Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 260
Filter
1.
Cytokine ; 144: 155596, 2021 08.
Article in English | MEDLINE | ID: mdl-34078571

ABSTRACT

Interleukin-1α (IL-1α) is an alarmin involved in the recruitment of macrophages and neutrophils during tissue inflammation. IL-1α can undergo cleavage by proteases, such as calpain-1, that enhances IL-1α binding to its receptor, although proteolytic cleavage is not necessary for biological activity. Macrophages and neutrophils are involved in the retinal inflammation associated with development of AIDS-related human cytomegalovirus (HCMV) retinitis. We therefore performed studies to test the hypothesis that IL-1α gene expression is stimulated intraocularly during retinitis development using two mouse models of murine cytomegalovirus (MCMV) retinitis that differ in method of immunosuppression, one by retrovirus-induced immunosuppression (MAIDS) and the other by corticosteroid-induced immunosuppression. MCMV-infected eyes of groups of retinitis-susceptible mice with MAIDS of 10 weeks duration (MAIDS-10 mice) and retinitis-susceptible corticosteroid-treated mice showed significant stimulation of IL-1α mRNA. Western blot analysis confirmed IL-1α protein production within the MCMV-infected eyes of MAIDS-10 mice. Whereas significant intraocular calpain-1 mRNA and protein production were also observed within MCMV-infected eyes of MAIDS-10 mice, the MCMV-infected eyes of retinitis-susceptible corticosteroid-treated mice showed a pattern of mRNA synthesis equivalent to that found within the MCMV-infected eyes of healthy mice that fail to develop retinitis. Our findings suggest a role for the alarmin IL-1α in the pathogenesis of MCMV retinitis in immunosuppressed mice. These findings may extend to the pathogenesis of HCMV retinitis in patients with AIDS or other forms of immunosuppression.


Subject(s)
Cytomegalovirus Retinitis/immunology , Interleukin-1alpha/immunology , Murine Acquired Immunodeficiency Syndrome/immunology , Muromegalovirus/immunology , Retina/immunology , Animals , Disease Models, Animal , Female , Immune Tolerance/immunology , Immunosuppression Therapy/methods , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA, Messenger/immunology
2.
J Neuroimmune Pharmacol ; 16(2): 437-453, 2021 06.
Article in English | MEDLINE | ID: mdl-32627098

ABSTRACT

HIV affects 37 million people worldwide, 25-69% of which develop HIV-associated neurocognitive disorders (HAND) regardless of antiviral treatment. HIV infection of the brain decreases cognitive function, disrupts/impairs learning and memory, and reduces quality of life for those affected. HIV-induced neuroinflammation has been associated with viral proteins such as gp120 and Tat, which remain elevated in the CNS even in patients with low peripheral viremia counts. In this study, we examined the effects of gp120 on neuroinflammation in immunodeficient vs. immunocompetent states by examining neuroinflammatory markers in gp120tg mice with or without systemic immunodeficiency caused by murine retroviral administration (LP-BM5 murine AIDS). Changes in inflammatory cytokine/chemokine mRNA expression was complex and dependent upon expression of gp120 protein, immunodeficiency status, brain region (hippocampus, frontal lobe, or striatum), and age. Gp120 expression reduced hippocampal synaptophysin expression but did not affect animals' learning/memory on the spontaneous T-maze test in our experimental conditions. Our results emphasize the critical role of the neuroinflammatory micro-environment and the peripheral immune system context in which gp120 acts. Multiple factors, particularly system-level differences in the immune response of different brain regions, need to be considered when developing treatment for HAND. Graphical Abstract.


Subject(s)
AIDS Dementia Complex/immunology , AIDS Dementia Complex/pathology , HIV Envelope Protein gp120 , Murine Acquired Immunodeficiency Syndrome/immunology , Murine Acquired Immunodeficiency Syndrome/pathology , Animals , Brain/immunology , Brain/pathology , Mice , Mice, Transgenic
3.
PLoS Pathog ; 16(11): e1009032, 2020 11.
Article in English | MEDLINE | ID: mdl-33156834

ABSTRACT

Human cytomegalovirus (HCMV) is an opportunistic human herpesvirus that causes a sight-threatening retinitis in immunosuppressed patients, especially those with AIDS. Using an established model of experimental murine cytomegalovirus (MCMV) retinitis in mice with retrovirus-induced immunodeficiency (MAIDS), we have been attempting to define with greater clarity the immunologic mechanisms that contribute to the progression of AIDS-related HCMV retinitis in the unique immunosuppressive setting of HIV infection. Toward this end, we provide herein a comprehensive assessment of immune response gene expression during the onset and development of MAIDS-related MCMV retinitis employing NanoString nCounter. In so doing, we analyzed and compared the intraocular expressions of 561 immune response genes within MCMV-infected eyes of groups of healthy mice, MCMV-infected mice with MAIDS of 4 weeks' (MAIDS-4) duration, and MCMV-infected eyes of mice with MAIDS of 10 weeks' (MAIDS-10) duration. These animal groups show a progression of retinal disease from absolute resistance to retinitis development in healthy mice to the development of classic full-thickness retinal necrosis in MAIDS-10 mice but through an intermediate stage of retinal disease development in MAIDS-4 mice. Our findings showed that increased susceptibility to MCMV retinitis during the progression of MAIDS is associated with robust upregulation or downregulation of a surprisingly large number of immune response genes that operate within several immune response pathways often unique to each animal group. Analysis of 14 additional immune response genes associated with programmed cell death pathways suggested involvement of necroptosis and pyroptosis during MAIDS-related MCMV retinitis pathogenesis. Use of the NanoString nCounter technology provided new and unexpected information on the immunopathogenesis of retinitis within MCMV-infected eyes of mice with retrovirus-induced immunosuppression. Our findings may provide new insights into the immunologic events that operate during the pathogenesis of AIDS-related HCMV retinitis.


Subject(s)
Cytomegalovirus Retinitis/immunology , Cytomegalovirus/immunology , HIV Infections/immunology , Immunity/genetics , Murine Acquired Immunodeficiency Syndrome/immunology , Muromegalovirus/immunology , Animals , Cytomegalovirus Retinitis/virology , Disease Models, Animal , Eye/immunology , Eye/virology , Female , Gene Expression Profiling , HIV Infections/virology , Humans , Immunosuppression Therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/virology
4.
J Virol ; 92(18)2018 09 15.
Article in English | MEDLINE | ID: mdl-29976680

ABSTRACT

AIDS-related human cytomegalovirus retinitis remains the leading cause of blindness among untreated HIV/AIDS patients worldwide. To study mechanisms of this disease, we used a clinically relevant animal model of murine cytomegalovirus (MCMV) retinitis with retrovirus-induced murine AIDS (MAIDS) that mimics the progression of AIDS in humans. We found in this model that MCMV infection significantly stimulates ocular suppressor of cytokine signaling 1 (SOCS1) and SOCS3, host proteins which hinder immune-related signaling by cytokines, including antiviral type I and type II interferons. The present study demonstrates that in the absence of retinal disease, systemic MCMV infection of mice without MAIDS, but not in mice with MAIDS, leads to mild stimulation of splenic SOCS1 mRNA. In sharp contrast, when MCMV is directly inoculated into the eyes of retinitis-susceptible MAIDS mice, high levels of intraocular SOCS1 and SOCS3 mRNA and protein are produced which are associated with significant intraocular upregulation of gamma interferon (IFN-γ) and interleukin-6 (IL-6) mRNA expression. We also show that infiltrating macrophages, granulocytes, and resident retinal cells are sources of intraocular SOCS1 and SOCS3 protein production during development of MAIDS-related MCMV retinitis, and SOCS1 and SOCS3 mRNA transcripts are detected in retinal areas histologically characteristic of MCMV retinitis. Furthermore, SOCS1 and SOCS3 are found in both MCMV-infected cells and uninfected cells, suggesting that these SOCS proteins are stimulated via a bystander mechanism during MCMV retinitis. Taken together, our findings suggest a role for MCMV-related stimulation of SOCS1 and SOCS3 in the progression of retinal disease during ocular, but not systemic, MCMV infection.IMPORTANCE Cytomegalovirus infection frequently causes blindness in untreated HIV/AIDS patients. This virus manipulates host cells to dysregulate immune functions and drive disease. Here, we use an animal model of this disease to demonstrate that cytomegalovirus infection within eyes during retinitis causes massive upregulation of immunosuppressive host proteins called SOCS. As viral overexpression of SOCS proteins exacerbates infection with other viruses, they may also enhance cytomegalovirus infection. Alternatively, the immunosuppressive effect of SOCS proteins may be protective against immunopathology during cytomegalovirus retinitis, and in such a case SOCS mimetics or overexpression treatment strategies might be used to combat this disease. The results of this work therefore provide crucial basic knowledge that contributes to our understanding of the mechanisms of AIDS-related cytomegalovirus retinitis and, together with future studies, may contribute to the development of novel therapeutic targets that could improve the treatment or management of this sight-threatening disease.


Subject(s)
Cytomegalovirus Retinitis/immunology , Immunosuppression Therapy , Murine Acquired Immunodeficiency Syndrome/immunology , Muromegalovirus/immunology , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Animals , Cytomegalovirus Retinitis/virology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Murine Acquired Immunodeficiency Syndrome/virology , Muromegalovirus/isolation & purification , Spleen/immunology , Suppressor of Cytokine Signaling 1 Protein/immunology , Suppressor of Cytokine Signaling 3 Protein/immunology
5.
J Med Food ; 21(7): 689-700, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29862890

ABSTRACT

The immune response is stimulated to protect the body from external antigens and is controlled by several types of immune cells. In the present study, the immunomodulatory effects of Curcuma longa L., purple sweet potato, and mixtures of the two (CPM) were investigated in C57BL/6 mice infected with LP-BM5 murine leukemia virus (MuLV). Mice were divided into seven groups as follows: normal control, infected control (LP-BM5 MuLV infection), positive control (LP-BM5 MuLV infection+dietary supplement of red ginseng 300 mg/kg body weight), the original powder of C. longa L. (C; LP-BM5 MuLV infection+dietary supplement of C 189 mg/kg body weight), the original powder of purple sweet potato (P; LP-BM5 MuLV infection+dietary supplement of P 1811 mg/kg body weight), CPM Low (CPL; LP-BM5 MuLV infection+CPM 2 g/kg body weight), and CPM High (CPH; LP-BM5 MuLV infection+CPM 5 g/kg body weight). Dietary supplementation lasted for 12 weeks. Dietary supplementation of CPM inhibited LP-BM5 MuLV-induced lymphadenopathy and splenomegaly and inhibited reduction of messenger RNA (mRNA) expression of major histocompatibility complex (MHC) I and II. Moreover, CPM reduced the decrease in T- and B cell proliferation, reduced the population of CD4(+)/CD8(+) T cells, and remedied the unbalanced production of T helper-1 (Th1)/T helper-2 (Th2) cytokines in LP-BM5 MuLV-infected mice. In addition, CPM inhibited reduction of phagocytosis in peritoneal macrophages and decreased serum levels of immunoglobulin A (IgA), immunoglobulin E (IgE), and immunoglobulin G (IgG). These results suggest that CPM had a positive effect on immunomodulation in C57BL/6 mice induced by LP-BM5 leukemia retrovirus infection.


Subject(s)
Curcuma/chemistry , Ipomoea batatas/chemistry , Leukemia Virus, Murine/physiology , Murine Acquired Immunodeficiency Syndrome/drug therapy , Murine Acquired Immunodeficiency Syndrome/immunology , Plant Extracts/administration & dosage , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , HIV Infections/drug therapy , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/physiology , Humans , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Major Histocompatibility Complex/drug effects , Male , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/genetics , Phagocytosis/drug effects , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology
6.
J Neuroimmunol ; 319: 117-129, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29526406

ABSTRACT

Opioid use accelerates neurocognitive impairment in HIV/AIDS patients. We assessed the effect of chronic morphine treatment and LP-BM5/murine AIDS (MAIDS) infection on cognition, cytokine production, and type 1 interferon (IFN) expression in the murine CNS. Morphine treatment decreased expression of pro-inflammatory factors (CCL5, iNOS) and reduced cognitive performance in LP-BM5-infected mice, correlating to increased hippocampal viral load and a blunted type 1 IFN response. In the striatum, morphine reduced viral load while increasing IFN-α RNA expression. Our results suggest that differentially regulated type 1 IFN responses may contribute to distinct regional outcomes in the hippocampus and striatum in LP-BM5/MAIDS.


Subject(s)
AIDS Dementia Complex/immunology , Analgesics, Opioid/toxicity , Hippocampus/drug effects , Hippocampus/immunology , Morphine/toxicity , Animals , Cognitive Dysfunction/etiology , Hippocampus/virology , Interferon Type I/immunology , Interferon Type I/metabolism , Male , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/immunology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/immunology , Viral Load
7.
Cytokine ; 97: 38-41, 2017 09.
Article in English | MEDLINE | ID: mdl-28558309

ABSTRACT

AIDS-related human cytomegalovirus retinitis remains a leading cause of blindness worldwide. We compared two C57BL/6 mouse models of experimental murine cytomegalovirus (MCMV) retinitis for intraocular expression of suppressors of cytokine signaling (SOCS)1 and SOCS3, host proteins that are inducible negative feedback regulators of cytokine signaling. These mouse models differed in method of immune suppression, one by retrovirus-induced immune suppression (MAIDS) and the other by corticosteroid-induced immune suppression. Following subretinal injection of MCMV to induce retinitis, intraocular SOCS1 and SOCS3 were only mildly stimulated, and often without significance, within MCMV-infected eyes during the progression of MCMV retinitis in corticosteroid-immunosuppressed mice, contrary to MCMV-infected eyes of mice with MAIDS that showed significant high stimulation of SOCS1 and SOCS3 expression in agreement with previous findings. Frequency and severity of retinitis as well as amounts of intraocular infectious MCMV in corticosteroid-immunosuppressed mice were also unexpectedly lower than values previously reported for MAIDS animals during MCMV retinitis. These data reveal a major difference between two mouse models of experimental MCMV retinitis and suggest a possible link between the amplitude of SOCS1 and SOCS3 stimulation and severity of disease in these models.


Subject(s)
Adrenal Cortex Hormones/administration & dosage , Cytomegalovirus Retinitis/immunology , Immune Tolerance , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Adrenal Cortex Hormones/immunology , Animals , Cytomegalovirus Retinitis/chemically induced , Cytomegalovirus Retinitis/virology , Disease Models, Animal , Disease Progression , Eye/immunology , Eye/metabolism , Eye/virology , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/immunology , Muromegalovirus/isolation & purification
8.
Virology ; 499: 9-22, 2016 12.
Article in English | MEDLINE | ID: mdl-27632561

ABSTRACT

Monocytic myeloid-derived suppressor cells (M-MDSCs) were increased during LP-BM5 retroviral infection, and were capable of suppressing not only T-cell, but also B-cell responses. In addition to previously demonstrating iNOS- and VISTA-dependent M-MDSC mechanisms, in this paper, we detail how M-MDSCs utilized soluble mediators, including the reactive oxygen and nitrogen species superoxide, peroxynitrite, and nitric oxide, and TGF-ß, to suppress B cells in a predominantly contact-independent manner. Suppression was independent of cysteine-depletion and hydrogen peroxide production. When two major mechanisms of suppression (iNOS and VISTA) were eliminated in double knockout mice, M-MDSCs from LP-BM5-infected mice were able to compensate using other, soluble mechanisms in order to maintain suppression of B cells. The IL-10 producing regulatory B-cell compartment was among the targets of M-MDSC-mediated suppression.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Murine Acquired Immunodeficiency Syndrome/immunology , Murine Acquired Immunodeficiency Syndrome/metabolism , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Reactive Nitrogen Species , Reactive Oxygen Species , Transforming Growth Factor beta/blood , Animals , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Cell Communication , Immune Tolerance , Male , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Peroxynitrous Acid/metabolism
9.
J Med Food ; 18(8): 882-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26076116

ABSTRACT

The immunomodulatory effects of a dietary supplement of purple sweet potato extract (PSPE) in LP-BM5 murine leukemia virus (MuLV)-induced immune-deficient mice were investigated. Mice were divided into six groups: normal control, infected control (LP-BM5 MuLV infection), positive control (LP-BM5 MuLV infection+dietary supplement of red ginseng 300 mg/kg), purple sweet potato water extract (PSPWE) (LP-BM5 MuLV infection+dietary supplement of PSPE 300 mg/kg), PSP10EE (LP-BM5 MuLV infection+dietary supplement of 10% ethanol PSPE 300 mg/kg), and PSP80EE (LP-BM5 MuLV infection+dietary supplement of 80% ethanol PSPE 300 mg/kg). Dietary supplementation began on the day of LP-BM5 MuLV infection and continued for 12 weeks. Dietary supplementation of PSPE inhibited LP-BM5 MuLV-induced splenomegaly and lymphadenopathy and attenuated the suppression of T- and B-cell proliferation and T helper 1/T helper 2 cytokine imbalance in LP-BM5 MuLV-infected mice. Dietary supplement of PSPE increased the activity of the antioxidant enzymes, superoxide dismutase and glutathione peroxidase. The data suggest that PSPE may ameliorate immune dysfunction due to LP-BM5 MuLV infection by modulating antioxidant defense systems.


Subject(s)
Antioxidants/pharmacology , Immunomodulation/drug effects , Ipomoea batatas/chemistry , Ipomoea batatas/immunology , Leukemia Virus, Murine , Murine Acquired Immunodeficiency Syndrome/diet therapy , Animals , Antioxidants/chemistry , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Cell Proliferation/drug effects , Cytokines/analysis , Disease Models, Animal , Glutathione Peroxidase/metabolism , Leukemia Virus, Murine/pathogenicity , Lymphatic Diseases/diet therapy , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/immunology , Murine Acquired Immunodeficiency Syndrome/pathology , Murine Acquired Immunodeficiency Syndrome/virology , Plant Extracts/pharmacology , Plant Preparations/chemistry , Splenomegaly/diet therapy , Superoxide Dismutase/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
10.
J Virol ; 88(4): 2349-53, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24335302

ABSTRACT

Interferon regulatory factor (IRF) regulation of the type I interferon response has not been extensively explored in murine retroviral infections. IRF-3(-/-) and select IRF-3/7(-/-) mice were resistant to LP-BM5-induced pathogenesis. However, further analyses strongly suggested that resistance could be attributed to strain 129-specific contamination of the known retrovirus resistance gene Fv1. Therefore, caution should be taken when interpreting phenotypes observed in these knockout mice, as strain 129-derived genetic polymorphisms may explain observed differences.


Subject(s)
Disease Models, Animal , Gammaretrovirus/immunology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-7/genetics , Interferon Type I/immunology , Murine Acquired Immunodeficiency Syndrome/immunology , Murine Acquired Immunodeficiency Syndrome/physiopathology , Animals , Electrophoresis , Gammaretrovirus/genetics , Mice , Mice, Knockout , Murine Acquired Immunodeficiency Syndrome/virology , Proteins/genetics , Proteins/immunology , Species Specificity , Statistics, Nonparametric
11.
J Neuroinflammation ; 10: 98, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23902750

ABSTRACT

BACKGROUND: Highly active antiretroviral therapy (HAART) restores inflammatory immune responses in AIDS patients which may unmask previous subclinical infections or paradoxically exacerbate symptoms of opportunistic infections. In resource-poor settings, 25% of patients receiving HAART may develop CNS-related immune reconstitution inflammatory syndrome (IRIS). Here we describe a reliable mouse model to study underlying immunopathological mechanisms of CNS-IRIS. METHODS: Utilizing our HSV brain infection model and mice with MAIDS, we investigated the effect of immune reconstitution on MAIDS mice harboring opportunistic viral brain infection. Using multi-color flow cytometry, we quantitatively measured the cellular infiltrate and microglial activation. RESULTS: Infection with the LP-BM5 retroviral mixture was found to confer susceptibility to herpes simplex virus (HSV)-1 brain infection to normally-resistant C57BL/6 mice. Increased susceptibility to brain infection was due to severe immunodeficiency at 8 wks p.i. and a marked increase in programmed death-1 (PD-1) expression on CD4+ and CD8+ T-cells. Both T-cell loss and opportunistic brain infection were associated with high level PD-1 expression because PD-1-knockout mice infected with LP-BM5 did not exhibit lymphopenia and retained resistance to HSV-1. In addition, HSV-infection of MAIDS mice stimulated peripheral immune cell infiltration into the brain and its ensuing microglial activation. Interestingly, while opportunistic herpes virus brain infection of C57BL/6 MAIDS mice was not itself lethal, when T-cell immunity was reconstituted through adoptive transfer of virus-specific CD3+ T-cells, it resulted in significant mortality among recipients. This immune reconstitution-induced mortality was associated with exacerbated neuroinflammation, as determined by MHC class II expression on resident microglia and elevated levels of Th1 cytokines in the brain. CONCLUSIONS: Taken together, these results indicate development of an immune reconstitution disease within the central nervous system (CNS-IRD). Experimental immune reconstitution disease of the CNS using T-cell repopulation of lymphopenic murine hosts harboring opportunistic brain infections may help elucidate neuroimmunoregulatory networks that produce CNS-IRIS in patients initiating HAART.


Subject(s)
AIDS-Related Opportunistic Infections/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Central Nervous System Viral Diseases/immunology , Herpes Simplex/immunology , Murine Acquired Immunodeficiency Syndrome/immunology , AIDS-Related Opportunistic Infections/mortality , AIDS-Related Opportunistic Infections/pathology , Animals , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Central Nervous System Viral Diseases/mortality , Central Nervous System Viral Diseases/pathology , Herpes Simplex/mortality , Herpes Simplex/pathology , Inflammation/immunology , Inflammation/mortality , Inflammation/pathology , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/mortality , Murine Acquired Immunodeficiency Syndrome/pathology
12.
J Neuroimmunol ; 261(1-2): 37-43, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23726765

ABSTRACT

B6 mice infected with LP-BM5 develop severe immunodeficiency (termed murine acquired immunodeficiency syndrome (MAIDS)) and peripheral neuropathy. To determine whether microglial CD40 is involved in LP-BM5-induced peripheral neuropathy, B6-CD40 knockout (KO) mice and B6-CD40 KO mice adoptively transferred either total leukocytes or B cells were examined for behavioral sensitivity, tissue viral loads, cytokine responses, and the development of MAIDS. All three CD40 KO groups developed MAIDS, the severity of which was correlated with peripheral cytokine responses. CD40 KO mice displayed significantly reduced mechanical hypersensitivity post-infection compared to wild-type mice regardless of cell transfer. These findings support microglial CD40 involvement in LP-BM5-induced peripheral neuropathy.


Subject(s)
CD40 Antigens/immunology , Leukemia Virus, Murine/immunology , Microglia/immunology , Murine Acquired Immunodeficiency Syndrome/immunology , Peripheral Nervous System Diseases/metabolism , Retroviridae/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , B-Lymphocytes/virology , CD40 Antigens/deficiency , Leukocytes/immunology , Leukocytes/pathology , Leukocytes/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Microglia/virology , Murine Acquired Immunodeficiency Syndrome/pathology , Murine Acquired Immunodeficiency Syndrome/virology , Peripheral Nervous System Diseases/immunology , Peripheral Nervous System Diseases/virology , Random Allocation
13.
Cytokine ; 61(3): 862-75, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23415673

ABSTRACT

Interleukin-17 (IL-17), a pro-inflammatory cytokine produced by CD4+ Th17 cells, has been associated with the pathogenesis of several autoimmune diseases including uveitis. The fate of IL-17 during HIV/AIDS, however, remains unclear, and a possible role for IL-17 in the pathogenesis of AIDS-related diseases has not been investigated. Toward these ends, we performed studies using a well-established animal model of experimental murine cytomegalovirus (MCMV) retinitis that develops in C57/BL6 mice with retrovirus-induced immunosuppression (MAIDS). After establishing baseline levels for IL-17 production in whole splenic cells of healthy mice, we observed a significant increase in IL-17 mRNA levels in whole splenic cells of mice with MAIDS of 4-weeks (MAIDS-4), 8-weeks (MAIDS-8), and 10-weeks (MAIDS-10) duration. In contrast, enriched populations of splenic CD4+ T cells, splenic macrophages, and splenic neutrophils exhibited a reproducible decrease in levels of IL-17 mRNA during MAIDS progression. To explore a possible role for IL-17 during the pathogenesis of MAIDS-related MCMV retinitis, we first demonstrated constitutive IL-17 expression in retinal photoreceptor cells of uninfected eyes of healthy mice. Subsequent studies, however, revealed a significant decrease in intraocular levels of IL-17 mRNA and protein in MCMV-infected eyes of MAIDS-10 mice during retinitis development. That MCMV infection might cause a remarkable downregulation of IL-17 production was supported further by the finding that systemic MCMV infection of healthy, MAIDS-4, or MAIDS-10 mice also significantly decreased IL-17 mRNA production by splenic CD4+ T cells. Based on additional studies using IL-10 -/- mice infected systemically with MCMV and IL-10 -/- mice with MAIDS infected intraocularly with MCMV, we propose that MCMV infection downregulates IL-17 production via stimulation of suppressor of cytokine signaling (SOCS)-3 and interleukin-10.


Subject(s)
Cytomegalovirus Retinitis/pathology , Cytomegalovirus Retinitis/virology , Down-Regulation , Interleukin-17/metabolism , Murine Acquired Immunodeficiency Syndrome/immunology , Murine Acquired Immunodeficiency Syndrome/virology , Muromegalovirus/physiology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cytomegalovirus Retinitis/genetics , Cytomegalovirus Retinitis/immunology , Disease Progression , Disease Susceptibility , Eye/metabolism , Female , Immunosuppression Therapy , Interleukin-17/biosynthesis , Interleukin-17/genetics , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/genetics , Murine Acquired Immunodeficiency Syndrome/pathology , Neutrophils/metabolism , Photoreceptor Cells, Vertebrate/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retroviridae/physiology , Spleen/metabolism , Spleen/pathology , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
14.
J Immunol ; 189(7): 3724-33, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22956581

ABSTRACT

Distal symmetrical polyneuropathy is the most common form of HIV infection-associated peripheral neuropathy and is often associated with pain. C57BL/6 (B6) mice infected with LP-BM5, a murine retroviral isolate, develop a severe immunodeficiency syndrome similar to that in humans infected with HIV-1, hence the term murine AIDS. We investigated the induction of peripheral neuropathy after LP-BM5 infection in B6 mice. Infected B6 mice, like HIV-infected humans, exhibited behavioral (increased sensitivity to mechanical and heat stimuli) and pathological (transient loss of intraepidermal nerve fibers) signs of peripheral neuropathy. The levels of viral gag RNA were significantly increased in all tissues tested, including spleen, paw skin, lumbar dorsal root ganglia, and lumbar spinal cord, postinfection (p.i.). Correlated with the development of peripheral neuropathy, the tissue levels of several cytokines, including IFN-γ, IL-1ß, IL-6, and IL-12, were significantly elevated p.i. These increases had cytokine-specific and tissue-specific profiles and kinetics. Further, treatment with the antiretroviral agent zidovudine either significantly reduced or completely reversed the aforementioned behavioral, pathologic, and cytokine changes p.i. These data suggest that LP-BM5 infection is a potential mouse model of HIV-associated distal symmetrical polyneuropathy that can be used for investigating the roles of various cytokines in infection-induced neuropathic pain. Further investigation of this model could give a better understanding of, and lead to more effective treatments for, HIV infection-associated painful peripheral neuropathy.


Subject(s)
Cytokines/biosynthesis , Leukemia Virus, Murine/immunology , Murine Acquired Immunodeficiency Syndrome/immunology , Murine Acquired Immunodeficiency Syndrome/metabolism , Peripheral Nervous System Diseases/immunology , Peripheral Nervous System Diseases/virology , Animals , Cytokines/genetics , Disease Models, Animal , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/virology , Leukemia Virus, Murine/isolation & purification , Leukemia, Experimental/immunology , Leukemia, Experimental/metabolism , Leukemia, Experimental/virology , Male , Mice , Mice, Inbred C57BL , Peripheral Nervous System Diseases/metabolism , RNA, Messenger/biosynthesis , Retroviridae Infections/immunology , Retroviridae Infections/metabolism
15.
Biosci Biotechnol Biochem ; 75(7): 1234-9, 2011.
Article in English | MEDLINE | ID: mdl-21737943

ABSTRACT

The purpose of this study was to determine the effect of isothiocyanates (ITCs) in delaying the progression of the murine immunodeficiency virus to murine AIDS, resulting in increased life span. Furthermore, we investigated the role of ITCs in modulating immune dysfunction caused by LP-BM5 retrovirus infection. Among the tested ITCs, oral administration of sulforaphane (SUL), benzyl isothiocyante (BITC), and phenethyl isothiocyanate (PEITC) showed the inhibition of premature death caused by LP-BM5 retrovirus infection, while indolo[3,2-b] carbazole (ICZ) and indole-3-carbinol (I3C) did not delay the progress of the LP-BM5 retrovirus to murine AIDS. Inhibition of premature death by BITC, PEITC, and SUL could be explained by restoration of the immune system and down regulation of free radicals. Dysfunction of T and B cell mitogenesis caused by retrovirus infection in primary cultured splenocytes has been partially recovered with administration of BITC, PEITC, and SUL. There was a shift from imbalanced cytokine production (increased Th2 and decreased Th1 cell cytokine production) into balanced Th1/Th2 cell secretion of cytokines under administration of these ITCs during the development of murine AIDS. Hepatic vitamin E level was significantly restored by administration of these ITCs, in accordance with reduced hepatic lipid peroxidation levels. This study suggests that certain types of ITCs have beneficial effects in preventing premature death during progression to murine AIDS by restoration of immune dysfunction and removal of excessive free radicals, implying that selective usage of ITCs would be helpful in retarding the progression from HIV infection to AIDS.


Subject(s)
Cytokines/drug effects , Isothiocyanates/pharmacology , Lipid Peroxidation/drug effects , Longevity/drug effects , Murine Acquired Immunodeficiency Syndrome/drug therapy , Murine Acquired Immunodeficiency Syndrome/immunology , Oxidative Stress/drug effects , Animals , Cytokines/immunology , Cytokines/metabolism , Disease Progression , Female , Longevity/immunology , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/metabolism , Sulfoxides , Th1 Cells/drug effects , Th1 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/metabolism , Thiocyanates/pharmacology , Vitamin E/metabolism
16.
J Immunol ; 186(9): 5119-30, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21430226

ABSTRACT

Type I protein kinase A (PKA) is targeted to the TCR-proximal signaling machinery by the A-kinase anchoring protein ezrin and negatively regulates T cell immune function through activation of the C-terminal Src kinase. RI anchoring disruptor (RIAD) is a high-affinity competitor peptide that specifically displaces type I PKA from A-kinase anchoring proteins. In this study, we disrupted type I PKA anchoring in peripheral T cells by expressing a soluble ezrin fragment with RIAD inserted in place of the endogenous A-kinase binding domain under the lck distal promoter in mice. Peripheral T cells from mice expressing the RIAD fusion protein (RIAD-transgenic mice) displayed augmented basal and TCR-activated signaling, enhanced T cell responsiveness assessed as IL-2 secretion, and reduced sensitivity to PGE(2)- and cAMP-mediated inhibition of T cell function. Hyperactivation of the cAMP-type I PKA pathway is involved in the T cell dysfunction of HIV infection, as well as murine AIDS, a disease model induced by infection of C57BL/6 mice with LP-BM5, a mixture of attenuated murine leukemia viruses. LP-BM5-infected RIAD-transgenic mice resist progression of murine AIDS and have improved viral control. This underscores the cAMP-type I PKA pathway in T cells as a putative target for therapeutic intervention in immunodeficiency diseases.


Subject(s)
A Kinase Anchor Proteins/immunology , Murine Acquired Immunodeficiency Syndrome/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , A Kinase Anchor Proteins/metabolism , Animals , Blotting, Western , Cell Separation , Cytoskeletal Proteins/immunology , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Murine Acquired Immunodeficiency Syndrome/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , Transfection
17.
J Pathol ; 221(1): 106-16, 2010 May.
Article in English | MEDLINE | ID: mdl-20217872

ABSTRACT

Anaplastic plasmacytomas (APCTs) from NFS.V(+) congenic mice and pristane-induced plasmacytic PCTs from BALB/c mice were previously shown to be histologically and molecularly distinct subsets of plasma cell neoplasms (PCNs). Here we extended these comparisons, contrasting primary APCTs and PCTs by gene expression profiling in relation to the expression profiles of normal naïve, germinal centre, and memory B cells and plasma cells. We also sequenced immunoglobulin genes from APCT and APCT-derived cell lines and defined surface phenotypes and chromosomal features of the cell lines by flow cytometry and by spectral karyotyping and fluorescence in situ hybridization. The results indicate that APCTs share many features with normal memory cells and the plasma cell-related neoplasms (PLs) of FASL-deficient mice, suggesting that APCTs and PLs are related and that both derive from memory B cells. Published in 2010 by John Wiley & Sons, Ltd.


Subject(s)
B-Lymphocyte Subsets/immunology , Immunologic Memory , Murine Acquired Immunodeficiency Syndrome/immunology , Neoplasms, Plasma Cell/immunology , Plasmacytoma/immunology , Animals , Base Sequence , Cell Survival/physiology , Chromosome Aberrations , Gene Expression Profiling/methods , Immunoglobulin Variable Region/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Murine Acquired Immunodeficiency Syndrome/complications , Murine Acquired Immunodeficiency Syndrome/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Plasma Cell/complications , Neoplasms, Plasma Cell/metabolism , Oligonucleotide Array Sequence Analysis/methods , Plasmacytoma/complications , Plasmacytoma/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction/physiology , Tumor Cells, Cultured
18.
Antiviral Res ; 81(2): 93-102, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18977391

ABSTRACT

In the attempt to eradicate HIV-1 infection, a strategy to eliminate macrophages, one of the most important cellular reservoirs in sustaining virus replication during HAART, could be of great benefit in the suppression of viral rebound. Aware of the ability of clodronate to cause macrophage depletion, the effect of the administration of clodronate encapsulated in erythrocytes on disease progression and on viral rebound was evaluated in a murine model of AIDS (MAIDS). One group of LP-BM5 retroviral complex-infected C57BL/6 mice received oral administrations of azidothymidine and dideoxyinosine daily for 12 weeks; two other groups received in addition, either clodronate-loaded erythrocytes or free clodronate at 7-10 day intervals. At the end of the treatment, the three groups maintained parameters characterizing disease progression similar to those of uninfected mice and showed a significantly lower level of BM5d DNA than infected mice in all organs and cells tested. To assess the viral rebound, some animals were left for an additional 4 month period without any treatment. After this time, the BM5d DNA content in blood leukocytes increased in all groups, but the group having received clodronate-loaded erythrocytes, in addition to transcriptase inhibitors, showed a significant delay in viral rebound.


Subject(s)
DNA, Viral/blood , Macrophages/immunology , Murine Acquired Immunodeficiency Syndrome/immunology , Viral Load , Animals , Anti-HIV Agents/administration & dosage , Clodronic Acid/administration & dosage , Didanosine/administration & dosage , Female , Immunologic Factors/administration & dosage , Leukocyte Reduction Procedures , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/drug therapy , Zidovudine/administration & dosage
19.
Immunogenetics ; 60(9): 485-94, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18612634

ABSTRACT

Murine acquired immunodeficiency syndrome (MAIDS) is caused by exposure to murine leukemia virus and serves as a model to study human AIDS. In MAIDS-susceptible C57BL/6 mice, virus exposure leads to progressive immune deficiency, while resistant strains such as BALB/c recover from infection and develop protective immunity. The goal of this study was to identify early gene expression patterns that may be important in establishing this strain-specific differential response. Total RNA was isolated from spleens and pooled lymph nodes of both mouse strains at 3 and 7 days post virus infection. The complementary DNA generated from this RNA was hybridized to mouse oligonucleotide DNA microarrays using a strategy that controlled for inherent variability and highlighted only virus-induced changes. Fluorescent intensities were normalized and analyzed for statistically significant differential expression between strains across both time points and lymphoid organs. The majority of the resistance-associated genes was identified at day 3 post-infection and demonstrated the highest fold differences between strains, while more susceptibility-associated sequences were seen at 7 days post-infection. Among the most highly differentially expressed sequences seen at the earlier time point were genes related to protein metabolism, especially serine proteases. Differential patterns of chemokine-related genes were observed at the later time point. The overall pattern of expression suggests strain-specific differences in proteases and chemokines within secondary lymphoid organs shortly after infection influence the likelihood of disease progression.


Subject(s)
Leukemia Virus, Murine/physiology , Lymphatic System/metabolism , Murine Acquired Immunodeficiency Syndrome/metabolism , Animals , Gene Expression Profiling , Leukemia Virus, Murine/immunology , Lymph Nodes/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/immunology , Murine Acquired Immunodeficiency Syndrome/virology , Oligonucleotide Array Sequence Analysis , Species Specificity , Spleen/metabolism , Time Factors , Virus Replication
20.
Virology ; 360(1): 58-71, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17113120

ABSTRACT

LP-BM5, a retroviral isolate, induces a disease featuring an acquired immunodeficiency syndrome termed murine AIDS (MAIDS). Many of the features of the LP-BM5-initiated disease are shared with HIV/AIDS. Our lab has shown that the interaction of B and CD4 T cells that is central to MAIDS pathogenesis requires ligation of CD40 on B cells by CD154 on CD4 T cells. Despite this strict requirement for CD154 expression, whether CD4 T cell receptor (TCR) occupancy is essential for the induction of MAIDS is unknown. To block TCR engagement, Tg mouse strains with monoclonal TCR of irrelevant peptide/MHC specificities, all on MAIDS-susceptible genetic backgrounds, were tested: the study of a panel of TCR Tg CD4 T cells controlled for the possibility of serendipitous crossreactive recognition of virus-associated or induced-self peptide, or superantigen, MHC complexes by a given TCR. The results argue that TCR engagement is not necessary for the induction of MAIDS.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD40 Ligand/metabolism , Murine Acquired Immunodeficiency Syndrome/immunology , Retroviridae/pathogenicity , Animals , Gene Deletion , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/deficiency , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, HIV , Virulence/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...