI. Western blots and reverse transcription polymerase chain reactions showed an early upregulation of TNF-alpha after injury. A peak TNF-alpha expression was observed within 1 h of injury. By 4 h after injury, the expression of iNOS and the level of NO were markedly increased in the injured spinal cord. Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end labeling (TUNEL)-positive cells were also first observed in the lesioned area 4 h after SCI. The largest number of TUNEL-positive cells was observed between 24-48 h after SCI. Injecting a neutralizing antibody against TNF-alpha into the lesion site after injury significantly reduced the expression of iNOS, the level of NO and the number of TUNEL-positive cells in the injured spinal cord. Injecting the NOS inhibitors, N(G)-monomethyl-L-arginine monoacetate and S-methylisothiourea sulfate, or an NO scavenger, carboxy-PTIO, into the lesion site also significantly reduced the level of NO and the degree of DNA laddering in the injured spinal cord. These data suggest that after SCI, apoptosis induced by TNF-alpha may be mediated in part by NO via upregulation of iNOS, induced in response to TNF-alpha.


Subject(s)
Apoptosis/physiology , Spinal Cord Injuries/physiopathology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Antibodies/pharmacology , Apoptosis/drug effects , Enzyme Induction , Male , Nerve Crush , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology , Time Factors , Tumor Necrosis Factor-alpha/immunology
11.
Ann N Y Acad Sci ; 1010: 530-3, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15033785

ABSTRACT

Complement activation is involved in the initiation of inflammation and antibody-mediated demyelination in experimental autoimmune encephalomyelitis (EAE). We investigated the role of MAC in apoptosis in myelin-induced EAE in complement C5-deficient (C5-d) and C5-sufficient (C5-s) mice. The number of apoptotic cells assessed by TUNEL assay was significantly increased in C5-d mice during clinical recovery as compared with C5-s mice. Most of the apoptotic cells were lymphocytes, monocytes, and oligodendrocytes. DNA microarray was performed using total RNA extracted from spinal cords. Genes expressed higher in C5-s included members of the caspase (caspase 6, 7), TNF and TNFR families (CD27, FasL, lymphotoxin-beta R) and survivin. These results indicate that C5 and possibly MAC may be required for the limitation of inflammatory response within the central nervous system.


Subject(s)
Apoptosis/physiology , Complement Membrane Attack Complex/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Animals , Complement C5/deficiency , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , In Situ Nick-End Labeling , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA/genetics , RNA/isolation & purification , Spinal Cord
12.
J Biol Chem ; 277(1): 502-8, 2002 Jan 04.
Article in English | MEDLINE | ID: mdl-11687586

ABSTRACT

Proliferation of aortic smooth muscle cells contributes to atherogenesis and neointima formation. Sublytic activation of complement, particularly C5b-9, induces cell cycle progression in aortic smooth muscle cells. RGC-32 is a novel protein that may promote cell cycle progression in response to complement activation. We cloned human RGC-32 cDNA from a human fetal brain cDNA library. The human RGC-32 cDNA encodes a 117-amino acid protein with 92% similarity to the rat and mouse protein. Human RGC-32 maps to chromosome 13 and is expressed in most tissues. Sublytic complement activation enhanced RGC-32 mRNA expression in human aortic smooth muscle cells and induced nuclear translocation of the protein. RGC-32 was physically associated with cyclin-dependent kinase p34CDC2 and increased the kinase activity in vivo and in vitro. In addition, RGC-32 was phosphorylated by p34CDC2-cyclin B1 in vitro. Mutation of RGC-32 protein at Thr-91 prevented the p34CDC2-mediated phosphorylation and resulted in loss of p34CDC2 kinase enhancing activity. Overexpression of RGC-32 induced quiescent aortic smooth muscle cells to enter S-phase. These data indicate that cell cycle activation by C5b-9 may involve p34CDC2 activity through RGC-32. RGC-32 appears to be a cell cycle regulatory factor that mediates cell proliferation, both as an activator and substrate of p34CDC2.


Subject(s)
Aorta/cytology , CDC2 Protein Kinase/biosynthesis , Cell Cycle Proteins , Muscle Proteins , Muscle, Smooth, Vascular/cytology , Nerve Tissue Proteins/physiology , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Cloning, Molecular , Complement System Proteins/physiology , Humans , Mice , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Phosphorylation , RNA, Messenger/analysis , S Phase
SELECTION OF CITATIONS
SEARCH DETAIL