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










Publication year range
2.
Sci Rep ; 11(1): 22901, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824340

ABSTRACT

Macrophages serve as viral reservoirs due to their resistance to apoptosis and HIV-cytopathic effects. We have previously shown that inhibitor of apoptosis proteins (IAPs) confer resistance to HIV-Vpr-induced apoptosis in normal macrophages. Herein, we show that second mitochondrial activator of caspases (SMAC) mimetics (SM) induce apoptosis of monocyte-derived macrophages (MDMs) infected in vitro with a R5-tropic laboratory strain expressing heat stable antigen, chronically infected U1 cells, and ex-vivo derived MDMs from HIV-infected individuals. To understand the mechanism governing SM-induced cell death, we show that SM-induced cell death of primary HIV-infected macrophages was independent of the acquisition of M1 phenotype following HIV infection of macrophages. Instead, SM-induced cell death was found to be mediated by IAPs as downregulation of IAPs by siRNAs induced cell death of HIV-infected macrophages. Moreover, HIV infection caused receptor interacting protein kinase-1 (RIPK1) degradation which in concert with IAP1/2 downregulation following SM treatment may result in apoptosis of macrophages. Altogether, our results show that SM selectively induce apoptosis in primary human macrophages infected in vitro with HIV possibly through RIPK1. Moreover, modulation of the IAP pathways may be a potential strategy for selective killing of HIV-infected macrophages in vivo.


Subject(s)
Anti-HIV Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , HIV Infections/drug therapy , HIV-1/pathogenicity , Macrophages/drug effects , Molecular Mimicry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Apoptosis Regulatory Proteins/genetics , Baculoviral IAP Repeat-Containing 3 Protein/genetics , Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Cytopathogenic Effect, Viral , HIV Infections/enzymology , HIV Infections/pathology , HIV Infections/virology , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Macrophages/enzymology , Macrophages/pathology , Macrophages/virology , Phenotype , U937 Cells , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
3.
J Immunol ; 207(9): 2359-2373, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34561230

ABSTRACT

Inflammatory macrophages have been implicated in many diseases, including rheumatoid arthritis and inflammatory bowel disease. Therefore, targeting macrophage function and activation may represent a potential strategy to treat macrophage-associated diseases. We have previously shown that IFN-γ-induced differentiation of human M0 macrophages toward proinflammatory M1 state rendered them highly susceptible to the cytocidal effects of second mitochondria-derived activator of caspases mimetics (SMs), antagonist of the inhibitors of apoptosis proteins (IAPs), whereas M0 and anti-inflammatory M2c macrophages were resistant. In this study, we investigated the mechanism governing SM-induced cell death during differentiation into M1 macrophages and in polarized M1 macrophages. IFN-γ stimulation conferred on M0 macrophages the sensitivity to SM-induced cell death through the Jak/STAT, IFN regulatory factor-1, and mammalian target of rapamycin complex-1 (mTORC-1)/ribosomal protein S6 kinase pathways. Interestingly, mTORC-1 regulated SM-induced cell death independent of M1 differentiation. In contrast, SM-induced cell death in polarized M1 macrophages is regulated by the mTORC-2 pathway. Moreover, SM-induced cell death is regulated by cellular IAP (cIAP)-2, receptor-interacting protein kinase (RIPK)-1, and RIPK-3 degradation through mTORC activation during differentiation into M1 macrophages and in polarized M1 macrophages. In contrast to cancer cell lines, SM-induced cell death in M1 macrophages is independent of endogenously produced TNF-α, as well as the NF-κB pathway. Collectively, selective induction of cell death in human M1 macrophages by SMs may be mediated by cIAP-2, RIPK-1, and RIPK-3 degradation through mTORC activation. Moreover, blocking cIAP-1/2, mTORC, or IFN regulatory factor-1 may represent a promising therapeutic strategy to control M1-associated diseases.


Subject(s)
Arthritis, Rheumatoid/immunology , Biomimetics/methods , Inflammatory Bowel Diseases/immunology , Macrophages/immunology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Apoptosis Regulatory Proteins/genetics , Cell Death , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Interferon Regulatory Factor-1/metabolism , Mitochondrial Proteins/genetics , NF-kappa B/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/metabolism
4.
J Immunol ; 207(9): 2310-2324, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34551966

ABSTRACT

IFN-γ, a proinflammatory cytokine produced primarily by T cells and NK cells, activates macrophages and engages mechanisms to control pathogens. Although there is evidence of IFN-γ production by murine macrophages, IFN-γ production by normal human macrophages and their subsets remains unknown. Herein, we show that human M1 macrophages generated by IFN-γ and IL-12- and IL-18-stimulated monocyte-derived macrophages (M0) produce significant levels of IFN-γ. Further stimulation of IL-12/IL-18-primed macrophages or M1 macrophages with agonists for TLR-2, TLR-3, or TLR-4 significantly enhanced IFN-γ production in contrast to the similarly stimulated M0, M2a, M2b, and M2c macrophages. Similarly, M1 macrophages generated from COVID-19-infected patients' macrophages produced IFN-γ that was enhanced following LPS stimulation. The inhibition of M1 differentiation by Jak inhibitors reversed LPS-induced IFN-γ production, suggesting that differentiation with IFN-γ plays a key role in IFN-γ induction. We subsequently investigated the signaling pathway(s) responsible for TLR-4-induced IFN-γ production in M1 macrophages. Our results show that TLR-4-induced IFN-γ production is regulated by the ribosomal protein S6 kinase (p70S6K) through the activation of PI3K, the mammalian target of rapamycin complex 1/2 (mTORC1/2), and the JNK MAPK pathways. These results suggest that M1-derived IFN-γ may play a key role in inflammation that may be augmented following bacterial/viral infections. Moreover, blocking the mTORC1/2, PI3K, and JNK MAPKs in macrophages may be of potential translational significance in preventing macrophage-mediated inflammatory diseases.


Subject(s)
Interferon-gamma/biosynthesis , Macrophages/drug effects , Poly I-C/pharmacology , COVID-19/immunology , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/immunology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/immunology , Macrophages/immunology , Phosphatidylinositol 3-Kinases/immunology , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/immunology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/immunology , Toll-Like Receptor 4/agonists
5.
BMC Infect Dis ; 21(1): 655, 2021 Jul 07.
Article in English | MEDLINE | ID: mdl-34233649

ABSTRACT

BACKGROUND: Macrophages, besides resting latently infected CD4+ T cells, constitute the predominant stable, major non-T cell HIV reservoirs. Therefore, it is essential to eliminate both latently infected CD4+ T cells and tissue macrophages to completely eradicate HIV in patients. Until now, most of the research focus is directed towards eliminating latently infected CD4+ T cells. However, few approaches have been directed at killing of HIV-infected macrophages either in vitro or in vivo. HIV infection dysregulates the expression of many host genes essential for the survival of infected cells. We postulated that exploiting this alteration may yield novel targets for the selective killing of infected macrophages. METHODS: We applied a pooled shRNA-based genome-wide approach by employing a lentivirus-based library of shRNAs to screen novel gene targets whose inhibition should selectively induce apoptosis in HIV-infected macrophages. Primary human MDMs were infected with HIV-eGFP and HIV-HSA viruses. Infected MDMs were transfected with siRNAs specific for the promising genes followed by analysis of apoptosis by flow cytometry using labelled Annexin-V in HIV-infected, HIV-exposed but uninfected bystander MDMs and uninfected MDMs. The results were analyzed using student's t-test from at least four independent experiments. RESULTS: We validated 28 top hits in two independent HIV infection models. This culminated in the identification of four target genes, Cox7a2, Znf484, Cstf2t, and Cdk2, whose loss-of-function induced apoptosis preferentially in HIV-infected macrophages. Silencing these single genes killed significantly higher number of HIV-HSA-infected MDMs compared to the HIV-HSA-exposed, uninfected bystander macrophages, indicating the specificity in the killing of HIV-infected macrophages. The mechanism governing Cox7a2-mediated apoptosis of HIV-infected macrophages revealed that targeting respiratory chain complex II and IV genes also selectively induced apoptosis of HIV-infected macrophages possibly through enhanced ROS production. CONCLUSIONS: We have identified above-mentioned novel genes and specifically the respiratory chain complex II and IV genes whose silencing may cause selective elimination of HIV-infected macrophages and eventually the HIV-macrophage reservoirs. The results highlight the potential of the identified genes as targets for eliminating HIV-infected macrophages in physiological environment as part of an HIV cure strategy.


Subject(s)
Apoptosis/genetics , Green Fluorescent Proteins , HIV Infections , Macrophages , RNA, Small Interfering , CD4-Positive T-Lymphocytes/virology , Genome-Wide Association Study , HIV Infections/genetics , HIV Infections/virology , HIV-1/physiology , Humans , T-Lymphocytes
6.
J Immunol ; 200(5): 1593-1606, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29358273

ABSTRACT

The inhibitors of apoptosis (IAP) proteins, initially described in the context of apoptosis regulation as promoting cell survival, have recently emerged as key regulators of innate immune signaling. As a result, downregulation of IAP via Smac mimetics (SMM) has both survival and immunoregulatory effects. IAPs modulate cytokine production in murine models either as a single agent or in response to LPS. However, the role of SMM and the involvement of IAPs in primary human cells and in particular macrophages with respect to cytokine production and innate immune responses remain largely unknown. IL-27, a member of the IL-12 cytokine family produced by APCs such as macrophages, has broad immunoregulatory properties in both innate and adaptive immune responses. Herein, we show that cellular IAPs (cIAPs) positively regulate LPS-induced IL-27 production in both primary human monocytes and macrophages. Investigations for the signaling mechanism of cIAPs involvement in IL-27 production in human macrophages revealed that LPS-induced IL-27 production is regulated by a novel signaling complex comprising cIAP1/2, TNFR-associated factor 2 (TRAF2), SHP-1, Src, and MyD88 leading to p38, c-Jun N-terminal kinases (JNK) and Akt activation and NF-κB signaling. In cancer cells, SMM induce the production of cytokines by activating the noncanonical alternate NF-κB pathway. However, in human macrophages, SMM do not induce the production of TNF-α and other cytokines while inhibiting LPS-induced IL-27 production by inhibiting the classical NF-κB pathway. These signaling pathways may constitute novel therapeutic avenues for immune modulation of IL-27 and provide insight into the modulatory immune effects of SMM.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Interleukins/immunology , Lipopolysaccharides/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Adaptive Immunity/immunology , Baculoviral IAP Repeat-Containing 3 Protein/immunology , Cells, Cultured , Gene Expression Regulation/immunology , Humans , Immunity, Innate/immunology , Inhibitor of Apoptosis Proteins/immunology , Macrophages/immunology , Monocytes/immunology , Myeloid Differentiation Factor 88/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Proto-Oncogene Proteins pp60(c-src)/immunology , TNF Receptor-Associated Factor 2/immunology , Tumor Necrosis Factor-alpha/immunology
7.
J Leukoc Biol ; 102(3): 925-939, 2017 09.
Article in English | MEDLINE | ID: mdl-28698313

ABSTRACT

Monocyte-derived Mϕs (MDMs) from HIV-infected patients and MDM infected in vitro with HIV exhibit a reduced ability to secrete various cytokines, including IL-12. Recently, IL-27, an IL-12 family cytokine, was shown to inhibit HIV replication in Mϕ. Whether HIV infection or HIV accessory protein(s) impact IL-27 production in Mϕs remains unknown. Herein, we show that in vitro HIV infection, as well as intracellular HIV-Tat (Tat) and Tat peptides, inhibit LPS-induced IL-27 production in human MDMs, suggesting impairment of the TLR4 signaling pathway. To understand the signaling pathways governing HIV or Tat-mediated inhibition of LPS-induced IL-27 production, we first demonstrated that p38 MAPK, PI3K, Src-homology region 2 domain-containing tyrosine phosphatase 1 (SHP-1), and Src kinases regulate LPS-induced IL-27 production in MDMs. Tat caused down-regulation of TNFR-associated factor (TRAF)-6 and inhibitor of apoptosis 1 (cIAP-1) and subsequently decreased phosphorylation of downstream PI3K and p38 MAPKs, which were implicated in LPS-induced IL-27 production. Whereas SHP-1 and Src kinases regulated LPS-induced IL-27 production, Tat did not inhibit these kinases, suggesting that they were not involved in Tat-mediated inhibition of LPS-induced IL-27 production. In contrast to Tat, in vitro HIV infection of MDM inhibited LPS-induced IL-27 production via inhibition of p38 MAPK activation. Overall, HIV and Tat inhibit LPS-induced IL-27 production in human macrophages via distinct mechanisms: Tat through the inhibition of cIAP-1-TRAF-6 and subsequent inhibition of PI3K and p38 MAPKs, whereas HIV through the inhibition of p38 MAPK activation.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , Interleukins/immunology , Lipopolysaccharides/pharmacology , Macrophages/immunology , tat Gene Products, Human Immunodeficiency Virus/immunology , Humans , Inhibitor of Apoptosis Proteins/immunology , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Phosphatidylinositol 3-Kinases/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , TNF Receptor-Associated Factor 6/immunology , p38 Mitogen-Activated Protein Kinases/immunology
8.
J Interferon Cytokine Res ; 33(8): 434-45, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23679818

ABSTRACT

Interferon (IFN)-γ is a potent stimulator of the IL-12 family Th1 cytokines, including IL-12/23p40 and IL-23, responsible for coordinating the innate and adaptive immune responses. Our results show that IFN-γ induced the production of IL-12/23p40 and IL-23p19 mRNA as well as IL-12p40 and IL-23 proteins in primary human monocytes isolated by positive selection through anti-CD14 microbeads. These results were confirmed by IFN-γ stimulation of CD14-activated monocytes resulting in IL-12/23p40 and IL-23 production. We investigated the signaling pathways governing the regulation of IL-23 and its subunits IL-23p40 and IL-23p19 following IFN-γ stimulation. We observed a differential regulation of IL-23p19, IL-12/23p40, and IL-23 following IFN-γ stimulation. IFN-γ-induced IL-23 and IL-12/23p40 expression was positively regulated by the p38 mitogen-activated protein kinases (MAPKs), independent of the Janus kinase (Jak)/signal transducers and activators of transcription (STAT) signaling. In contrast, IL-12 and IL-23 were negatively regulated by the Jak/STAT, phosphatidylinositol 3-kinase (PI3K), and the c-Jun-N-terminal kinase (JNK) MAPKs in IFN-γ-stimulated monocytes. Overall, our results suggest for the first time a differential positive regulation of IL-12p40 and IL-23 by p38 MAPKs independent of the Jak/STAT pathways and negative regulation by the Jak/STAT, JNK, and PI3K pathways in CD14-activated primary human monocytes stimulated with IFN-γ.


Subject(s)
Interferon-gamma/pharmacology , Interleukin-12 Subunit p40/metabolism , Interleukin-23/metabolism , Monocytes/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-12 Subunit p40/genetics , Interleukin-23/genetics , Interleukin-23 Subunit p19/genetics , Interleukin-23 Subunit p19/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Janus Kinases/metabolism , Lipopolysaccharide Receptors/immunology , Lipopolysaccharide Receptors/metabolism , Monocytes/immunology , Monocytes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Interferon/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Interferon gamma Receptor
9.
Biores Open Access ; 2(2): 128-37, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23593565

ABSTRACT

Trappin-2/elafin is a novel innate immune factor that belongs to the serine protease inhibitor family and has known antibacterial, antifungal, and antiviral properties. In this study, we further investigated the anti-HIV activity of elafin using different cellular models and both X4- and R5-HIV-1 laboratory strains. We compared the antiviral activity of human recombinant elafin (rElafin) with three well-known antiretroviral drugs, AZT, tenofovir, and enfuvirtide. We have found that when the virus is pre-incubated with rElafin prior to the infection of the cells, HIV-1 replication is significantly inhibited. In target T cells and human peripheral blood mononuclear cells, maximal inhibition was achieved using submicromolar concentrations, and rElafin was found to be as potent as enfuvirtide, showing its potential for therapeutic application. We also show data on the mechanism of the antiviral activity of rElafin. We have demonstrated that rElafin neither binds to CD4, CXCR4, or CCR5 host cell receptors, nor to the viral glycoproteins gp120 and gp41. Furthermore, in our cell-to-cell fusion assays, in contrast to enfuvirtide, rElafin failed to block cell fusion. Altogether our results indicate that rElafin interferes with HIV replication at the early steps of its cycle but with a different mechanism of action than enfuvirtide. This study provides the first experimental evidence that elafin inhibits HIV replication in its natural target cells; therefore, elafin might have potential for its development as a new anti-HIV drug or microbicide.

10.
Expert Opin Biol Ther ; 13(7): 973-85, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23289797

ABSTRACT

OBJECTIVE: Cationic lipidic formulations have been successfully used to deliver small interfering RNA (siRNA) into cells but they show limitations for in vivo application due to their cytotoxicity and instability in the presence of serum. To overcome these limitations, the authors developed an anionic lipid-based carrier named Neutraplex (Nx). Here, they wanted to investigate the influence of the lipoplex (Lx) surface charge on cytotoxicity, delivery and silencing activity of siRNAs. METHODS: The efficiency of three Nx formulations (cationic, close to neutrality and anionic) to deliver anti-CXCR4 siRNAs in MAGI cells was investigated and compared with the cationic commercial transfection reagent Lipofectamine RNAiMAX. Cellular uptake and intracellular localization of a fluorescent siRNA was monitored in live cells using fluorescence microscopy and silencing activity was measured by flow cytometry and RT-PCR analysis. RESULTS: The authors found that the Lx surface charge influenced cellular uptake and silencing activity of siRNA in cell cultures. Although cationic Lx formulations were the most efficient carriers to deliver active silencing siRNAs, negatively charged lipoplexes were taken up by cells, delivered active siRNAs and presented low cytotoxicity. CONCLUSIONS: Altogether, the findings support further investigation for in vivo delivery of therapeutic siRNAs using Nx. Furthermore, this study indicates that anionic delivery systems may have potential for in vivo RNAi therapeutics.


Subject(s)
Cell Proliferation , Drug Carriers/chemistry , Drug Delivery Systems , HIV-1/genetics , Lipids/chemistry , RNA, Small Interfering/administration & dosage , Receptors, CXCR4/antagonists & inhibitors , Blotting, Western , Flow Cytometry , Fluorescent Antibody Technique , Humans , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Surface Properties
12.
Viral Immunol ; 23(1): 17-28, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20121399

ABSTRACT

Studies from our laboratory and those of others have implicated lipopolysaccharide (LPS)-induced MAPK signaling as an important pathway in the regulation of cytokine expression. In this article, the regulation of IL-12 expression in two different human myeloid cell populations was evaluated. In primary monocytes, the inhibition of p38 enhanced IL-12 production, whereas it downregulated IL-12 production in THP-1 cells. The role of MAPK signaling in transcription factor binding to the IL-12p40 promoter was subsequently determined. In primary monocytes, ERK and p38 inhibition increased binding of AP-1 and Sp1, respectively, to the IL-12p40 promoter, while JNK inhibition increased NF-kappaB, AP-1, and Sp1 binding. In THP-1 cells, p38, ERK, and JNK inhibition increased NF-kappaB and Sp1 binding to the IL-12p40 promoter, while inhibiting AP-1 binding. In monocytes, mutations in the NF-kappaB, AP-1, Sp1, or Ets-2 binding sites resulted in complete inhibition of LPS-stimulated IL-12p40 promoter activity using a luciferase-based assay. In contrast, promoter activity was abrogated in THP-1 cells only when the Sp1 or Ets-2 binding sites were mutated. Transcription factor binding to the IL-12p40 promoter following in-vitro HIV infection demonstrated several differences between monocytes and THP-1 cells. Infection with HIV produced an increase in NF-kappaB, AP-1, and Sp1 binding in primary monocytes. In contrast, binding of Ets-2 was dramatically impaired following HIV infection of monocytes, but was unaffected in THP-1 cells. These data clearly show that although LPS induces IL-12p40 expression in primary monocytes and THP-1 cells, the signaling pathways involved and the effect of HIV infection differ and can have disparate effects in these two cell types.


Subject(s)
HIV Infections/immunology , Interleukin-12 Subunit p40/biosynthesis , Lipopolysaccharides/immunology , MAP Kinase Signaling System , Myeloid Cells/immunology , Signal Transduction , Cell Line , Cells, Cultured , DNA/metabolism , Humans , NF-kappa B/metabolism , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Protein c-ets-2/metabolism , Sp1 Transcription Factor/metabolism , Transcription Factor AP-1/metabolism
13.
J Biol Chem ; 284(12): 7578-87, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19019824

ABSTRACT

Impaired cellular immunity caused by decreased production of Th1-type cytokines, including interleukin-12 (IL-12) is a major feature of HIV-1-associated immunodeficiency and acquired immunodeficiency syndrome. IL-12p40, an inducible subunit shared between IL-12 and IL-23, plays a critical role in the development of cellular immunity, and its production is significantly decreased during HIV infection. The mechanism by which HIV induces loss of IL-12p40 production remains poorly understood. We have previously shown that lipopolysaccharide (LPS)-induced IL-12p40 production in monocytic cells is regulated by NFkappaB and AP-1 transcription factors through the activation of two distinct upstream signaling pathways, namely the c-Jun-N-terminal kinase (JNK) and the calmodulin-dependent protein kinase-II-activated pathways. Herein, we show that intracellular nef expressed through transduction of primary monocytes and promonocytic THP-1 cells with retroviral-mediated nef gene inhibited LPS-induced IL-12p40 transcription by inhibiting the JNK mitogen-activated protein kinases without affecting the calmodulin-dependent protein kinase-II-activated pathway. In addition, nef inhibited JNK-activated NFkappaB without affecting the AP-1 activity. Overall, our results suggest for the first time that intracellular nef inhibited LPS-activated JNK, which may cause inhibition of IL-12p40 expression in human monocytic cells by selectively inhibiting NFkappaB activity.


Subject(s)
Acquired Immunodeficiency Syndrome/metabolism , Gene Expression Regulation , HIV-1/metabolism , Interleukin-12 Subunit p40/biosynthesis , Lipopolysaccharides/pharmacology , MAP Kinase Kinase 4/metabolism , Monocytes/metabolism , NF-kappa B/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/immunology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , HIV-1/immunology , Humans , Immunity, Cellular/drug effects , Interleukin-23/immunology , Interleukin-23/metabolism , MAP Kinase Kinase 4/immunology , Mice , Monocytes/immunology , NF-kappa B/immunology , NIH 3T3 Cells , Transcription Factor AP-1/immunology , Transcription Factor AP-1/metabolism , Transcription, Genetic/drug effects , nef Gene Products, Human Immunodeficiency Virus/immunology
14.
J Biol Chem ; 281(42): 31647-58, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-16920714

ABSTRACT

Human immunodeficiency virus (HIV)-Tat plays an important role in virus replication and in various aspects of host immune responses, including dysregulation of cytokine production. IL-10, an anti-inflammatory cytokine, is up-regulated during the course of HIV infection representing an important pathway by which HIV may induce immunodeficiency. Here we show that extracellular as well as intracellular Tat induced IL-10 expression in normal human monocytes and promonocytic THP-1 cells. The signaling pathways involved in the regulation of IL-10 production by endogenous Tat remain unknown. To understand the molecular mechanism underlying intracellular Tat-induced IL-10 transcription, we employed a retroviral expression system to investigate the role of MAPKs and the transcription factor(s) involved. Our results suggest that an inhibitor specific for the ERK1/2, PD98059, selectively blocked intracellular Tat-induced IL-10 expression in THP-1 cells. Furthermore, intracellular Tat activated the CREB-1 transcription factor through Ser(133) phosphorylation that was regulated by ERK MAPK as determined by IL-10 promoter analysis and gel shift assays. Overall, our results suggest that intracellular HIV-Tat induces IL-10 transcription by ERK MAPK-dependent CREB-1 transcription factor activation through Ser(133) phosphorylation.


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , Gene Products, tat/chemistry , Interleukin-10/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/metabolism , Serine/chemistry , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Primers/chemistry , Enzyme Inhibitors/pharmacology , Humans , Interleukin-10/metabolism , Luciferases/metabolism , MAP Kinase Signaling System , Monocytes/virology , Phosphorylation , Retroviridae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...