Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
2.
Gene Ther ; 16(3): 340-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19112449

ABSTRACT

The effectiveness of genetic engineering with lentivectors to protect transplanted cells from allogeneic rejection was examined using, as a model, type 1 diabetes treatment with beta-cell transplantation, whose widespread use has been limited by the requirement for sustained immunosuppressive treatment to prevent graft rejection. We examined whether lentivectors expressing select immunosuppressive proteins encoded by the adenoviral genome early region 3 (AdE3) would protect transplanted beta-cells from an alloimmune attack. The insulin-producing beta-cell line beta TC-tet (C3HeB/FeJ-derived) was transduced with lentiviruses encoding the AdE3 proteins gp19K and RID alpha/beta. The efficiency of lentiviral transduction of beta TC-tet cells exceeded 85%. Lentivector expression of gp19K decreased surface class I major histocompatibility complex expression by over 90%, whereas RID alpha/beta expression inhibited cytokine-induced Fas upregulation by over 75%. beta TC-tet cells transduced with gp19K and RID alpha/beta lentivectors, but not with a control lentivector, provided prolonged correction of hyperglycemia after transplantation into diabetic BALB/c severe combined immunodeficient mice reconstituted with allogeneic immune effector cells or into diabetic allogeneic BALB/c mice. Thus, genetic engineering of beta-cells using gp19K- and RID alpha/beta-expressing lentiviral vectors may provide an alternative that has the potential to eliminate or reduce treatment with the potent immunosuppressive agents necessary at present for prolonged engraftment with transplanted islets.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Genetic Engineering/methods , Graft Rejection/prevention & control , Insulin-Secreting Cells/immunology , Islets of Langerhans Transplantation/methods , Adenovirus E3 Proteins/genetics , Adenovirus E3 Proteins/immunology , Adenovirus Early Proteins/genetics , Adenovirus Early Proteins/immunology , Animals , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Female , Genetic Vectors , Graft Rejection/immunology , Immune Tolerance , Lentivirus/genetics , Mice , Mice, Inbred C3H , Mice, Inbred Strains , Reverse Transcriptase Polymerase Chain Reaction/methods , Transduction, Genetic
4.
Curr Top Microbiol Immunol ; 273: 113-35, 2004.
Article in English | MEDLINE | ID: mdl-14674600

ABSTRACT

Adenoviruses contain genes that have evolved to control the host immune and inflammatory responses; however, it is not clear whether these genes function primarily to facilitate survival of the virus during acute infection or during its persistent phase. These issues have assumed greater importance as the use of adenoviruses as vectors for gene therapy has been expanded. This review will focus on the mechanism of immune evasion mediated by the proteins encoded within the early region 3 (E3) transcription region, which affect the functions of a number of cell surface receptors including Fas, intracellular cell signaling events involving NF-kappaB, and the secretion of pro-inflammatory molecules such as chemokines. The successful use of E3 genes in facilitating allogeneic transplantation and in preventing autoimmune diabetes in several transgenic mouse models will also be described.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae/physiology , Adenovirus E3 Proteins/immunology , Adenoviridae/immunology , Adenoviridae Infections/virology , Adenovirus E3 Proteins/genetics , Adenovirus E3 Proteins/physiology , Animals , Autoimmune Diseases/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Gene Expression Regulation , Humans
5.
Gene Ther ; 9(15): 981-90, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12101428

ABSTRACT

Host immune responses limit the duration of expression of transgenes introduced by recombinant adenoviruses, preclude gene transfer upon vector readministration and cause liver injury. CTLA4Ig inhibits immune response by blocking the co-stimulatory interaction between CD28 on T cells and B7 on antigen-presenting cells. We have constructed a recombinant adenovirus, Ad-hUGT1A1-CTLA4Ig that coexpresses human bilirubin-uridinediphosphoglucuronate glucuronosyltransferase (hUGT1A1) and soluble murine CTLA4Ig, both driven by CMV immediate-early promoters. After intravenous injection of this vector (6 x 10(11) p.f.u.) into UGT1A1-deficient jaundiced Gunn rats, serum CTLA4Ig levels peaked at 1.8-2.0 mg/ml on day 7 and declined thereafter to 0.2 mg/ml by day 180. Serum bilirubin declined from mean preinjection levels of 8.0 mg/dl to 0.48-0.6 mg/dl in 3 days, remained normal for 28 weeks, and then gradually increased to 8 mg/dl by day 350. A second injection of Ad-hUGT1A1-CTLA4Ig normalized serum bilirubin. In two rats in this group that were followed longer, serum bilirubin increased to 3.1 and 3.5 mg/dl in 40 weeks, but was normalized again after a third injection. The antibody and cytotoxic lymphocyte (CTL) responses were negligible, and liver biopsy showed no inflammatory cell infiltration. Rats receiving a tertiary challenge with Ad-LacZ (expressing E. coli beta-galactosidase) (5 x 10(11) p.f.u.), 2 months after the second dose of Ad-hUGT1A1-CTLA4Ig, showed beta-galactosidase expression in over 80% of hepatocytes. In contrast, after Ad-hUGT1A1 (which expresses UGT1A1 alone) injection, serum bilirubin remained normal for only 4 weeks, and returned to preinjection levels by day 120. Bilirubin levels did not decline upon reinjection, and beta-galactosidase was not expressed after Ad-LacZ. High levels of adenovirus-specific antibodies and CTL, and hepatic inflammation were found. This is the first demonstration that coexpression of CTLA4Ig permits prolonged expression and repeatable gene transfer by an adenoviral vector.


Subject(s)
Adenoviridae/genetics , Antigens, Differentiation/genetics , Crigler-Najjar Syndrome/therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Immunoconjugates , Abatacept , Animals , Antigens, CD , Bilirubin/genetics , Bilirubin/metabolism , CTLA-4 Antigen , Female , Gene Expression , Genetic Vectors/genetics , Glucuronosyltransferase/genetics , Humans , Liver/metabolism , Male , Mice , Rats , Rats, Gunn , Time Factors , Transgenes
6.
Biochem Biophys Res Commun ; 285(2): 555-60, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11444880

ABSTRACT

FIP-3 (NEMO/IKKgamma) is an essential modulator of the activity of NF-kappaB by mechanisms that include alterations in the phosphorylation, ubiquination, and degradation of IkappaBalpha. The multiple protein-protein interactions of FIP-3 (NEMO/IKKgamma) in a high molecular weight IKK complex indicated that this protein may be a link between some of the receptor-proximal upstream signal transduction molecules such as RIP and the downstream effects on IkappaBalpha. Although FIP-3 (NEMO/IKKgamma) has no intrinsic kinase activity, it has been shown to increase the kinase activity of IKKbeta. In this manuscript, the results of serine to alanine mutations at five sites on FIP-3 (NEMO/IKKgamma) are described, and functional assays demonstrated that two of these mutants affect both the phosphorylation and kinase activity of IKKbeta. Protein kinase Calpha appeared to be the kinase that was required for the posttranslational modification of FIP-3 (NEMO/IKKgamma). One of the serine targets of the protein kinase Calpha enzyme at amino acid 141 was within a leucine zipper-like sequence of FIP-3 (NEMO/IKKgamma), which might affect its interactions with other proteins on the signal transduction pathway. The second serine, which augmented the inhibition, was at amino acid 85 within the FIP-3 (NEMO/IKKgamma) interaction site with IKKbeta. When both serines were mutated simultaneously, the effect on IKKbeta and IkappaBalpha phosphorylation was more profoundly affected.


Subject(s)
Isoenzymes/metabolism , NF-kappa B/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Alanine , Amino Acid Sequence , Amino Acid Substitution , Cell Line , Humans , I-kappa B Kinase , Leucine Zippers , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Point Mutation , Protein Kinase C-alpha , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serine , Transfection
7.
Transplantation ; 71(6): 801-11, 2001 Mar 27.
Article in English | MEDLINE | ID: mdl-11330546

ABSTRACT

BACKGROUND: Hepatocyte cell lines are beginning to be developed as universal donors for isolated liver cell transplantation, which is a less invasive method than orthotopic liver transplantation for treatment of metabolic liver disease. The immune response to isolated liver cell transplantation and its modification by costimulatory blockade are as yet not well delineated. METHODS: Adenovirus expressing CTLA4Ig was used to study blockade of the costimulatory CD28/B7 pathway in murine models of hepatocyte transplantation, and the effects on alloreactive T and B cells were studied. RESULTS: CTLA4Ig delayed rejection of subcutaneously administered C57L-derived murine hepatoma cells in CBA/J recipients for >50 days. Activation and cytokine secretion by allospecific CD4+ and CD8+ T cells were initially blocked by CTLA4Ig; delayed rejection was associated with tumor infiltration by CD8+ T cells that did not secrete interferon-gamma. CTLA4Ig failed to block transplant rejection in primed mice, indicating that memory effector T cells were resistant to its action. In contrast, CTLA4Ig suppressed both naive and memory alloreactive B cells. High levels of CTLA4Ig mediated acceptance of hepatoma cells delivered directly into the spleen. However, isolated primary C57BL/6 mouse hepatocytes delivered into the spleen were rejected with only moderately delayed kinetics. CONCLUSIONS: Transplant antigenicity, transplant site, and CTLA4Ig dose all affected the survival of transplanted liver cells. CD8+ T cells are significant mediators of hepatocyte transplant rejection and are relatively resistant to costimulatory blockade with CTLA4Ig. Strategies to specifically antagonize CD8+ T cells or to modulate MHC class I expression in association with costimulatory blockade by CTLA4Ig may enhance the clinical feasibility of transplanting allogeneic hepatocytes.


Subject(s)
B7-1 Antigen/pharmacology , CD28 Antigens/pharmacology , Hepatocytes/transplantation , Immunoconjugates , Abatacept , Animals , Antigens, CD , Antigens, Differentiation/blood , Antigens, Differentiation/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/physiology , CTLA-4 Antigen , Graft Rejection , Immunoglobulin G/blood , Immunoglobulin G/pharmacology , Isoantigens/immunology , Liver Transplantation/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, SCID , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Transplantation, Homologous/immunology , Tumor Cells, Cultured/immunology , Tumor Cells, Cultured/transplantation
8.
J Autoimmun ; 16(3): 211-7, 2001 May.
Article in English | MEDLINE | ID: mdl-11334485

ABSTRACT

The induction of autoimmunity by viruses has been attributed to numerous mechanisms. Coxsackievirus B4 (CB4) induces insulin-dependent diabetes mellitus (IDDM) in mice resembling the final step of disease progression in humans. Following viral infection, autoreactive lymphocytes are activated through exposure to damaged islets consequently precipitating IDDM. However, the viral and host requirements leading up to this final step have yet to be elucidated. We provide evidence that disease induction requires a pre-existing accumulation of beta-cell specific autoreactive T cells within the pancreas, as well as the infection of islet beta-cells. Therefore, the primary role of CB4 in the development of IDDM is to infect tissue, resulting in the presentation of sequestered islet antigen, the stimulation of preexisting autoreactive T cells, and the initiation of disease.


Subject(s)
Coxsackievirus Infections/immunology , Diabetes Mellitus, Type 1/virology , Enterovirus B, Human/immunology , Islets of Langerhans/immunology , Animals , Diabetes Mellitus, Type 1/immunology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic
9.
Virology ; 281(2): 205-15, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11277693

ABSTRACT

Coxsackieviral infections have been linked etiologically to multiple diseases. The serotype CB4 is associated with acute pancreatitis and autoimmune type 1 diabetes. To delineate the mechanisms of host survival after an acute infection with CB4 (strain E2), we have investigated the role of nitric oxide (NO), generated by the inducible form of nitric oxide synthase (NOS2), in viral clearance and pancreatic beta-cell maintenance. Mice deficient in NOS2 (NOS2-/- mice) and their wild-type (wt) counterparts were injected with CB4, after which both groups developed severe pancreatitis, hepatitis, and hypoglycemia within 3 days. Within 4 to 7 days postinfection (p.i.), most of the NOS2-/- mice died and at a strikingly higher mortality rate than wt mice. Histological examination of pancreata from both infected NOS2-/- and infected wt mice revealed early and complete destruction of the pancreatic acinar tissue, but intact, insulin-stained islets. When examined up to 8 weeks p.i., neither surviving NOS2-/-mice nor surviving wt mice developed hyperglycemia. However, the clearance of infectious CB4 was different between the mice. The spleens of NOS2-/- survivors were cleared of infectious virus with kinetics similar to that of wt mice, but the livers, pancreata, kidneys, and hearts of the NOS2-/- groups cleared virus more slowly than those of the wt group. This delayed clearance was particularly prominent in the livers of infected NOS2-/- mice, which also showed prolonged histopathological features of viral hepatitis. Taken together, this outcome suggests that NOS2 (and NO) is not required for the prevention of pancreatic beta-cell depletion after CB4 infection. Instead the critical actions of NOS2 apparently occur early in the host immune response, allowing mice to survive and clear virus. Moreover, the data support the existence of an organ-specific dependency on NO for a rapid clearance of CB4.


Subject(s)
Coxsackievirus Infections/virology , Enterovirus/pathogenicity , Nitric Oxide Synthase/physiology , Animals , Coxsackievirus Infections/enzymology , Coxsackievirus Infections/mortality , Enterovirus/isolation & purification , Female , Hepatitis/enzymology , Hepatitis/pathology , Hypoglycemia/enzymology , Hypoglycemia/pathology , Immunohistochemistry , Islets of Langerhans/enzymology , Islets of Langerhans/pathology , Liver/pathology , Liver/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Pancreas/pathology , Pancreas/virology , Pancreatitis/enzymology , Pancreatitis/pathology , Time Factors , Viral Plaque Assay , Virus Replication , Viscera/virology
11.
Nat Cell Biol ; 3(1): 1-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11146619

ABSTRACT

DAP kinase is a pro-apoptotic calcium-regulated serine/threonine kinase, whose expression is frequently lost in human tumours. Here we show that DAP kinase counteracts oncogene-induced transformation by activating a p19ARF/p53-dependent apoptotic checkpoint. Ectopic expression of DAP kinase suppressed oncogenic transformation of primary embryonic fibroblasts by activating p53 in a p19ARF-dependent manner. Consequently, the fibroblasts underwent apoptosis, characterized by caspase activation and DNA fragmentation. In response to c-Myc or E2F-1, the endogenous DAP kinase protein was upregulated. Furthermore, functional or genetic inactivation of the endogenous DAP kinase reduced the extent of induction of p19ARF/p53 and weakened the subsequent apoptotic responses to c-Myc or E2F-1. These results establish a role for DAP kinase in an early apoptotic checkpoint designed to eliminate pre-malignant cells during cancer development.


Subject(s)
Apoptosis/genetics , Calcium-Calmodulin-Dependent Protein Kinases/deficiency , Carrier Proteins , Cell Cycle Proteins , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins , Genes, Tumor Suppressor/physiology , Genes, cdc/physiology , Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis Regulatory Proteins , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Division/genetics , Cell Line, Transformed/cytology , Cell Line, Transformed/enzymology , Cell Transformation, Neoplastic/genetics , Death-Associated Protein Kinases , E2F Transcription Factors , E2F1 Transcription Factor , Fetus , Fibroblasts/cytology , Fibroblasts/enzymology , Gene Expression Regulation, Neoplastic/physiology , Genes, myc/physiology , Mice , Mice, Knockout , Oncogenes/physiology , Proteins/genetics , Retinoblastoma-Binding Protein 1 , Signal Transduction/genetics , Transcription Factor DP1 , Transcription Factors/genetics , Tumor Suppressor Protein p14ARF , Tumor Suppressor Protein p53/genetics
12.
Nat Med ; 6(6): 693-7, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10835688

ABSTRACT

Cardiovascular disease is one of the leading causes of death worldwide, and has been associated with many environmental risk factors. Recent evidence has indicated the involvement of pathogens such as viruses as causative agents, and specifically identified the coxsackievirus B serogroup as the leading culprit. Not only has coxsackievirus B3 (CB3) been identified from patients with cardiovascular disease, but also infection of mice with CB3 strains can reproduce human clinical heart disease in rodents. Several mechanisms have been proposed in an attempt to distinguish between pathology mediated by direct viral destruction of cardiac muscle cells or by the virus-induced immune response directed at infected myocytes or at 'mimicked' epitopes shared between viral and cardiac antigens. To distinguish between these mechanisms, we infected a unique mouse that diminishes the extent of infection and spread of the virus, but allows complete immunity to the virus. Transgenic mice expressing interferon-gamma in their pancreatic beta cells failed to develop CB-3-induced myocarditis. This work challenges the idea of the function of the immune response and 'molecular mimicry' in the CB-3-induced autoimmune myocarditis model, and instead favors the idea of virus-mediated damage. These results emphasize the benefit of reducing the level of viremia early during infection, thereby reducing the incidence of virus-mediated heart damage and autoimmunity.


Subject(s)
Coxsackievirus Infections/immunology , Enterovirus B, Human/immunology , Interferon-gamma/immunology , Myocarditis/immunology , Pancreas/immunology , Animals , Autoantibodies/immunology , Coxsackievirus Infections/pathology , Coxsackievirus Infections/virology , Disease Models, Animal , Gene Expression , HeLa Cells , Heart/virology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Mice , Mice, Inbred NOD , Mice, Transgenic , Myocarditis/pathology , Myocarditis/virology , Myocardium/immunology , Myocardium/metabolism , Myocardium/pathology , Myosins/immunology , Pancreas/metabolism , Pancreas/pathology
13.
J Virol ; 74(10): 4705-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10775608

ABSTRACT

Adenoviruses (Ad) code for immunoregulatory and cytokine regulatory proteins, one of which is the early region 3, 14.7-kDa protein (Ad E3-14.7K), which has been shown to inhibit tumor necrosis factor alpha-induced apoptosis. In an effort to understand the mechanism of action of Ad E3-14.7K, we previously searched for cell proteins with which it interacted. Three Ad E3-14.7K-interacting proteins (FIP-1, -2, and -3) were isolated. FIP-1 is a small GTPase which was used in this report as bait in the yeast two-hybrid system to find other interacting cell targets. The search resulted in the isolation of a protein, which we called GIP-1 (GTPase-interacting protein) that subsequently was shown to be identical to one of the light-chain components of human dynein (TCTEL1). FIP-1 was able to bind both TCTEL1 and Ad E3-14.7K simultaneously and was necessary to form a complex in which the viral protein was associated with a microtubule-binding motor protein. The functional significance of these interactions is discussed with respect to the steps of the Ad life cycle which are microtubule associated.


Subject(s)
Adenovirus E3 Proteins/metabolism , Apoptosis , Dyneins/metabolism , GTP Phosphohydrolases/metabolism , Microtubule-Associated Proteins , Nuclear Proteins , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adenoviridae/genetics , Adenoviridae/metabolism , Adenovirus E3 Proteins/genetics , Amino Acid Sequence , Carrier Proteins/metabolism , Cell Cycle , Cell Line , Dyneins/chemistry , GTP Phosphohydrolases/chemistry , Humans , Microtubule Proteins/genetics , Microtubule Proteins/metabolism , Molecular Sequence Data , Monomeric GTP-Binding Proteins/metabolism , Plasmids/genetics , RNA, Messenger/metabolism , Two-Hybrid System Techniques , t-Complex Genome Region
14.
J Biol Chem ; 275(13): 9882-9, 2000 Mar 31.
Article in English | MEDLINE | ID: mdl-10734145

ABSTRACT

FIP3, isolated as a type 2 adenovirus E3-14.7-kDa interacting protein, is an essential component of the multimeric IkappaB-alpha kinase (IKK) complex and has been shown to interact with various components (Fas receptor-interacting protein, NF-kappaB-inducing kinase, IKKbeta) of the NF-kappaB activation pathway. FIP3 has also been shown to repress basal and tumor necrosis factor (TNF) alpha-induced NF-kappaB activity as well as to induce cell death when overexpressed. The adenovirus E3-14.7-kDa protein (E3-14.7K) is an inhibitor of TNFalpha-induced cell death. In the current study, we generated deletion mutants to map the domains of FIP3, which are responsible for its various functions. The NF-kappaB inhibitory activity and the E3-14.7K binding domains were mapped at the carboxyl half of the FIP3 protein. We also found that the carboxyl-terminal half of FIP3 blocked TNFalpha-induced IkappaB-alpha phosphorylation and subsequent degradation, which suggests that the stabilization of the cytoplasmic inhibitor of NF-kappaB underlies the FIP3 inhibition of NF-kappaB activity. The amino-terminal 119 amino acids were responsible for the FIP3-IKKbeta and FIP3-IKKalpha interaction, and the middle of the protein (amino acids 201-300) appeared to be both the FIP3 self-association domain as well as the FIP3-Fas receptor-interacting protein interaction domain. Thus, FIP3 might serve as a scaffold protein to organize the various components of the IkappaB-alpha kinase complex. Whereas the full-length protein is required for efficient cell death, the amino-terminal 200 amino acids are sufficient to cause rounding and detachment of the cells from the monolayer.


Subject(s)
NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Viral Proteins/metabolism , Amino Acids/chemistry , Down-Regulation , I-kappa B Kinase , Phosphorylation , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Viral Proteins/chemistry
15.
Immunol Rev ; 169: 241-53, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10450521

ABSTRACT

Conceptually, the initiation of autoimmune disease can be described as a three-stage process involving both genetic and environmental influences. This process begins with the development of an autoimmune cellular repertoire, followed by activation of these autoreactive cells in response to a localized target and, finally, the immune system's failure to regulate these self-reactive constituents. Viruses have long been associated with inciting autoimmune disorders. Two mechanisms have been proposed to explain how a viral infection can overcome immunological tolerance to self-components and initiate an organ-specific autoreactive process; these mechanisms are molecular mimicry and bystander activation. Both pathways, as discussed here, could play pivotal roles in the development of autoimmunity without necessarily excluding each other. Transgene technology has allowed us and others to examine more closely the roles of these mechanisms in mice and to dissect the requirements for initiating disease. These results demonstrate that bystander activation is the most likely explanation for disease development. Additional evidence suggests a further role for viruses in the reactivation and chronicity of autoimmune diseases. In this scenario, a second invasion by a previously infecting virus may restimulate already existing autoreactive lymphocytes, and thereby contribute to the diversity of the immune response.


Subject(s)
Autoimmunity , Viruses/immunology , Viruses/pathogenicity , Animals , Disease Models, Animal , Humans , Inflammation/immunology , Lymphocyte Activation , Mice , Mice, Transgenic , Molecular Mimicry , Organ Specificity , Virus Diseases/immunology
16.
J Neurovirol ; 5(1): 55-64, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10190691

ABSTRACT

Mouse adenovirus-type 1 (MAV-1) has recently been shown to cause a fatal hemorrhagic encephalopathy in certain strains of mice whereas other strains are resistant. Morbidity is associated with a productive infection of cerebrovascular endothelial cells, resulting in necrosis of the vasculature, infarction, hemorrhage and death within 4 - 6 days. Previous studies were not able to define a role for the innate or acquired immune response. In the current study we have addressed the effect of MAV-1 on chemokine and chemokine receptor expression in the central nervous system (CNS) and spleen of susceptible (C57BL/6) and resistant (BALB/c) strains of mice. Intra-peritoneal infection with MAV-1 in C57BL/6 animals resulted in early and prominent induction of IP-10/crg-2 in the spleen and CNS. Increased expression of MCP-1, MIP-1alpha, MIP-1beta and RANTES was also noted in the CNS of MAV-1-infected C57BL/6 animals commencing around 72 h post-infection. In contrast, chemokine expression in BALB/c animals was more restricted with prominent upregulation only of MIP-2 in the CNS. In situ hybridization identified the vascular endothelium and CNS glia as the principal site of IP-10/crg-2 production in the C57BL/6 animals. The chemokine receptors CCR1-5 were upregulated in the CNS of both strains of mice. These data show that productive infection of the CNS with MAV-1 leads to the upregulation of a characteristic pattern of chemokines and their receptors, which may point to a role for these factors in disease pathogenesis.


Subject(s)
Adenoviridae Infections/metabolism , Brain Diseases/metabolism , Chemokines/metabolism , Mastadenovirus , Animals , Brain Diseases/immunology , Brain Diseases/virology , Chemokine CXCL10 , Chemokines/genetics , Chemokines, CXC/metabolism , Disease Susceptibility , Female , L Cells , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA, Messenger/analysis , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Spleen/metabolism , Time Factors
17.
Lab Invest ; 79(2): 235-42, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10068211

ABSTRACT

To recognize and respond immunologically to foreign antigens, T lymphocytes require the presentation of foreign peptides by MHC molecules. To determine which cells of the central nervous system (CNS) are capable of expressing MHC molecules, we used confocal microscopy and dual immunofluorescence with cell-specific and MHC-specific antibodies to study brain sections of adult mice. We took advantage of transgenic mice that initiate CNS-specific expression of IFN-gamma at 8 weeks of age. This inflammatory cytokine is a strong inducer of MHC expression both in culture and in vivo. From this analysis, we clearly found MHC class I and II expression on endothelial, microglial, and oligodendrocyte cell types, but did not find astrocytes or neurons capable of expressing either MHC class I or II molecules under these conditions. This finding suggests that, although microglia and oligodendrocytes may participate in the antigen presentation process in the organism, we found no in vivo evidence to support the concept that astrocytes act as antigen-presenting cells.


Subject(s)
Central Nervous System/drug effects , Interferon-gamma/pharmacology , Major Histocompatibility Complex/physiology , Animals , Astrocytes/immunology , Central Nervous System/cytology , Central Nervous System/immunology , Endothelium/cytology , Endothelium/immunology , Fluorescent Antibody Technique , Histocompatibility Antigens Class I/analysis , Histocompatibility Antigens Class II/analysis , Interferon-gamma/genetics , Mice , Mice, Transgenic/genetics , Microglia/immunology , Microscopy, Confocal , Neurons/immunology , Oligodendroglia/immunology
18.
J Virol ; 73(3): 1756-66, 1999 Mar.
Article in English | MEDLINE | ID: mdl-9971752

ABSTRACT

Coxsackievirus infection causes severe pancreatitis and myocarditis in humans, often leading to death in young or immunocompromised individuals. In susceptible strains of mice, coxsackievirus strain CB4 causes lethal hypoglycemia. To investigate the potential of gamma interferon (IFN-gamma) in protection and clearance of the viral infection, IFN-gamma knockout mice and transgenic (Tg) mice specifically expressing IFN-gamma in their pancreatic beta cells were infected with CB4. Lack of IFN-gamma in mice normally resistant to CB4-mediated disease resulted in hypoglycemia and rapid death. However, expression of IFN-gamma in the beta cells of Tg mice otherwise susceptible to lethal infection allowed for survival and protected them from developing the accompanying hypoglycemia. While all the mice had high levels of viral replication in their pancreata and comparable tissue pathology following viral infection, the Tg mice had significantly lower levels of virus at the peak of infection, significantly higher numbers of activated macrophages before and after infection, and less damage to their acinar tissue. Additionally, despite having increased levels of inducible nitric oxide synthetase (iNOS) expression, treatment of Tg mice with the iNOS inhibitor aminoguanidine did not alter the level of protection afforded by IFN-gamma expression. In conclusion, IFN-gamma protects from lethal coxsackievirus infection by activating macrophages in an iNOS-independent manner.


Subject(s)
Coxsackievirus Infections/immunology , Interferon-gamma/physiology , Pancreatitis/prevention & control , Animals , Lymphocytes/physiology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Nitric Oxide/physiology
19.
Proc Natl Acad Sci U S A ; 96(3): 1042-7, 1999 Feb 02.
Article in English | MEDLINE | ID: mdl-9927690

ABSTRACT

FIP-3 (14.7K interacting protein) was discovered during a search for cell proteins that could interact with an adenovirus protein (Ad E3-14.7K) that had been shown to prevent tumor necrosis factor (TNF)-alpha-induced cytolysis. FIP-3, which contains leucine zippers and a zinc finger domain, inhibits both basal and induced transcriptional activity of NF-kappaB and causes a late-appearing apoptosis with unique morphologic manifestations. Ad E3-14.7K can partially reverse apoptotic death induced by FIP-3. FIP-3 also was shown to bind to other cell proteins, RIP and NIK, which previously had been described as essential components of TNF-alpha-induced NF-kappaB activation. In addition, FIP-3 inhibited activation of NF-kappaB induced by TNF-alpha, the TNFR-1 receptor, RIP, NIK, and IKKbeta, as well as basal levels of endogenous NF-kappaB in 293 cells. Because the activation of NF-kappaB has been shown to inhibit apoptosis, FIP-3 appears both to activate a cell-death pathway and to inhibit an NF-kappaB-dependent survival mechanism.


Subject(s)
Carrier Proteins/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/physiology , Adenoviridae/genetics , Adenovirus E3 Proteins/genetics , Adenovirus E3 Proteins/metabolism , Amino Acid Sequence , Apoptosis , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Cell Survival/drug effects , Glutathione Transferase , Humans , I-kappa B Kinase , Leucine Zippers , Molecular Sequence Data , Molecular Weight , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases , Recombinant Fusion Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Zinc Fingers , NF-kappaB-Inducing Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...