Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 98(21): 12162-7, 2001 Oct 09.
Article in English | MEDLINE | ID: mdl-11593031

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) is well recognized for its role in mediating innate immune responses. However, the mechanisms of TNF-alpha that influence the adaptive immune response to virus infections are not well understood. In this study, we have investigated the role of TNF-alpha in activating the cellular and humoral responses to systemic viral challenge with recombinant replication-defective adenovirus (rAd). Evaluation of T cell function in TNF-alpha-deficient (TNFKO) mice revealed impaired virus-specific proliferation of T cells derived from the draining lymph nodes of the liver. Analysis of dendritic cells (DC) isolated from local draining lymph nodes after systemic challenge showed that DC from TNFKO mice were relatively immature compared with those from strain-matched wild-type mice. In vitro, TNF-alpha was required to mature DC efficiently during virus-mediated stimulation. Adoptive transfer of primed, mature DC into TNFKO mice restored T cell responses and reconstituted anti-adenovirus antibody responses. Thus, TNF-alpha plays a significant role in the maturation of DC after adenovirus challenge both in vitro and in vivo, highlighting the importance of this innate cytokine in activating adaptive immunity to viral challenge.


Subject(s)
Adenoviridae Infections/immunology , Dendritic Cells/immunology , Tumor Necrosis Factor-alpha/immunology , Adaptation, Physiological/immunology , Adenoviruses, Human/immunology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , Bone Marrow Cells/immunology , Cell Differentiation , Defective Viruses/immunology , Dendritic Cells/cytology , Immunity, Active , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/genetics
2.
Surgery ; 129(1): 48-54, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11150033

ABSTRACT

BACKGROUND: Tumor necrosis factor-alpha (TNF) is thought to act as a stimulator for initiating hepatocyte proliferation after partial hepatectomy (PH). At the same time, TNF induces a series of inflammatory responses that may be detrimental for the liver and other remote organs. The purpose of this study was to investigate the effect of TNF on the pathophysiologic state after PH. METHODS: Wild-type (TNF+/+) and TNF-deficient (TNF-/-) mice underwent 70% PH. Hepatocyte proliferation was assessed by bromodeoxyuridine labeling and mitotic index. Liver function was evaluated by alanine aminotransferase (ALT) and total bilirubin levels in serum after PH. Myeloperoxidase activity in the liver and lung was measured as a marker for neutrophil activation. RESULTS: No differences were observed in liver regeneration or hepatocyte proliferation between TNF+/+ and TNF-/- mice. The survival of TNF-/- mice on day 1 after PH was significantly higher than that of TNF+/+ mice, but both groups had similar survival thereafter. The ALT level was significantly higher in TNF+/+ mice 6 hours after PH and myeloperoxidase activities in both liver and lung were markedly elevated in TNF+/+ mice compared with TNF-/- mice. CONCLUSIONS: These findings demonstrate that TNF gene-depleted mice do not demonstrate delayed liver regeneration but do suppress neutrophil activation after PH compared with results in wild-type (TNF +/+) mice.


Subject(s)
Liver Regeneration/physiology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , Animals , Cell Division , Female , Gene Deletion , Hepatectomy , Hepatocytes/cytology , Interleukin-6/blood , Liver/enzymology , Liver Regeneration/genetics , Lung/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxidase/metabolism , Tumor Necrosis Factor-alpha/physiology
3.
Infect Immun ; 69(1): 129-36, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11119498

ABSTRACT

The contribution of granulocyte-macrophage colony-stimulating factor (GM-CSF), a hematopoietic and immunoregulatory cytokine, to resistance to blood-stage malaria was investigated by infecting GM-CSF-deficient (knockout [KO]) mice with Plasmodium chabaudi AS. KO mice were more susceptible to infection than wild-type (WT) mice, as evidenced by higher peak parasitemia, recurrent recrudescent parasitemia, and high mortality. P. chabaudi AS-infected KO mice had impaired splenomegaly and lower leukocytosis but equivalent levels of anemia compared to infected WT mice. Both bone marrow and splenic erythropoiesis were normal in infected KO mice. However, granulocyte-macrophage colony formation was significantly decreased in these tissues of uninfected and infected KO mice, and the numbers of macrophages in the spleen and peritoneal cavity were significantly lower than in infected WT mice. Serum levels of gamma interferon (IFN-gamma) were found to be significantly higher in uninfected KO mice, and the level of this cytokine was not increased during infection. In contrast, IFN-gamma levels were significantly above normal levels in infected WT mice. During infection, tumor necrosis factor alpha (TNF-alpha) levels were significantly increased in KO mice and were significantly higher than TNF-alpha levels in infected WT mice. Our results indicate that GM-CSF contributes to resistance to P. chabaudi AS infection and that it is involved in the development of splenomegaly, leukocytosis, and granulocyte-macrophage hematopoiesis. GM-CSF may also regulate IFN-gamma and TNF-alpha production and activity in response to infection. The abnormal responses seen in infected KO mice may be due to the lack of GM-CSF during development, to the lack of GM-CSF in the infected mature mice, or to both.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Malaria/immunology , Plasmodium chabaudi , Anemia/etiology , Animals , Cytokines/biosynthesis , Erythropoiesis , Hematopoiesis , Leukocytosis/etiology , Malaria/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Splenomegaly/etiology
4.
Neuromuscul Disord ; 10(8): 612-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11053690

ABSTRACT

We have previously demonstrated a role for T cells in Duchenne muscular dystrophy (DMD) using the mdx mouse and have shown that T cell killing of dystrophic muscle can occur through perforin-dependent and perforin-independent mechanisms. In this investigation, we explore the possibility that one perforin-independent mechanism utilized by the T cells is cytokine-based killing, specifically by tumor necrosis factor (TNF). We tested this hypothesis by generating mice that are TNF-deficient and dystrophin-deficient (TNF-/mdx). Body mass and muscle mass of the TNF-/mdx mice were significantly less than TNF+/mdx mice at 8 weeks of age. Creatine kinase levels and overall muscle strength were unchanged. Histopathology measurements showed different results in the diaphragm and quadriceps muscles. These data suggest that removal of TNF in vivo in dystrophic mice has differential effects on diaphragm and quadriceps suggesting that TNF is an unfavorable target for immunotherapy for DMD.


Subject(s)
Diaphragm/metabolism , Diaphragm/pathology , Dystrophin/deficiency , Leg/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/immunology , Tumor Necrosis Factor-alpha/deficiency , Age Factors , Animals , Body Weight/physiology , Creatine Kinase/metabolism , Diaphragm/physiopathology , Disease Models, Animal , Dystrophin/genetics , Female , Immunotherapy/methods , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Leg/physiopathology , Male , Mice , Mice, Knockout/anatomy & histology , Mice, Knockout/metabolism , Muscle Weakness/diagnosis , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/genetics
5.
Infect Immun ; 68(11): 6289-93, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11035737

ABSTRACT

Tumor necrosis factor (TNF)-deficient mice were challenged with Leishmania donovani to characterize TNF in the response of visceral intracellular infection to antileishmanial chemotherapy. In wild-type controls (i) liver infection peaked at week 2 and resolved, (ii) discrete liver granulomas developed at weeks 2 to 4 and involuted, and (iii) leishmanicidal responses to antimony (Sb), amphotericin B (AmB), and miltefosine were intact. In TNF knockout (KO) mice (i) initial liver infection was unrestrained, plateaued, and then declined somewhat by week 6, (ii) an absent early granulomatous reaction abruptly accelerated with striking tissue inflammation, widespread hepatic necrosis, and 100% mortality by week 10, and (iii) while the initial response to AmB and miltefosine was intact, killing induced by Sb therapy was reduced by >50%. Although initial AmB treatment during weeks 2 to 3 killed 98% of liver parasites, 75% of AmB-treated KO mice subsequently relapsed and died by week 12; however, additional maintenance AmB preserved long-term survival. These results for a model of visceral infection indicate that endogenous TNF is required early on to control intracellular L. donovani, support granuloma development, and mediate optimal initial effects of Sb and prevent relapse after ordinarily curative AmB treatment. A compensatory, TNF-independent antileishmanial mechanism developed in TNF KO mice; however, its effect was uncontrolled fatal inflammation. Chemotherapeutic elimination of the parasite stimulus reversed the hyperinflammatory response and preserved survival.


Subject(s)
Leishmania donovani , Leishmaniasis, Visceral/immunology , Tumor Necrosis Factor-alpha/physiology , Amphotericin B/therapeutic use , Animals , Female , Leishmaniasis, Visceral/drug therapy , Leishmaniasis, Visceral/mortality , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Th1 Cells/physiology
6.
Eur J Immunol ; 30(9): 2586-92, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11009092

ABSTRACT

T cell production by the thymus, thymic size, cellularity and output all decrease drastically after puberty. Among the candidates that may mediate this decrease are the sex steroids: hypersecretion or pharmacological administration of these hormones has long been known to induce thymic hypocellularity, and their depletion yields thymic hypercellularity. Here we show that a typical sex steroid, testosterone, specifically targets CD8+CD4+ double-positive (DP) thymocytes for apoptosis via TNF-alpha. Anti-TNF-alpha monoclonal antibodies abrogated testosterone-induced DP apoptosis, and TNF-alpha-/- DP thymocytes were largely resistant to testosterone-mediated apoptosis in vivo. Testosterone accomplished this effect by upregulating TNF-alpha production and by simultaneously sensitizing DP thymocytes to TNF-alpha. Thus, TNF-alpha is the critical mediator of sex steroid-induced apoptosis in thymocytes, and its manipulation should provide a point of intervention to modulate T cell production in sex hormone disorders.


Subject(s)
Apoptosis/drug effects , CD4 Antigens/analysis , CD8 Antigens/analysis , T-Lymphocyte Subsets/drug effects , Testosterone/pharmacology , Tumor Necrosis Factor-alpha/physiology , Animals , Mice , Mice, Inbred C57BL , T-Lymphocyte Subsets/physiology
7.
J Leukoc Biol ; 68(1): 151-7, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10914503

ABSTRACT

Mice with combined lymphotoxin-alpha (LTalpha) and tumor necrosis factor (TNF) deficiencies show defects in the structure of peripheral lymphoid organs such as spleen, lymph nodes, and gut-associated lymphoid tissues. To identify genes associated with this defective phenotype in spleen, we applied a gene profiling approach, including subtractive cloning and gene array hybridizations, to mice with combined TNF/LT deficiency. The differentially expressed genes identified by these techniques was then evaluated by Northern blot analysis for splenic expression in knockout mice with single LTalpha or single TNF deficiency. Most of the genes detected in this analysis are directly or indirectly associated with disrupted LT and not TNF signaling.


Subject(s)
Gene Expression Profiling , Lymphoid Tissue/pathology , Lymphotoxin-alpha/genetics , Spleen/metabolism , Tumor Necrosis Factor-alpha/deficiency , Animals , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Chemokines/biosynthesis , Chemokines/genetics , DNA, Complementary/genetics , Expressed Sequence Tags , Gene Expression Profiling/methods , Gene Expression Regulation , Group II Phospholipases A2 , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Organ Specificity , Pancreas/enzymology , Phenotype , Phospholipases A/biosynthesis , Phospholipases A/genetics , Phospholipases A/isolation & purification , Receptors, Chemokine/biosynthesis , Receptors, Chemokine/genetics , Specific Pathogen-Free Organisms , Spleen/pathology , Subtraction Technique , Tumor Necrosis Factor-alpha/genetics
8.
Lab Invest ; 80(6): 901-14, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10879741

ABSTRACT

Tumour necrosis factor-alpha (TNF) plays a central role in the recruitment and activation of mononuclear cells in mycobacterial infection. In the absence of type 1 TNF receptor, Mycobacterium bovis Bacillus Calmette-Guerin (BCG) infection of mice is not contained, leading to fatal disease. Because type 1 TNF receptor binds both TNF and lymphotoxin-a, we used TNF-deficient mice to determine the specific role of TNF in the host resistance to BCG infection. The bacterial burden of the lungs of TNF-deficient mice was substantially increased and the mice succumbed to pneumonia between 8 and 12 weeks with a defective granuloma response. Atypical granulomas developed by 4 weeks expressing low levels of MHC class II, intracellular adhesion molecule (ICAM-1), CD11b and CD11c. Macrophages showed little signs of activation and had low levels of acid phosphatase activity and inducible nitric oxide synthase (INOS) expression. Despite the defective cellular recruitment, the chemokines, monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-1 (MIP-1alpha), were increased in broncho-alveolar lavage fluid of TNF-deficient mice. The defective host response was corrected by the transplantation of normal bone marrow cells into irradiated TNF-deficient mice. These results demonstrate that TNF derived from hemopoietic cells rather than from mesenchymal origin are essential for a normal host response to BCG infection. Furthermore, TNF dependent expression of adhesion molecules may be essential for the recruitment of mononuclear cells for the formation of bactericidal BCG granulomas.


Subject(s)
Antigens, CD/physiology , Bone Marrow Transplantation , Mycobacterium bovis/immunology , Receptors, Tumor Necrosis Factor/physiology , Tuberculosis/immunology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/immunology , Acid Phosphatase/analysis , Animals , Antigens, CD/genetics , Granuloma/microbiology , Histocompatibility Antigens Class II/analysis , Immunity, Innate , Integrin alphaXbeta2/analysis , Intercellular Adhesion Molecule-1/analysis , Lung/immunology , Lung/microbiology , Lung/pathology , Macrophage-1 Antigen/analysis , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium bovis/isolation & purification , Pneumonia/microbiology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I , Tuberculosis/complications , Tuberculosis/genetics , Tumor Necrosis Factor-alpha/genetics
9.
J Immunol ; 164(4): 1689-94, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657611

ABSTRACT

Apoptosis is one of the key regulatory mechanisms in tissue modeling and development. In the thymus, 95-98% of all thymocytes die by apoptosis because they failed to express a TCR with an optimal affinity for the selecting intrathymic peptide-MHC complexes. We studied the possible role of two prominent nerve growth factor (NGF-TNF) family member systems, Fas ligand (FasL)-Fas receptor (FasR) and TNF-alpha-TNFR, in apoptosis of murine CD8+4+ double-positive (DP) thymocytes induced via TCR-CD3- and cAMP-mediated signaling. TCR-CD3epsilon-mediated apoptosis of DP thymocytes was found not to be dependent on either of the two systems. The FasL-FasR system was also found to be dispensable for the cAMP-mediated apoptosis. By contrast, cAMP agonists (dibutyryl-cAMP and forskolin) induced apoptosis via TNF-alpha, as evidenced by 1) the ability of anti-TNF-alpha mAbs to abrogate cAMP analogue-induced DP apoptosis in a dose-dependent manner; and 2) increased resistance of DP thymocytes from TNF-alpha-/- and TNFR I-/-II-/- animals to cAMP agonist-mediated apoptosis. cAMP agonists induced DP thymocyte death by a combination of two mechanisms: first, they induced selective up-regulation of TNF-alpha production, and, second, they sensitized DP thymocytes to TNF-alpha. The latter effect may be due to the down-regulation of TNFR-associated factor 2 protein. These results identify TNF-alpha as the critical mediator of cAMP-induced apoptosis in thymocytes and provide a molecular explanation for how the cAMP stimulators, including the sex steroids, may modulate T cell production output, as observed under physiological and pharmacological conditions.


Subject(s)
Apoptosis/immunology , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cyclic AMP/physiology , T-Lymphocyte Subsets/immunology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis/genetics , Bucladesine/pharmacology , Colforsin/pharmacology , Cyclic AMP/agonists , Dose-Response Relationship, Immunologic , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Knockout , Receptor-CD3 Complex, Antigen, T-Cell/physiology , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism
10.
Immunity ; 11(3): 379-89, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10514016

ABSTRACT

Tumor necrosis factor (TNF) receptor-associated factor 2 (TRAF2) can interact with various members of the TNF receptor family. Previously, we reported that TRAF2-deficient mice die prematurely and have elevated serum TNF levels. In this study, we demonstrate that TRAF2-deficient macrophages produce increased amounts of nitric oxide (NO) and TNF in response to TNF stimulation. Furthermore, we could enhance the survival of TRAF2-deficient mice by eliminating either TNF or TNFR1. Using these double-knockout mice, we show that in the absence of TRAF2, the T helper-dependent antibody response, CD40-mediated proliferation, and NF-kappaB activation are defective. These data demonstrate two important roles of TRAF2, one as a negative regulator of certain TNFR1 signals and the other as a positive mediator of CD40 signaling.


Subject(s)
Antigens, CD/metabolism , CD40 Antigens/metabolism , Proteins/physiology , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Animals , Cell Division , Cells, Cultured , Female , Immunoglobulin Class Switching , Immunoglobulin Isotypes , Interleukin-12/biosynthesis , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Phenotype , Proteins/genetics , Receptors, Tumor Necrosis Factor, Type I , Spleen/cytology , TNF Receptor-Associated Factor 2 , Tumor Necrosis Factor-alpha/biosynthesis , Vesicular stomatitis Indiana virus
11.
Cancer Res ; 59(18): 4516-8, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10493498

ABSTRACT

To examine the hypothesis that tumor necrosis factor (TNF) alpha is an essential cytokine in carcinogenesis, we conducted two-stage carcinogenesis experiments with an initiator, 7,12-dimethylbenz(a)anthracene (DMBA), plus either of two tumor promoters, okadaic acid and 12-O-tetradecanoylphorbol-13-acetate (TPA), on the skin of TNF-alpha-deficient (TNF-/-) mice. TNF-/- mice treated with DMBA plus okadaic acid developed no tumors for up to 19 weeks, and at 20 weeks, the percentage of tumor-bearing TNF-/- mice was 10%, whereas the percentage of tumor-bearing TNF+/+ mice was 100%. In TNF-/- mice treated with DMBA plus TPA, tumor onset was delayed 4 weeks, and the time to development of small tumors in 100% of mice was 9 weeks later than that seen in TNF+/+ CD-1 mice. The average number of tumors in TPA-treated TNF-/- mice was 2.8, compared with 11.8 for TNF+/+ CD-1 mice. To understand the residual tumor-promoting activity in TNF-/- mice, we also investigated the possible significance of interleukin (IL) 1 as an additional cytokine in tumor promotion. A single application of TPA and okadaic acid increased IL-1alpha and IL-1beta gene expression in TNF-/- mice. All of our results demonstrate that TNF-alpha is the key cytokine for tumor promotion in mouse skin and, very possibly, for carcinogenesis in humans as well.


Subject(s)
Carcinogens/toxicity , Genes, ras , Skin Neoplasms/pathology , Tumor Necrosis Factor-alpha/physiology , 3T3 Cells , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Cell Division/drug effects , Cell Line, Transformed , Female , Interleukin-1/genetics , Interleukin-1/pharmacology , Interleukin-1/physiology , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Okadaic Acid/toxicity , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Tetradecanoylphorbol Acetate/toxicity , Transfection , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
12.
Proc Natl Acad Sci U S A ; 96(15): 8721-6, 1999 Jul 20.
Article in English | MEDLINE | ID: mdl-10411942

ABSTRACT

The present study evaluated behavioral and histopathological outcome after controlled cortical impact (CCI) brain injury in mice deficient in tumor necrosis factor [TNF(-/-)] and their wild-type (wt) littermates. Mice were subjected to CCI brain injury [TNF(-/-), n = 10; wt, n = 10] or served as uninjured controls [TNF(-/-), n = 10; wt, n = 10] and were evaluated for deficits in memory retention at 7 days postinjury. Although both brain-injured wt and TNF(-/-) mice exhibited significant memory dysfunction compared to uninjured controls (P < 0.02), the deficits in memory retention in injured TNF(-/-) mice were significantly less severe than in injured wt mice (P < 0.02). A second group of mice was subjected to CCI brain injury [TNF(-/-), n = 20; wt, n = 20] or served as uninjured controls [TNF(-/-), n = 15; wt, n = 15] and were evaluated over a 4-week period for neurological motor function. In the acute posttraumatic period (48 h postinjury), brain-injured TNF(-/-) mice were significantly less impaired than injured wt mice on composite neuroscore (P < 0.001), rotarod (P < 0.05), and beam balance (P < 0. 02) tests. However, wt mice recovered from brain injury by 2-3 weeks postinjury, whereas TNF(-/-) mice continued to demonstrate persistent motor deficits up to 4 weeks postinjury. Histopathological analysis at 2 and 4 weeks postinjury revealed that brain-injured TNF(-/-) mice had significantly more cortical tissue loss than wt mice (P < 0.02). Our results suggest that although the presence of TNF in the acute posttraumatic period may be deleterious, this cytokine may play a role in facilitating long-term behavioral recovery and histological repair after brain injury.


Subject(s)
Brain Injuries/physiopathology , Tumor Necrosis Factor-alpha/deficiency , Animals , Behavior, Animal , Brain/pathology , Brain Injuries/genetics , Maze Learning , Memory , Mice , Mice, Knockout , Motor Activity , Postural Balance , Time Factors , Tumor Necrosis Factor-alpha/genetics
13.
J Virol ; 73(6): 5098-109, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10233973

ABSTRACT

The cellular and humoral immune responses to adenovirus (Ad) remain a major barrier to Ad-mediated gene therapy. We recently reported that mice deficient in tumor necrosis factor alpha (TNF-alpha) or Fas (APO-1, CD95) have prolonged expression of an Ad transgene expressing a foreign protein in the liver. To determine whether blockade of TNF-alpha or Fas would have the same effect in normal mice, we created transgenes that expressed soluble murine CD8 or CD8 fused to the extracellular regions of TNF receptor 1 (TNFR) or Fas and inserted into the left-end region of first-generation (E1/E3-) Ad vectors. Consistent with the results observed in TNF-deficient mice, expression of the TNFR-CD8 fusion protein was prolonged in vivo compared to that of control proteins. Not only did expression of TNFR-CD8 persist in the liver and the lung, but when coadministered with another first-generation vector, the protein provided "transprotection" for the companion vector and transgene. In addition, TNFR-CD8 attenuated the humoral immune response to the Ad. Together, these findings demonstrate that blockade of TNF-alpha is likely to be useful in extending the expression of an Ad-encoded transgene in a gene therapy application.


Subject(s)
Adenoviridae/genetics , Liver/metabolism , Lung/metabolism , Recombinant Fusion Proteins/pharmacology , Transgenes , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Antibodies, Viral/biosynthesis , Antigens, CD/physiology , CD8 Antigens/physiology , Enzyme-Linked Immunosorbent Assay , Genetic Therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, SCID , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type I , fas Receptor/physiology
14.
Proc Natl Acad Sci U S A ; 96(6): 2994-9, 1999 Mar 16.
Article in English | MEDLINE | ID: mdl-10077625

ABSTRACT

Mice lacking the RelA (p65) subunit of NF-kappaB die between days 14 and 15 of embryogenesis because of massive liver destruction. Fibroblasts and macrophages isolated from relA-/- embryos were found to be highly sensitive to tumor necrosis factor (TNF) cytotoxicity, raising the possibility that endogenous TNF is the cause of liver cell apoptosis. To test this idea, we generated mice lacking both TNF and RelA. Embryogenesis proceeds normally in such mice, and TNF/RelA double-deficient mice are viable and have normal livers. Thus, the RelA-mediated antiapoptotic signal that protects normal cells from TNF injury in vitro can be shown to be operative in vivo.


Subject(s)
Embryonic and Fetal Development/genetics , NF-kappa B/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Apoptosis/genetics , Embryonic and Fetal Development/immunology , Fetal Death/genetics , Fetal Death/immunology , Gene Expression Regulation, Developmental/immunology , Mice , Mice, Knockout , NF-kappa B/immunology , Transcription Factor RelA , Tumor Necrosis Factor-alpha/immunology
15.
Eur J Immunol ; 28(12): 3980-8, 1998 12.
Article in English | MEDLINE | ID: mdl-9862334

ABSTRACT

Granulocyte-macrophage colony-stimulating factor-deficient (GM-CSF-/-) mice produce far lower serum levels of IFN-gamma in response to LPS than GM-CSF+/+ mice. CD4+ and CD8+ T cells from LPS-injected GM-CSF-/- mice showed a deficiency in IFN-gamma production and proliferative activity in response to IL-2 and IL-12, whereas IFN-gamma production by NK cells was not compromised. These defects of T cells were reversed by administration of GM-CSF in vivo, but not by supplementation with GM-CSF in vitro. GM-CSF-/- mice do not have an intrinsic defect in IFN-gamma production, because IL-12 injection induces the same high levels of IFN-gamma in GM-CSF-/- and GM-CSF+/+ mice. To investigate the inhibitory effect of LPS on GM-CSF-/- T cells and the indirect restorative activity of GM-CSF, we tested the action of supernatants from cultured dendritic cells (DC). A factor or factors in the DC supernatant normalized serum IFN-gamma levels and T cell responses in LPS-injected GM-CSF-/- mice. IL-18 reproduced some but not all of these in vivo and in vitro effects of DC supernatants. Our results indicate that GM-CSF is important in protecting T cells from inhibitory signals generated during immunization or exposure to LPS, and that this effect of GM-CSF is indirect and mediated by factors produced by DC.


Subject(s)
Antigen Presentation/immunology , Gene Expression Regulation/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Interferon-gamma/immunology , Animals , Antigen Presentation/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interferon-gamma/biosynthesis , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Mice , Mice, Knockout , T-Lymphocytes/immunology
16.
Proc Natl Acad Sci U S A ; 95(23): 13806-11, 1998 Nov 10.
Article in English | MEDLINE | ID: mdl-9811882

ABSTRACT

Inflammation is associated with production of cytokines and chemokines that recruit and activate inflammatory cells. Interleukin (IL) 12 produced by macrophages in response to various stimuli is a potent inducer of interferon (IFN) gamma production. IFN-gamma, in turn, markedly enhances IL-12 production. Although the immune response is typically self-limiting, the mechanisms involved are unclear. We demonstrate that IFN-gamma inhibits production of chemokines (macrophage inflammatory proteins MIP-1alpha and MIP-1beta). Furthermore, pre-exposure to tumor necrosis factor (TNF) inhibited IFN-gamma priming for production of high levels of IL-12 by macrophages in vitro. Inhibition of IL-12 by TNF can be mediated by both IL-10-dependent and IL-10-independent mechanisms. To determine whether TNF inhibition of IFN-gamma-induced IL-12 production contributed to the resolution of an inflammatory response in vivo, the response of TNF+/+ and TNF-/- mice injected with Corynebacterium parvum were compared. TNF-/- mice developed a delayed, but vigorous, inflammatory response leading to death, whereas TNF+/+ mice exhibited a prompt response that resolved. Serum IL-12 levels were elevated 3-fold in C. parvum-treated TNF-/- mice compared with TNF+/+ mice. Treatment with a neutralizing anti-IL-12 antibody led to resolution of the response to C. parvum in TNF-/- mice. We conclude that the role of TNF in limiting the extent and duration of inflammatory responses in vivo involves its capacity to regulate macrophage IL-12 production. IFN-gamma inhibition of chemokine production and inhibition of IFN-gamma-induced IL-12 production by TNF provide potential mechanisms by which these cytokines can exert anti-inflammatory/repair function(s).


Subject(s)
Anti-Inflammatory Agents/pharmacology , Interferon-gamma/pharmacology , Interleukin-12/biosynthesis , Macrophages, Peritoneal/immunology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Drug Antagonism , Female , Inflammation/prevention & control , Interleukin-12/antagonists & inhibitors , Macrophage Inflammatory Proteins/biosynthesis , Mice , Mice, Inbred C3H , Mice, Inbred C57BL
17.
Nat Med ; 4(1): 78-83, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9427610

ABSTRACT

Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS) characterized by localized areas of demyelination. Although the etiology and pathogenesis of MS remain largely unknown, it is generally assumed that immune responses to myelin antigens contribute to the disease process. The exact sequence of events, as well as the molecular mediators that lead to myelin destruction, is yet to be defined. As a potent mediator of inflammation, the cytopathic cytokine, tumor necrosis factor (TNF) has been considered to be a strong candidate in the pathogenesis of MS and its animal model, experimental autoimmune encephalomyelitis (EAE). However, its role in immune-mediated demyelination remains to be elucidated. To determine the contribution of TNF to the pathogenesis of the MS-like disease provoked by the myelin oligodendrocyte glycoprotein (MOG), we have tested mice with an homologous disruption of the gene encoding TNF. Here we report that upon immunization with MOG, mice lacking TNF develop severe neurological impairment with high mortality and extensive inflammation and demyelination. We show further that inactivation of the TNF gene converts MOG-resistant mice to a state of high susceptibility. Furthermore, treatment with TNF dramatically reduces disease severity in both TNF-/- mice and in other TNF+/+ mice highly susceptible to the MOG-induced disease. These findings indicate that TNF is not essential for the induction and expression of inflammatory and demyelinating lesions, and that it may limit the extent and duration of severe CNS pathology.


Subject(s)
Multiple Sclerosis/physiopathology , Spinal Cord/pathology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/physiology , Animals , Crosses, Genetic , Female , Heterozygote , Inflammation , Lymphocytes/immunology , Lymphocytes/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Inbred Strains , Mice, Knockout , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Myelin Proteins , Myelin Sheath/pathology , Myelin-Associated Glycoprotein , Myelin-Oligodendrocyte Glycoprotein , Oligodendroglia/physiology , Recombinant Proteins/therapeutic use , Spinal Cord/immunology , Tumor Necrosis Factor-alpha/therapeutic use
18.
Am J Trop Med Hyg ; 59(6): 852-8, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9886187

ABSTRACT

Previous studies demonstrated that Plasmodium yoelii 17XL, a lethal strain of rodent malaria, causes a syndrome in SW mice that resembles human cerebral malaria. The mouse brain pathology is characterized by cytoadherence of parasitized erythrocytes. Here, the possible mechanisms mediating cerebral malaria in this model were studied and the results were compared with a nonlethal strain of this parasite, P. yoelii 17XNL (nonlethal), which does not cause cerebral malaria. Immunostaining for intercellular adhesion molecule-1 (ICAM-1) revealed an increase in expression of this protein in the small venules and capillaries of the brains of infected mice that increased with time after infection. Staining was more pronounced during the lethal infection than the nonlethal infection. Some staining with monoclonal antibody to vascular cell adhesion molecule-1 was also observed, but it was quantitatively less than ICAM-1 staining and was limited to larger venules. During the lethal infection, levels of tumor necrosis factor-alpha (TNF-alpha) increased rapidly, peaking on day 4. In contrast, mice infected with nonlethal P. yoelii had a slower serum TNF-alpha response that peaked on day 10, prior to the maximum parasitemia. In addition, mice with a targeted disruption of the TNF-alpha gene (TNF-alpha-/- mice) were infected with the lethal and nonlethal strains of P. yoelii 17X. The TNF-alpha-/- mice infected with the nonlethal parasite had significantly higher levels of parasitemia than controls, whereas TNF-alpha-/- mice infected with the lethal strain had slightly higher levels of infected erythrocytes but were equally susceptible to death from this infection. Thus, TNF-alpha does not appear to be essential in mediating death. These results demonstrate that P. yoelii 17XL infection has features in common with human cerebral malaria and suggest that this model may be useful in testing strategies to alleviate this syndrome.


Subject(s)
Disease Models, Animal , Intercellular Adhesion Molecule-1/biosynthesis , Malaria, Cerebral/metabolism , Plasmodium yoelii , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Brain/metabolism , Endothelium, Vascular/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Vascular Cell Adhesion Molecule-1/biosynthesis
19.
Proc Natl Acad Sci U S A ; 94(23): 12557-61, 1997 Nov 11.
Article in English | MEDLINE | ID: mdl-9356488

ABSTRACT

Immunological functions were analyzed in mice lacking granulocyte/macrophage colony-stimulating factor (GM-CSF). The response of splenic T cells to allo-antigens, anti-mouse CD3 mAb, interleukin 2 (IL-2), or concanavalin A was comparable in GM-CSF +/+ and GM-CSF -/- mice. To investigate the responses of CD8(+) and CD4+ T cells against exogenous antigens, mice were immunized with ovalbumin peptide or with keyhole limpet hemocyanin (KLH). Cytotoxic CD8+ T cells with specificity for ovalbumin peptide could not be induced in GM-CSF -/- mice. After immunization with KLH, there was a delay in IgG generation, particularly IgG2a, in GM-CSF -/- mice. Purified CD4+ T cells from GM-CSF -/- mice immunized with KLH showed impaired proliferative responses and produced low amounts of interferon-gamma (IFN-gamma) and IL-4 when KLH-pulsed B cells or spleen cells were used as antigen presenting cells (APC). When enriched dendritic cells (DC) were used as APC, CD4+ T cells from GM-CSF -/- mice proliferated as well as those from GM-CSF +/+ mice and produced high amounts of IFN-gamma and IL-4. To analyze the rescue effect of DC on CD4(+) T cells, supernatants from (i) CD4(+) T cells cultured with KLH-pulsed DC or (ii) DC cultured with recombinant GM-CSF were transferred to cultures of CD4(+) T cells and KLH-pulsed spleen cells from GM-CSF -/- mice. Supernatants from both DC sources contained a factor or factors that restored proliferative responses and IFN-gamma production of CD4(+) T cells from GM-CSF -/- mice.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunity, Cellular/genetics , T-Lymphocytes/immunology , Animals , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Mice , Mice, Knockout , T-Lymphocyte Subsets/immunology
20.
Nature ; 389(6651): 610-4, 1997 Oct 09.
Article in English | MEDLINE | ID: mdl-9335502

ABSTRACT

Obesity is highly associated with insulin resistance and is the biggest risk factor for non-insulin-dependent diabetes mellitus. The molecular basis of this common syndrome, however, is poorly understood. It has been suggested that tumour necrosis factor (TNF)-alpha is a candidate mediator of insulin resistance in obesity, as it is overexpressed in the adipose tissues of rodents and humans and it blocks the action of insulin in cultured cells and whole animals. To investigate the role of TNF-alpha in obesity and insulin resistance, we have generated obese mice with a targeted null mutation in the gene encoding TNF-alpha and those encoding the two receptors for TNF-alpha. The absence of TNF-alpha resulted in significantly improved insulin sensitivity in both diet-induced obesity and that resulting for the ob/ob model of obesity. The TNFalpha-deficient obese mice had lower levels of circulating free fatty acids, and were protected from the obesity-related reduction in the insulin receptor signalling in muscle and fat tissues. These results indicate that TNF-alpha is an important mediator of insulin resistance in obesity through its effects on several important sites of insulin action.


Subject(s)
Insulin Resistance , Muscle Proteins , Obesity/metabolism , Tumor Necrosis Factor-alpha/physiology , Adipose Tissue/metabolism , Animals , Fatty Acids/blood , Glucose/metabolism , Glucose Tolerance Test , Glucose Transporter Type 4 , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Monosaccharide Transport Proteins/metabolism , Mutation , Phosphorylation , Receptor, Insulin/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Triglycerides/blood , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...