Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Pathol Int ; 57(7): 413-20, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17587240

ABSTRACT

The role of chemokines, especially CXCL10/interferon-gamma-inducible protein 10 kDa (IP-10), a chemokine to attract CXCR3(+) T-helper 1-type CD4(+) T cells, is largely unknown in the pathophysiology of inflammatory bowel disease; ulcerative colitis and Crohn's disease. The authors have earlier shown that IP-10 neutralization protected mice from acute colitis by protecting crypt epithelial cells of the colon. To investigate the therapeutic effect of neutralization of IP-10 on chronic colitis, an anti-IP-10 antibody was injected into mice with newly established murine AIDS (MAIDS) colitis. Anti-IP-10 antibody treatment reduced the number of colon infiltrating cells when compared to those mice given a control antibody. The treatment made the length of the crypt of the colon greater than control antibody. The number of Ki67(+) proliferating epithelial cells was increased by the anti-IP-10 antibody treatment. Terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)(+) apoptotic cells were observed in the epithelial cells of the luminal tops of crypts in control MAIDS colitis, whereas TUNEL(+) apoptotic epithelial cells were rarely observed with anti-IP-10 antibody treatment. In conclusion, blockade of IP-10 attenuated MAIDS colitis through blocking cellular trafficking and protecting intestinal epithelial cells, suggesting that IP-10 plays a key role in the development of inflammatory bowel disease as well as in chronic experimental colitis.


Subject(s)
Chemokines, CXC/antagonists & inhibitors , Colitis/prevention & control , Enterocytes/pathology , Murine Acquired Immunodeficiency Syndrome/prevention & control , Animals , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Chemokine CXCL10 , Chemokines, CXC/immunology , Chronic Disease , Colitis/metabolism , Colon/drug effects , Colon/metabolism , Colon/pathology , Disease Models, Animal , Enterocytes/drug effects , Enterocytes/metabolism , Female , Fluorescent Antibody Technique, Indirect , In Situ Nick-End Labeling , Ki-67 Antigen/metabolism , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/metabolism , Protein Transport/drug effects
2.
J Immunol ; 172(8): 4917-25, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15067071

ABSTRACT

We describe successful immunotherapy of murine AIDS (MAIDS) in C57BL/6J mice based on the elimination of replicating CD4(+) regulator T cells. We demonstrate that a single injection of the antimitotic drug vinblastine (Vb) given 14 days postinfection (p.i.) with LP-BM5 can prevent MAIDS progression. Treatment with anti-CD4 mAb at 14 days p.i. is similarly able to prevent MAIDS. Treatment at other time points with Vb or anti-CD4 mAb is ineffective. The effect is based on ablation of a replicating dominantly suppressive CD4(+) T cell population, as indicated by adoptive transfer and in vivo depletion experiments using mAbs against CD4 as well as combinations of mAbs against the known regulatory cell surface markers CD25, GITR, and CTLA-4. Cell surface marker analysis shows a population of CD4(+)CD25(+) cells arising shortly before day 14 p.i. Cytokine analyses show a peak in IL-10 production from day 12 to day 16 p.i. MAIDS-infected mice also have CD4(+) T cells with significantly higher expression levels of CD38 and particularly CD69, which have been demonstrated to be regulator T cell markers in the Friend retroviral model. The immunotherapy appears to prevent disease progression, although no protection against reinfection with LP-BM5 is generated. These data define a new therapy for murine retroviral infection, which has potential for use in other diseases where T regulator cell-mediated immunosuppression plays a role in the disease process.


Subject(s)
CD4-Positive T-Lymphocytes/pathology , Lymphocyte Depletion/methods , Murine Acquired Immunodeficiency Syndrome/immunology , Murine Acquired Immunodeficiency Syndrome/prevention & control , Animals , Antibodies, Blocking/administration & dosage , Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Cycle/drug effects , Cell Cycle/immunology , Disease Progression , Drug Administration Schedule , Female , Growth Inhibitors/administration & dosage , Growth Inhibitors/therapeutic use , Immunization Schedule , Immunization, Secondary , Injections, Intraperitoneal , Interleukin-10/antagonists & inhibitors , Interleukin-10/biosynthesis , Interleukin-4/antagonists & inhibitors , Interleukin-4/biosynthesis , Lectins, C-Type , Leukemia Virus, Murine/immunology , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/drug therapy , Murine Acquired Immunodeficiency Syndrome/pathology , Receptors, Interleukin-2/biosynthesis , Spleen/immunology , Spleen/pathology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Vinblastine/administration & dosage , Vinblastine/therapeutic use , Viral Load
3.
FASEB J ; 16(13): 1755-63, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12409318

ABSTRACT

LP-BM5 murine leukemia virus (MuLV) infection causes murine acquired immunodeficiency syndrome (MAIDS), a disease characterized by varied functional abnormalities of immunocompetent cells. We found that MAIDS progression was severely retarded in IL-15 transgenic (Tg) mice constructed with cDNA encoding secretable IL-15 under the control of an MHC class I promoter. Several immune defects, including impaired natural killer activity, depressed IFN-gamma production by T cells stimulated with anti-T cell receptor cross-linking, and increased susceptibility to Mycobacterium bovis infection, were prevented in IL-15 Tg mice inoculated with LP-BM5 MuLV. Cytotoxic T lymphocyte response to a highly antigenic 10-mer peptide encoded by LP-BM5-defective virus gag p12 gene was detected in the spleen and peritoneal exudate cells from IL-15 Tg mice infected with LP-BM5 MuLV. Intramuscular injection of cDNA encoding secretable IL-15 also prevented the development of MAIDS. These results indicate that IL-15 prevents the progression of MAIDS and may provide insight into an immunotherapeutic approach using the IL-15 gene for controlling retrovirus-induced immunodeficiency.


Subject(s)
Interleukin-15/physiology , Leukemia Virus, Murine , Murine Acquired Immunodeficiency Syndrome/prevention & control , Animals , Female , Gene Expression , Genetic Therapy/methods , Interleukin-15/genetics , Interleukin-15/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Murine Acquired Immunodeficiency Syndrome/genetics , Murine Acquired Immunodeficiency Syndrome/mortality , Mycobacterium Infections/immunology , Mycobacterium bovis/immunology , Splenomegaly/pathology , Survival Rate , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Time Factors
4.
Virology ; 278(1): 95-102, 2000 Dec 05.
Article in English | MEDLINE | ID: mdl-11112485

ABSTRACT

We previously showed that certain tyrphostin derivatives, known as protein tyrosine kinase inhibitors, also act as topoisomerase I-specific antagonists and inhibit Moloney murine leukemia virus replication in vitro in acutely and chronically infected cells. However, an accurate portrayal of retroviral-induced disease cannot rely exclusively on extrapolations from in vitro data. Therefore, experiments with animal models are essential for evaluating the efficacy of a specific drug in vivo. In this study, we examined the effect of tyrphostin AG-1387 on murine AIDS (MAIDS) development in C57BL/6J mice injected with the LP-BM5 virus mixture. A single dose of tyrphostin, administered together with or 24 h post virus inoculation, decreased the development of MAIDS symptoms as measured by spleen and lymph node weight, the T-cell response to concanavalin A (con A), and spleen architecture. Furthermore, weekly treatment with tyrphostins totally abolished MAIDS symptoms and prevented the viral infection of the spleen cells as measured by the absence of viral RNA and the restoration of T-cell function in these spleens. These results implicate that prolonged treatment with tyrphostins is needed for the prevention of MAIDS development in infected mice and suggest that it may be applied as a legitimate remedy for the treatment of retroviral-induced diseases.


Subject(s)
Antiviral Agents/therapeutic use , Moloney murine leukemia virus , Murine Acquired Immunodeficiency Syndrome/prevention & control , Tyrphostins/therapeutic use , Animals , Blotting, Northern , Concanavalin A/pharmacology , Disease Models, Animal , Lymph Nodes/pathology , Lymphatic Diseases/drug therapy , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/immunology , Murine Acquired Immunodeficiency Syndrome/virology , RNA, Viral/analysis , Spleen/drug effects , Spleen/pathology , Spleen/virology , Splenomegaly/drug therapy , T-Lymphocytes/drug effects , T-Lymphocytes/pathology
5.
J Acquir Immune Defic Syndr ; 23(3): 209-20, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10839656

ABSTRACT

Anti-HIV-1 combination therapies, including protease and reverse transcriptase inhibitors, can reduce plasma viremia to undetectable levels within the first 2 weeks of treatment. This reduction is followed by a slower decline that primarily results from the presence of viral reservoirs such as CD4+ memory cells, dendritic cells, and macrophages. For this reason, we evaluated a new drug combination therapy that includes a lympholytic drug: (2-fluoro-ara-AMP, fludarabine) to eliminate cells already infected and an antiviral drug (azidothymidine [AZT]) to protect cells not yet infected. We used C57BL/6 mice infected with the retroviral complex LP-BM5, which developed severe immunodeficiency (i.e., murine AIDS), to select the most effective fludarabine regimen to inhibit disease progression, and then to evaluate the efficacy and toxicity of the fludarabine and AZT combinations. The results obtained show that intraperitoneal administration of fludarabine at 3 mg/mouse twice a day for 4 weeks is the most effective regimen in reducing splenomegaly, lymphadenopathy, hypergammaglobulinemia, and proviral DNA content in spleen and lymph nodes and in restoring the architecture of lymph nodes. Subsequently, we evaluated the combined or sequential administration of fludarabine and AZT. The data reported in this paper show that the sequential administration of the two drugs provides additive antiviral effects that reduce spleen and lymph node weights to normal values and proviral DNA content by approximately 95% in all infected organs; the phenotypes of blood T and B cells moved toward control values, although the number of B cells was significantly reduced by fludarabine treatment. Finally, we evaluated the outcome of the disease after suspension or continuation of different treatment regimens. In all treatment groups, the disease progressed and increased proviral DNA content was found in infected organs, but animals receiving the sequential administration of fludarabine and AZT were less affected than those receiving only fludarabine or the simultaneous administration of both. The results obtained suggest that fludarabine could be part of a new therapeutic approach aiming at eradicating HIV from those cells that have been already infected and that are not protected by highly active antiretroviral therapy (HAART).


Subject(s)
Anti-HIV Agents/therapeutic use , Immunosuppressive Agents/therapeutic use , Murine Acquired Immunodeficiency Syndrome/prevention & control , Vidarabine Phosphate/analogs & derivatives , Zidovudine/therapeutic use , Administration, Oral , Animals , Anti-HIV Agents/administration & dosage , Bone Marrow/virology , DNA, Viral/analysis , Drug Therapy, Combination , Female , Flow Cytometry , Immunoglobulin G/blood , Immunophenotyping , Immunosuppressive Agents/administration & dosage , Injections, Intraperitoneal , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/isolation & purification , Liver/pathology , Lymph Nodes/pathology , Lymph Nodes/virology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Proviruses/genetics , Proviruses/isolation & purification , Spleen/pathology , Spleen/virology , Vidarabine Phosphate/administration & dosage , Vidarabine Phosphate/therapeutic use , Zidovudine/administration & dosage
6.
Alcohol Clin Exp Res ; 23(5): 856-62, 1999 May.
Article in English | MEDLINE | ID: mdl-10371406

ABSTRACT

Acquired immune deficiency syndrome (AIDS) is a clinical disorder caused by the human immunodeficiency virus (HIV) after development of severe immunosuppressive changes. Chronic ethanol (EtOH) consumption accentuates the severity of murine AIDS (MAIDS). Because hormone production is often suppressed by chronic EtOH intake, as well as retrovirus infection, we investigated whether hormone supplementation during chronic EtOH consumption contributes to slowing immune dysfunction caused by LP-BM5 infection and/or EtOH use. Because dehydroepiandrosterone sulfate (DHEAS) was previously shown to have immune-enhancing properties during MAIDS, we determined whether DHEAS reduced cytokine dysregulation otherwise exacerbated by chronic EtOH intake during MAIDS. Adult female C57BL/6 mice were infected with LP-BM5 murine retrovirus. Some were fed 40% EtOH in drinking water and agar gel for 16 weeks postinfection. EtOH consumption further inhibited T- and B-cell proliferation beyond suppression due to retrovirus infection. Interleukin (IL)-2 release produced by concanavalin A-stimulated splenocytes was reduced by EtOH use by infected and uninfected mice. DHEAS overcame much of the effects induced by retrovirus infection and/or EtOH use. IL-4 secretion and IL-6 secretion were enhanced. Hepatic vitamin E levels were decreased by murine retrovirus infection, as well as by EtOH use in both uninfected and infected mice. In addition, DHEAS (0.01%) supplementation during MAIDS prevented the further dysregulation of cytokines and hepatic lipid peroxidation due to EtOH intake, partially restored T- and B-cell proliferation, and maintained hepatic vitamin E levels to near normal levels.


Subject(s)
Alcohol Drinking/immunology , Dehydroepiandrosterone Sulfate/pharmacology , Murine Acquired Immunodeficiency Syndrome/immunology , Adult , Alcohol Drinking/adverse effects , Animals , B-Lymphocytes/immunology , Body Weight , Cytokines/immunology , Dehydroepiandrosterone Sulfate/therapeutic use , Enzyme-Linked Immunosorbent Assay , Female , Humans , Leukemia Virus, Murine/immunology , Lipid Peroxidation/drug effects , Lipid Peroxidation/immunology , Liver/chemistry , Liver/immunology , Liver/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/prevention & control , Spleen/cytology , Spleen/immunology , T-Lymphocytes/immunology , Vitamin E/analysis
8.
J Acquir Immune Defic Syndr Hum Retrovirol ; 18(4): 316-22, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9704936

ABSTRACT

LP-BM5 murine leukemia virus (MuLV) infection causes severe immunodeficiency termed murine AIDS (MAIDS). The acyclic nucleoside phosphonates, (R)-9-(2-phosphonylmethoxypropyl)adenine (PMPA) and 9-(2-phosphonylmethoxyethyl)adenine (PMEA) were examined, in comparison with zidovudine (AZT), for their inhibitory effect on the development of MAIDS. Although no significant difference in inhibition of LP-BM5 MuLV replication was identified between PMPA and PMEA in cell cultures, PMPA was obviously less cytotoxic to the host lymphocytes. None of the mice treated in vivo with 5 or 25 mg/kg of PMPA or 25 mg/kg of PMEA developed MAIDS at 5 weeks after viral infection. However at 9 weeks, none of the 25 mg/kg PMPA-treated mice progressed to MAIDS, except for one that developed mild MAIDS, whereas PMEA, even at 100 mg/kg, could not prevent disease progression. MAIDS-associated activation of lymphocytes and viral replication were drastically inhibited by PMPA treatment. These results indicate that PMPA is a highly effective antiretroviral agent in vivo.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/therapeutic use , Leukemia Virus, Murine/drug effects , Murine Acquired Immunodeficiency Syndrome/prevention & control , Organophosphonates , Organophosphorus Compounds/therapeutic use , Adenine/pharmacology , Adenine/therapeutic use , Animals , Anti-HIV Agents/pharmacology , Cell Survival/drug effects , Clone Cells , Female , Leukemia Virus, Murine/physiology , Lymph Nodes/pathology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Organophosphorus Compounds/pharmacology , Specific Pathogen-Free Organisms , Spleen/pathology , Tenofovir , Virus Replication/drug effects , Zidovudine/pharmacology , Zidovudine/therapeutic use
9.
J Virol ; 72(6): 5285-90, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9573306

ABSTRACT

Mouse AIDS (MAIDS) induced in C57BL/6 mice by infection with a replication-defective retrovirus (Du5H) combines extensive lymphoproliferation and profound immunodeficiency. Although B cells are the main target of viral infection, recent research has focused on CD4(+) T cells, the activation of which is a key event in MAIDS induction and progression. A preliminary observation of increased expression of B7 molecules on B cells in MAIDS prompted us to address the possible involvement of the CD28/B7 costimulatory pathway in MAIDS. Mice infected with the MAIDS-inducing viral preparation were treated with murine fusion protein CTLA4Ig (3 x 50 microg/week given intraperitoneally), a competitive inhibitor of physiological CD28-B7 interactions. In CTLA4Ig-treated animals, the onset of the disease was delayed, lymphoproliferation progressed at a much slower rate than in untreated mice, and the loss of in vitro responsiveness to mitogens was reduced. Relative expression of Du5H did not differ between treated and untreated animals. These results suggest that the CD28/B7 costimulatory pathway contributes to MAIDS development.


Subject(s)
Antigens, Differentiation/administration & dosage , B-Lymphocytes/immunology , B7-1 Antigen/immunology , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Immunity, Cellular , Immunoconjugates , Immunosuppressive Agents/administration & dosage , Murine Acquired Immunodeficiency Syndrome/immunology , Abatacept , Animals , Antigens, CD , Antigens, Differentiation/immunology , CTLA-4 Antigen , Mice , Murine Acquired Immunodeficiency Syndrome/prevention & control
10.
Cell Immunol ; 172(1): 126-34, 1996 Aug 25.
Article in English | MEDLINE | ID: mdl-8806815

ABSTRACT

Murine retrovirus infection causes an aberrant stimulation of several subsets of T helper 2 cells identified by their T cell receptors (TCR). C57BL/6 mice were treated with synthetic peptides based upon different human TCR V beta CDR1 sequences following experimental infection with the murine retrovirus. Previous studies established that retrovirally infected mice produced autoantibodies to certain of these peptides, and their administration after infection diminished many of the cytokine abnormalities induced by the virus. This study determined whether the complete 16-mer synthetic peptides modeling the V beta CDR1/FR3 were required, and whether admixture of autoantigenic peptides synergized immune preservation. Treatment with complete TCR pep beta 3 and pep V beta 5.2 peptide alone and combined largely prevented the retrovirus-induced reduction in B and T cell proliferation and Th1 cytokine secretion while suppressing excessive production of Th2 cytokines, which are stimulated by retrovirus infection. Treatment with overlapping short peptides corresponding to the N-terminal 11-mer and C-terminal 12-mer did not significantly prevent the immune dysfunction in retrovirus-infected mice. These data suggest that immune dysfunction and abnormal cytokine production, induced by murine retrovirus infection, were largely prevented by TCR V beta CDR1 peptides, and the complete CDR1 in association with the five residues from FR2 was required.


Subject(s)
B-Lymphocytes/immunology , Leukemia Virus, Murine/immunology , Murine Acquired Immunodeficiency Syndrome/prevention & control , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Body Weight , Cell Division , Cells, Cultured , Concanavalin A/pharmacology , Cytokines/immunology , Female , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mitogens/pharmacology , Molecular Sequence Data , Murine Acquired Immunodeficiency Syndrome/immunology , Murine Acquired Immunodeficiency Syndrome/pathology , Peptides/chemical synthesis , Peptides/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Vaccination
11.
Cell Immunol ; 169(1): 24-9, 1996 Apr 10.
Article in English | MEDLINE | ID: mdl-8612290

ABSTRACT

Infection of certain strains of mice, such as C57BL/6 and C57BL/10 [B10], with LP-BM5 murine leukemia virus (MuLV) rapidly causes a profound and lethal immune suppression. The H2d congenic strain of B10, B10.D2, is resistant to disease, but B10 x B10.D2 F1 mice are susceptible, indicating that disease sensitivity is dominant. To determine whether disease resistance could be adoptively transferred to a sensitive host, radiation chimeras (B10.D2 --> B10 x B10.D2 F1 and F1 --> F1) were challenged with LP-BM5 virus. Infected B10.D2 --> F1 chimeras showed no loss of immune function, whereas F1 --> F1 chimeras infected with LP-BM5 MuLV developed MAIDS and became completely immune suppressed. These results, coupled with previous studies, indicate resistance or sensitivity to disease is an inherent property of the hematopoietic system that can be transferred by bone marrow grafts.


Subject(s)
Immunity, Innate/immunology , Immunosuppression Therapy , Immunotherapy, Adoptive , Leukemia Virus, Murine , Leukemia, Experimental/prevention & control , Murine Acquired Immunodeficiency Syndrome/prevention & control , Tumor Virus Infections/prevention & control , Animals , Bone Marrow Transplantation/immunology , Leukemia, Experimental/immunology , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/immunology , Tumor Virus Infections/immunology
12.
Immunology ; 87(2): 198-204, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8698380

ABSTRACT

C57BL/6 mice were injected with different doses of human T-cell receptor (TCR) V beta 8.1 CDR1 peptide at different times after murine retrovirus (LP-BM5) infection. Injection with TCR V beta 8.1 CDR1 peptide largely prevented the retrovirus-induced reduction in B- and T-cell proliferation, and T-helper 1 (Th1) cytokines [interleukin-2 (IL-2) and interferon-gamma (IFN-gamma)] secretion. It also suppressed T-helper 2 (Th2) cytokines (IL-6 and IL-10) production, which was stimulated by retrovirus infection. These effects were accomplished using at least 100 micrograms of peptide per mouse and the most effective dose of peptide had to be given within 4 weeks after retrovirus infection. Immunization with doses above 100 micrograms/mouse as long as 4 weeks postinfection maintained natural killer (NK) cell activity during retrovirus infection. Reducing the dose of peptide or delaying it until the disease progressed towards early murine acquired immune deficiency syndrome (AIDS) allowed development of immune dysfunction. These studies provide data suggesting that immune dysfunction, induced by murine retrovirus infection, was largely prevented by TCR V beta CDR1 peptide injection.


Subject(s)
Immunization Schedule , Murine Acquired Immunodeficiency Syndrome/immunology , Receptors, Antigen, T-Cell, alpha-beta , Receptors, Antigen, T-Cell/immunology , Adjuvants, Immunologic , Animals , Body Weight , Cell Culture Techniques , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Dose-Response Relationship, Immunologic , Female , Immunity, Cellular , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/prevention & control , Murine Acquired Immunodeficiency Syndrome/therapy , Spleen/immunology
13.
Arch Virol ; 129(1-4): 155-66, 1993.
Article in English | MEDLINE | ID: mdl-8385912

ABSTRACT

Among murine leukemia viruses (MuLV) present in the LP-BM5 virus mixture, the agent etiologic for an acquired immunodeficiency syndrome (MAIDS) is replication defective, containing only a single open reading frame which includes all of gag. The Gag polyprotein encoded by the defective virus, termed BM5def, differs most in p12 from that of nonpathogenic ecotropic virus (BM5eco). As one approach to examining the role of p12 in disease, the ecotropic and defective virus forms of the protein, synthesized in bacteria, were used to immunize three strains of mice differing in their sensitivity to MAIDS. In each strain, both proteins elicited substantial antibody responses that were cross-reactive with either p12 and recognized the proteins as part of intact viral Gag polyproteins. Immunization with either p12 before infection with LP-BM5 viruses had no effect on the sensitivity or resistance of mice to MAIDS or on the extent of helper virus spread. The variant p12 of BM5def, when presented on its own, is thus not a crucial antigenic determinant of disease. Alternative mechanisms by which BM5def may contribute to MAIDS are discussed.


Subject(s)
Defective Viruses/immunology , Gene Products, gag/immunology , Leukemia Virus, Murine/immunology , Murine Acquired Immunodeficiency Syndrome/prevention & control , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Base Sequence , Immunization , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Murine Acquired Immunodeficiency Syndrome/immunology
14.
Antiviral Res ; 16(4): 307-26, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1663732

ABSTRACT

LP-BM5 MuLV infection of C57BL/6 mice induces a well characterized, lymphoproliferative, immunodeficiency disease (MAIDS), which is useful for evaluation of potential antiviral agents, because of the reproducibility of virological and clinical endpoints. This MAIDS retrovirus model was used to evaluate 3'azido-2,3'dideoxythymidine (AZT), using different doses, methods of administration and timing for initiation and continuation of therapy. AZT therapy 1 mg/ml in the drinking water given 30 days prior to virus challenge, and continued for 16 weeks, prevented LP-BM5 MuLV dissemination and disease in 13 of 15 treated mice. Efficacy was dose dependent for AZT concentrations of 1, 0.5, and 0.1 mg/ml in drinking water. One mg/ml AZT was most effective in preventing infection if therapy was begun within days prior to virus challenge or within the first four hours after virus inoculation. If treatment was initiated later, disease was delayed. Continuous infusion of AZT, 25 micrograms/h, was effective since virus was not detected in spleens of any mice during the 21 days of AZT treatment. However, after treatment was stopped treated mice became virus positive and disease progressed. Likewise, AZT administration at 1 mg/ml in the drinking water for only 21 days post virus inoculation (p.i.), was not sufficient to prevent virus dissemination or disease.


Subject(s)
Murine Acquired Immunodeficiency Syndrome/prevention & control , Zidovudine/administration & dosage , Administration, Oral , Animals , Chromatography, High Pressure Liquid , Immunoglobulins/analysis , Infusions, Intravenous , Leukemia Virus, Murine , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/immunology , Murine Acquired Immunodeficiency Syndrome/microbiology , Murine Acquired Immunodeficiency Syndrome/pathology , Spleen/pathology , Zidovudine/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...