Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Clin Exp Immunol ; 198(3): 359-366, 2019 12.
Article in English | MEDLINE | ID: mdl-31461782

ABSTRACT

The complement system is now a therapeutic target for the management of serious and life-threatening conditions such as paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, glomerulonephritis and other diseases caused by complement deficiencies or genetic variants. As complement therapeutics expand into more clinical conditions, monitoring complement activation is increasingly important, as is the baseline levels of complement activation fragments in blood or other body fluid levels. Although baseline complement levels have been reported in the literature, the majority of these data were generated using non-standard assays and with variable sample handling, potentially skewing results. In this study, we examined the plasma and serum levels of the soluble membrane attack complex of complement (sMAC). sMAC is formed in the fluid phase when complement is activated through the terminal pathway. It binds the regulatory proteins vitronectin and/or clusterin and cannot insert into cell membranes, and can serve as a soluble diagnostic marker in infectious disease settings, as previously shown for intraventricular shunt infections. Here we show that in healthy adults, serum sMAC levels were significantly higher than those in plasma, that plasma sMAC levels were similar between in African Americans and Caucasians and that plasma sMAC levels increase with age. Plasma sMAC levels were significantly higher in virally suppressed people living with HIV (PLWH) compared to non-HIV infected healthy donors. More specifically, PLWH with CD4+ T cell counts below 200 had even greater sMAC levels, suggesting diagnostic value in monitoring sMAC levels in this group.


Subject(s)
Complement Activation/immunology , Complement Membrane Attack Complex/immunology , HIV Infections/immunology , Immune Reconstitution/immunology , Adult , Atypical Hemolytic Uremic Syndrome/blood , Atypical Hemolytic Uremic Syndrome/diagnosis , Atypical Hemolytic Uremic Syndrome/immunology , Biomarkers/blood , Clusterin/blood , Clusterin/immunology , Complement Membrane Attack Complex/genetics , Complement Membrane Attack Complex/metabolism , Female , HIV Infections/blood , HIV Infections/metabolism , Hemoglobinuria, Paroxysmal/blood , Hemoglobinuria, Paroxysmal/diagnosis , Hemoglobinuria, Paroxysmal/immunology , Humans , Male , Vitronectin/blood , Vitronectin/immunology , Young Adult
2.
Transfus Med ; 24(2): 114-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24588971

ABSTRACT

OBJECTIVE: To determine if the complement system, a potent mediator of inflammation, contributes to haemolysis during red blood cell (RBC) storage. BACKGROUND: RBCs in storage undergo structural and biochemical changes that may result in adverse patient outcomes post-transfusion. Complement activation on leukodepletion and during storage may contribute to the RBC storage lesion. METHODS/MATERIALS: We performed a cross-sectional analysis of aliquots of leukoreduced RBC units, stored for 1-6 weeks, for the levels of C3a, C5a, Bb, iC3b, C4d and C5b-9 [membrane attack complex (MAC)] by enzyme-linked immunosorbent assay (ELISA). RESULTS: We observed that only MAC levels significantly increased in RBC units as a function of storage time. We also observed that the level of C5b-9 bound to RBCs increased as a function of storage time. CONCLUSION: MAC levels increased over time, suggesting that MAC is the primary complement-mediated contributor to changes in stored RBCs. Inhibition of the terminal complement pathway may stabilise RBC functionality and extend shelf life.


Subject(s)
Blood Preservation , Complement Membrane Attack Complex/metabolism , Erythrocytes/cytology , Erythrocytes/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Time Factors
3.
Parasite Immunol ; 34(11): 547-50, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22882618

ABSTRACT

Complement receptors for C3-derived fragments (CR1-4) play critical roles in innate and adaptive immune responses. Of these receptors, CR3 and CR4 are important in binding and phagocytosis of complement-opsonized pathogens including parasites. The role of CR3 and CR4 in malaria or in cerebral malaria (CM) has received little attention and remains poorly understood in both human disease and rodent models of malaria. CR3 and CR4 are members of the ß(2) -integrin family of adhesion molecules and are expressed on all leucocytes that participate in the development of CM, most importantly as it relates to parasite phagocytosis (monocytes/macrophages) and antigen processing and presentation (dendritic cells). Thus, it is possible that these receptors might play an important role in disease development. To address this question, we examined the role of CR3(-/-) and CR4(-/-) in experimental cerebral malaria (ECM). We found that both CR3(-/-) and CR4(-/-) mice were fully susceptible to ECM and developed disease comparable to wild-type mice. Our results indicate that CR3 and CR4 are not critical to the pathogenesis of ECM despite their role in elimination of complement-opsonized pathogens. These findings support recent studies indicating the importance of the terminal complement pathway and the membrane attack complex in ECM pathogenesis.


Subject(s)
Disease Susceptibility , Integrin alphaXbeta2/genetics , Macrophage-1 Antigen/genetics , Malaria, Cerebral/genetics , Malaria, Cerebral/immunology , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Sequence Deletion
4.
Parasite Immunol ; 34(8-9): 444-7, 2012.
Article in English | MEDLINE | ID: mdl-22708514

ABSTRACT

Complement contributes to inflammation during pathogen infections; however, less is known regarding its role during malaria and in the severest form of the disease, cerebral malaria. Recent studies have shown that deletion of the complement anaphylatoxins receptors, C3aR and C5aR, does not alter disease susceptibility in experimental cerebral malaria (ECM). This does not, however, preclude C3a- and C5a-mediated contributions to inflammation in ECM and raises the possibility that carboxypeptidase regulation of anaphylatoxin activity rapidly over rides their functions. To address this question, we performed ECM using carboxypeptidase N-deficient (CPN(-/-)) mice. Unexpectedly, we found that CPN(-/-) mice survived longer than wild-type mice, but they were fully susceptible to ECM. CD4(+) and CD8(+) T cell infiltration was not reduced at the peak of disease in CPN(-/-) mice, and there was no corresponding reduction in pro-inflammatory cytokine production. Our results indicate that carboxypeptidases contribute to the pathogenesis of ECM and that studies examining the contribution of other carboxypeptidase families and family members may provide greater insight into the role these enzymes play in malaria.


Subject(s)
Lysine Carboxypeptidase/deficiency , Lysine Carboxypeptidase/metabolism , Malaria, Cerebral/pathology , Malaria, Cerebral/parasitology , Animals , Brain/immunology , Brain/pathology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Mice , Mice, Knockout , Survival Analysis , Time Factors
5.
Arch Microbiol ; 189(4): 357-66, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18283436

ABSTRACT

Heterocyst differentiation in cyanobacteria is accompanied by developmentally regulated DNA rearrangements that occur within the nifD, fdxN, and hupL genes. These genetic elements are excised from the genome by site-specific recombination during the latter stages of differentiation. The nifD element is excised by the recombinase, XisA, located within the element. Our objective was to examine the XisA-mediated excision of the nifD element. To accomplish this, we observed the ability of XisA to excise substrate plasmids that contained the flanking regions of the nifD element in an E. coli host. Using PCR directed mutagenesis, nucleotides in the nifD element flanking regions in substrate plasmids were altered and the effect on recombination was determined. Results indicate that only certain nucleotides within and surrounding the direct repeats are involved in excision. In some nucleotide positions, the presence of a purine versus a pyrimidine greatly affected recombination. Our results also indicated that the site of excision and branch migration occurs in a 6 bp region within the direct repeats.


Subject(s)
Nitrogenase/chemistry , Nostoc/genetics , Recombinases/metabolism , Recombination, Genetic , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Escherichia coli/genetics , Nitrogenase/genetics , Nostoc/chemistry , Nostoc/metabolism , Plasmids/chemistry , Plasmids/genetics , Recombinases/genetics
6.
Brain Res ; 894(2): 321-6, 2001 Mar 16.
Article in English | MEDLINE | ID: mdl-11251209

ABSTRACT

Expression of the C5a receptor in the central nervous system has been demonstrated on microglia, astrocytes and neurons. In the present study, we demonstrate C5aR expression in vitro by rat and murine O2-A progenitor cells and oligodendrocytes. We also observed that in vitro differentiation of O2-A progenitors into mature oligodendrocytes is accompanied by down-regulation of C5aR mRNA expression. These results suggest that the C5aR may be a marker for oligodendroglial differentiation and play a role in oligodendrocyte function.


Subject(s)
Antigens, CD/genetics , Oligodendroglia/physiology , Receptors, Complement/genetics , Animals , Cell Differentiation/physiology , Cell Lineage/physiology , Cells, Cultured , Flow Cytometry , Gene Expression/physiology , In Situ Hybridization , Oligodendroglia/cytology , RNA, Messenger/analysis , Rats , Receptor, Anaphylatoxin C5a , Stem Cells/cytology , Stem Cells/physiology
7.
J Immunol ; 165(10): 5867-73, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11067947

ABSTRACT

The exact mechanisms leading to CNS inflammation and myelin destruction in multiple sclerosis and in its animal model, experimental allergic encephalomyelitis (EAE) remain equivocal. In both multiple sclerosis and EAE, complement activation is thought to play a pivotal role by recruiting inflammatory cells, increasing myelin phagocytosis by macrophages, and exerting direct cytotoxic effects through the deposition of the membrane attack complex on oligodendrocytes. Despite this assumption, attempts to evaluate complement's contribution to autoimmune demyelination in vivo have been limited by the lack of nontoxic and/or nonimmunogenic complement inhibitors. In this report, we used mice deficient in either C3 or factor B to clarify the role of the complement system in an Ab-independent model of EAE. Both types of complement-deficient mice presented with a markedly reduced disease severity. Although induction of EAE led to inflammatory changes in the meninges and perivascular spaces of both wild-type and complement-deficient animals, in both C3(-/-) and factor B(-/-) mice there was little infiltration of the parenchyma by macrophages and T cells. In addition, compared with their wild-type littermates, the CNS of both C3(-/-) and factor B(-/-) mice induced for EAE are protected from demyelination. These results suggest that complement might be a target for the therapeutic treatment of inflammatory demyelinating diseases of the CNS.


Subject(s)
Complement C3/deficiency , Complement C3/genetics , Complement Factor B/deficiency , Complement Factor B/genetics , Demyelinating Autoimmune Diseases, CNS/genetics , Demyelinating Autoimmune Diseases, CNS/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Animals , Demyelinating Autoimmune Diseases, CNS/metabolism , Demyelinating Autoimmune Diseases, CNS/pathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Immunohistochemistry , Incidence , Mice , Mice, Inbred C57BL , Mice, Knockout , Spinal Cord/pathology , Spinal Cord/ultrastructure
8.
J Neuroimmunol ; 109(2): 164-72, 2000 Sep 22.
Article in English | MEDLINE | ID: mdl-10996218

ABSTRACT

The anaphylatoxin C5a is a potent mediator of inflammation in the CNS. We analyzed the intracerebral expression of the C5a receptor (C5aR) in a model of closed head injury (CHI) in mice. Up-regulation of C5aR mRNA and protein expression was observed mainly on neurons in sham-operated and head-injured wild-type mice at 24 h. In contrast, in TNF/lymphotoxin-alpha knockout mice, the intracerebral C5aR expression remained at low constitutive levels after sham operation, whereas it strongly increased in response to trauma between 24 and 72 h. Interestingly, by 7 days after CHI, the intrathecal C5aR expression was clearly attenuated in the knockout animals. These data show that the posttraumatic neuronal expression of the C5aR is, at least in part, regulated by TNF and lymphotoxin-alpha at 7 days after trauma.


Subject(s)
Antigens, CD/genetics , Head Injuries, Closed/immunology , Lymphotoxin-alpha/genetics , Receptors, Complement/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Antigens, CD/analysis , Antigens, CD/immunology , Brain Chemistry/immunology , Gene Expression/immunology , Head Injuries, Closed/physiopathology , In Situ Hybridization , Lymphotoxin-alpha/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/analysis , Receptor, Anaphylatoxin C5a , Receptors, Complement/analysis , Receptors, Complement/immunology , Tumor Necrosis Factor-alpha/immunology
9.
J Rheumatol ; 27(2): 402-4, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10685805

ABSTRACT

OBJECTIVE: Upregulated matrix synthesis is a hallmark of systemic sclerosis (SSc). There are indications that growth factors such as platelet derived growth factor (PDGF) are involved in proliferative pathways in SSc lesions. As activated complement releases PDGF from endothelial cells, we searched for activated complement and the complement receptor for C5a (C5aR) in skin biopsies of patients with SSc. METHODS: Snap frozen sections of 8 patients with early SSc and 5 patients with longterm SSc were examined. Using monoclonal antibodies against activated complement complex C5b-9 and the C5aR, skin biopsies derived from both clinically involved and non-involved skin were examined by APAAP immunohistochemistry. RESULTS: A pattern of activated complement C5b-9 and the CSaR could be detected in SSc microvasculature. Eleven of the 13 patients (7/8 patients with early SSc) showed positive staining for C5b-9. The CSaR was detected in 6 of the 8 patients with early SSc. In 3 patients with longterm disease, C5aR expression could also be detected in non-involved skin. CONCLUSION: Activated complement and complement receptors could be detected in early and late stages of SSc skin lesions. The presence of complement receptors in non-involved skin may indicate preclinical activation of pathways resulting in growth factor dependent matrix synthesis.


Subject(s)
Antigens, CD/metabolism , Complement C5a/metabolism , Complement Membrane Attack Complex/metabolism , Receptors, Complement/metabolism , Scleroderma, Systemic/metabolism , Skin/metabolism , Antigens, CD/immunology , Biopsy , Complement C5a/immunology , Complement Membrane Attack Complex/immunology , Humans , Receptor, Anaphylatoxin C5a , Receptors, Complement/immunology , Scleroderma, Systemic/immunology , Scleroderma, Systemic/pathology , Skin/immunology , Skin/pathology
10.
Curr Microbiol ; 40(4): 283-7, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10688700

ABSTRACT

The low molecular weight (LMW) heat shock protein (HSP), HSP16.6, in the unicellular cyanobacterium, Synechocystis sp. PCC 6803, protects cells from elevated temperatures. A 95% reduction in the survival of mutant cells with an inactivated hsp16.6 was observed after exposure for 1 h at 47 degrees C. Wild-type cell survival was reduced to only 41%. HSP16.6 is also involved in the development of thermotolerance. After a sublethal heat shock at 43 degrees C for 1 h and subsequent challenge exposure at 49 degrees C for 40 min, mutant cells did not survive, while 64% of wild-type cells survived. Ultrastructural changes in the integrity of thylakoid membranes of heat-shocked mutant cells also are discussed. These results demonstrate an important protective role for HSP16.6 in the protection of cells and, in particular, thylakoid membrane against thermal stress.


Subject(s)
Cell Membrane/metabolism , Cyanobacteria/physiology , Heat-Shock Proteins/metabolism , Cell Membrane/ultrastructure , Cyanobacteria/ultrastructure , Heat-Shock Proteins/genetics , Microscopy, Electron , Mutation , Temperature , Time Factors
11.
J Immunol ; 163(12): 6551-6, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10586048

ABSTRACT

Although generally thought of as a T cell-driven autoimmune disease, recent studies in experimental allergic encephalomyelitis (EAE), the animal model of multiple sclerosis, suggest a significant role for innate immune mechanisms. To address the possibility that the complement system plays a central role in these diseases, we developed a transgenic mouse with astrocyte-targeted production of a soluble inhibitor of complement activation, complement receptor-related protein y (sCrry). Here, we show that sCrry transgenic mice are either fully protected against EAE or develop significantly delayed clinical signs. These results indicate that complement activation may have an essential role in the pathogenesis of the disease and that complement-mediated events may occur early during the effector phase of EAE. Furthermore, this work underscores the importance of humoral immunity in amplifying a T cell-initiated pathogenic process.


Subject(s)
Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Receptors, Complement/biosynthesis , Animals , Central Nervous System/cytology , Central Nervous System/metabolism , Cerebellum/chemistry , Cerebellum/metabolism , Cerebellum/pathology , Complement C4/metabolism , DNA, Complementary/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation/immunology , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/immunology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteins , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , Promoter Regions, Genetic/immunology , Receptors, Complement/genetics , Receptors, Complement 3b
13.
Trends Neurosci ; 22(9): 397-402, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10441300

ABSTRACT

Activation of the complement system has been reported in a variety of inflammatory diseases and neurodegenerative processes of the CNS. Recent evidence indicates that complement proteins and receptors are synthesized on or by glial cells and, surprisingly, neurons. Among these proteins are the receptors for the chemotactic and anaphylactic peptides, C5a and C3a, which are the most-potent mediators of complement inflammatory functions. The functions of glial-cell C3a and C5a receptors (C3aR and C5aR) appear to be similar to immune-cell C3aRs and C5aRs. However, little is known about the roles these receptors might have on neurons. Indeed, when compared with glial cells, neurons display a distinct pattern of C3aR and C5aR expression, in either the normal or the inflamed CNS. These findings suggest unique functions for these receptors on neurons.


Subject(s)
Anaphylatoxins/metabolism , Central Nervous System Diseases/metabolism , Neurons/metabolism , Receptors, Complement/metabolism , Animals , Brain Injuries/metabolism , Complement C3a/biosynthesis , Complement C5a/biosynthesis , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Meningitis, Bacterial/metabolism , Multiple Sclerosis/metabolism , Neuroglia/metabolism , Neuronal Plasticity
14.
Glia ; 27(2): 162-70, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10417815

ABSTRACT

The expression of the murine complement regulatory protein, Crry, in the CNS remains largely unexplored. In this study, we examined murine astrocytes and microglia purified from neonatal brain and sections of adult murine brain for the expression of Crry. Using RT-PCR, immunohistochemistry, in situ hybridization, flow cytometry, and Western blot analysis, we demonstrated that astrocytes and microglia express Crry protein and RNA. Crry expression is greater on microglia than astrocytes and, as determined by Western blot analysis, each cell type expresses a Crry protein of different molecular weight. Interestingly, neuronal expression of Crry was seen only at the RNA level. These data demonstrate Crry expression by astrocytes, microglia, and neurons in the murine CNS and suggest that Crry may play an important role in protecting the CNS against complement-mediated damage.


Subject(s)
Astrocytes/metabolism , Gene Expression Regulation , Microglia/metabolism , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , Receptors, Complement/biosynthesis , Animals , Blotting, Western , Complement Activation , Flow Cytometry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Complement/genetics , Receptors, Complement 3b , Reverse Transcriptase Polymerase Chain Reaction
15.
Glia ; 26(3): 201-11, 1999 May.
Article in English | MEDLINE | ID: mdl-10340761

ABSTRACT

Little is known about the expression of the receptor for complement anaphylatoxin C3a (C3aR) in the central nervous system (CNS). In this study, we provide the first evidence that neurons are the predominant cell type expressing C3aR in the normal CNS. By using in situ hybridization (ISH) and immunohistochemistry, we found that C3aR is constitutively expressed at high levels in cortical and hippocampal neurons as well as in Purkinje cells. Moreover, we showed that primary culture of human astrocytes and microglia express the C3aR mRNA as assessed by RT-PCR. In situ hybridization performed on rat primary astrocytes confirmed the RT-PCR result demonstrating C3aR expression by astrocytes. In experimental allergic encephalitis (EAE), C3aR expression was elevated on microglia, infiltrating monocyte-macrophage cells and a few astrocytes, whereas neuronal expression remained unchanged during the course of the disease. These data demonstrate that the C3aR is expressed primarily by neurons in the normal CNS and that its neuronal expression is not dramatically upregulated under inflammation. This is in contrast to the increased neuronal expression of the C5aR in several inflammatory CNS conditions. The high constitutive expression of the C3aR by neurons suggests this receptor may play an important role in normal physiological conditions in the CNS.


Subject(s)
Anaphylatoxins/metabolism , Membrane Proteins , Neuroglia/metabolism , Neurons/metabolism , Receptors, Complement/metabolism , Animals , Autoradiography , Blotting, Southern , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
16.
J Immunol ; 162(7): 4018-23, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10201923

ABSTRACT

The anaphylatoxin C5a is a potent mediator of inflammation that exerts a broad range of activity on cells of the myeloid lineage. In this study, we present the first evidence that human T cells express the C5a receptor (C5aR) and are chemotactic to C5a. Using FACS analysis, we found that the C5aR was expressed at a low basal level on unstimulated T cells and was strikingly up-regulated upon PHA stimulation in a time- and dose-dependent manner. CD3+ sorted T cells as well as Jurkat T cells were shown to express C5aR mRNA as assessed by RT-PCR. Moreover, semiquantitative RT-PCR analysis demonstrated that C5aR mRNA was down-regulated in purified T cells upon long-term PHA stimulation. To demonstrate that C5a was biologically active on T cells, we investigated the chemotactic activity of C5a and observed that purified CD3+ T cells are chemotactic to C5a at nanomolar concentrations. Finally, using a combination of in situ hybridization and immunohistochemistry, we showed that the T cells infiltrating the central nervous system during experimental allergic encephalomyelitis express the C5aR mRNA. In summary, these results suggest that C5a exerts direct effects on T cells and could be involved in the trafficking of T cells under physiological and pathological conditions, including inflammatory diseases of the central nervous system.


Subject(s)
Antigens, CD/biosynthesis , Chemotaxis, Leukocyte/immunology , Complement C5a/pharmacology , Receptors, Complement/biosynthesis , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antigens, CD/genetics , Cell Movement/immunology , Cells, Cultured , Central Nervous System/immunology , Central Nervous System/pathology , Complement C5a/metabolism , Down-Regulation/drug effects , Down-Regulation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Jurkat Cells , Phytohemagglutinins/antagonists & inhibitors , Phytohemagglutinins/pharmacology , RNA, Messenger/biosynthesis , Rats , Rats, Inbred Lew , Receptor, Anaphylatoxin C5a , Receptors, Complement/genetics , T-Lymphocytes/pathology , Up-Regulation/drug effects , Up-Regulation/immunology
17.
J Neuroimmunol ; 91(1-2): 147-55, 1998 Nov 02.
Article in English | MEDLINE | ID: mdl-9846831

ABSTRACT

In this study, we investigated the expression of the C5aR in spinal cords of Lewis rats with experimental allergic encephalomyelitis (EAE). Using in situ hybridization (ISH) we analyzed the kinetics of C5aR at different time points of EAE (preclinical stage, clinical peak, remission phase). We observed that C5aR mRNA was readily detected in the CNS of EAE rats at all the stages of the disease. Using a combination of ISH and immunohistochemistry, we formally demonstrated that C5aR is strongly expressed on microglial cells and hypertrophic astrocytes during EAE. The potential involvement of C5a receptor in EAE physiopathology is discussed.


Subject(s)
Antigens, CD/genetics , Antigens, CD/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Receptors, Complement/genetics , Receptors, Complement/immunology , Animals , Endothelium, Vascular/chemistry , Endothelium, Vascular/immunology , Female , Gene Expression/immunology , Kinetics , Macrophages/chemistry , Macrophages/immunology , Microglia/chemistry , Microglia/immunology , Monocytes/chemistry , Monocytes/immunology , Multiple Sclerosis/immunology , RNA, Messenger/analysis , Rats , Rats, Inbred Lew , Receptor, Anaphylatoxin C5a , Spinal Cord/blood supply , Spinal Cord/cytology , Spinal Cord/immunology
18.
Curr Microbiol ; 37(6): 403-7, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9806978

ABSTRACT

The low molecular weight (LMW) heat shock protein (HSP) gene hsp16.6 was identified and cloned from the unicellular cyanobacterium Synechocystis sp. PCC 6803 through comparisons of genomic sequences and conserved gene sequences of the LMW HSPs. Hsp16.6 was isolated using PCR and cloned into the pGEMT plasmid. Hsp16.6 showed a significant increase in transcription after heat shock at 42 degreesC that indicated hsp16.6 was a heat shock gene. To determine the role that hsp16.6 plays in the heat shock response, a mutant Synechocystis cell line was generated. Cell growth and oxygen evolution rates of wild type and mutant cells were compared after heat shock. Results showed significantly decreased cell growth rates and a 40% reduction in oxygen evolution rates in mutants after heat shock treatments. These data indicate a protective role for hsp16.6 in the heat shock response.


Subject(s)
Bacterial Proteins/physiology , Cyanobacteria/chemistry , Heat-Shock Proteins/physiology , Heat-Shock Response/physiology , Amino Acid Sequence , Bacterial Proteins/genetics , Cloning, Molecular , Cyanobacteria/genetics , Cyanobacteria/growth & development , Cyanobacteria/radiation effects , Genes, Bacterial/physiology , Heat-Shock Proteins/genetics , Molecular Sequence Data , Sequence Alignment , Time Factors
19.
Glia ; 24(3): 338-45, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9775985

ABSTRACT

Evidence from several central nervous system (CNS) inflammatory disease models suggests that intrathecal complement synthesis may contribute to early inflammatory events in the brain. In this study, we examined the expression of the receptor for C5a (C5aR), a potent inflammatory and chemotactic factor, in the brains of transgenic mice with constitutive astrocyte expression of interleukin-3 (IL-3), a hematopoietic and immunomodulatory cytokine. By in situ hybridization, we demonstrated that cells infiltrating the cerebellar meninges, the cerebellum, and demyelinating lesions in the cerebellum were strongly positive for C5aR mRNA. By immunohistochemistry, the infiltrating cells expressing the C5aR were identified as macrophages based on staining with antibodies to the complement receptor type 3 and F4/80, a mouse macrophage-specific marker. In addition, some of the cells in cerebellar lesions were positive for the astrocyte-specific marker, glial fibrillary acidic protein, suggesting that a subpopulation of astrocytes in these lesions express elevated levels of the C5aR. Increased C5aR expression was also observed in cortical neurons in the occipital cortex and in pyramidal neurons in the cornu ammonis and subiculum of the hippocampus, at both the protein and mRNA levels. These data suggest that IL-3 may play an immunomodulatory role in C5aR expression on several cell types in the brain and that increased C5aR expression correlates with the pathology seen in this model. The transgenic mice used in this study provide a useful tool for characterizing the mechanism of regulation of the C5aR expression and for examining the functions of this chemotactic receptor in CNS inflammation.


Subject(s)
Antigens, CD/biosynthesis , Astrocytes/physiology , Complement C5a/metabolism , Interleukin-3/biosynthesis , Neuroglia/metabolism , Neurons/metabolism , Receptors, Complement/biosynthesis , Animals , Astrocytes/metabolism , Cerebellum/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Hippocampus/metabolism , Immunohistochemistry , In Situ Hybridization , Interleukin-3/genetics , Macrophages/metabolism , Meninges/metabolism , Mice , Mice, Transgenic , Microglia/metabolism , RNA, Messenger/metabolism , Receptor, Anaphylatoxin C5a
20.
Mol Immunol ; 35(1): 1-11, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9683259

ABSTRACT

Previously, we reported that TGF-beta2 regulates the C3 gene expression in a dose- and time-dependent manner in monocytes. To extend these studies, we examined the role of PKC in the TGF-beta2-mediated induction of C3 expression by the human monocyte cell line, U937. Treatment of U937 cells with the PKC inhibitors, H7 and calphostin C, suppressed TGF-beta2-mediated induction of C3 protein levels, but not mRNA levels, in a dose-dependent manner. At the highest concentrations of H7 and calphostin C, C3 protein levels were inhibited 50% and 93%, respectively, compared to control levels. Treatment of U937 cells with HA1004, a weak PKC inhibitor used as a control for H7, did not inhibit induction of C3 protein levels. Down-modulating PKC with a prolonged exposure of U937 cells to PMA also suppressed TGF-beta2-mediated C3 protein induction by as much as 82%. Incubating cell extracts isolated from TGF-beta2-treated U937 cells with the PKC substrate, MIBP(4-14), resulted in increased substrate phosphorylation compared to cell extracts isolated from untreated cells. Addition of calphostin C suppressed the increased substrate phosphorylation by TGF-beta2. Furthermore, biosynthetic labeling of U937 cells treated with TGF-beta2 and calphostin C demonstrated an accumulation of C3 protein within cell lysates compared to controls. Collectively, these studies suggest a role for PKC in the secretion of C3 protein during TGF-beta2-mediated regulation of C3 expression in U937 cells.


Subject(s)
Complement C3/metabolism , Monocytes/immunology , Protein Kinase C/metabolism , Sulfonamides , Transforming Growth Factor beta/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Drug Interactions , Humans , Inflammation/immunology , Isoenzymes/metabolism , Isoquinolines/pharmacology , Naphthalenes/pharmacology , Protein Kinase C/antagonists & inhibitors , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...