1
|
Lee MYH, Upadhyay AA, Walum H, Chan CN, Dawoud RA, Grech C, Harper JL, Karunakaran KA, Nelson SA, Mahar EA, Goss KL, Carnathan DG, Cervasi B, Gill K, Tharp GK, Wonderlich ER, Velu V, Barratt-Boyes SM, Paiardini M, Silvestri G, Estes JD, Bosinger SE. Tissue-specific transcriptional profiling of plasmacytoid dendritic cells reveals a hyperactivated state in chronic SIV infection. PLoS Pathog 2021; 17:e1009674. [PMID: 34181694 PMCID: PMC8270445 DOI: 10.1371/journal.ppat.1009674] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 07/09/2021] [Accepted: 05/28/2021] [Indexed: 12/15/2022] Open
Abstract
HIV associated immune activation (IA) is associated with increased morbidity in people living with HIV (PLWH) on antiretroviral therapy, and remains a barrier for strategies aimed at reducing the HIV reservoir. The underlying mechanisms of IA have not been definitively elucidated, however, persistent production of Type I IFNs and expression of ISGs is considered to be one of the primary factors. Plasmacytoid DCs (pDCs) are a major producer of Type I IFN during viral infections, and are highly immunomodulatory in acute HIV and SIV infection, however their role in chronic HIV/SIV infection has not been firmly established. Here, we performed a detailed transcriptomic characterization of pDCs in chronic SIV infection in rhesus macaques, and in sooty mangabeys, a natural host non-human primate (NHP) species that undergoes non-pathogenic SIV infection. We also investigated the immunostimulatory capacity of lymph node homing pDCs in chronic SIV infection by contrasting gene expression of pDCs isolated from lymph nodes with those from blood. We observed that pDCs in LNs, but not blood, produced high levels of IFNα transcripts, and upregulated gene expression programs consistent with T cell activation and exhaustion. We apply a novel strategy to catalogue uncharacterized surface molecules on pDCs, and identified the lymphoid exhaustion markers TIGIT and LAIR1 as highly expressed in SIV infection. pDCs from SIV-infected sooty mangabeys lacked the activation profile of ISG signatures observed in infected macaques. These data demonstrate that pDCs are a primary producer of Type I IFN in chronic SIV infection. Further, this study demonstrated that pDCs trafficking to LNs persist in a highly activated state well into chronic infection. Collectively, these data identify pDCs as a highly immunomodulatory cell population in chronic SIV infection, and a putative therapeutic target to reduce immune activation.
Collapse
Affiliation(s)
- Michelle Y.-H. Lee
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Amit A. Upadhyay
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Hasse Walum
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Chi N. Chan
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Reem A. Dawoud
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Christine Grech
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Justin L. Harper
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Kirti A. Karunakaran
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Sydney A. Nelson
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Ernestine A. Mahar
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Kyndal L. Goss
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Diane G. Carnathan
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Barbara Cervasi
- Flow Cytometry Core, Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
| | - Kiran Gill
- Flow Cytometry Core, Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
| | - Gregory K. Tharp
- Yerkes NHP Genomics Core Laboratory, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | | | - Vijayakumar Velu
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Simon M. Barratt-Boyes
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mirko Paiardini
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Guido Silvestri
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Jacob D. Estes
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Steven E. Bosinger
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
- Yerkes NHP Genomics Core Laboratory, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
2
|
Sun S, Yang Q, Sheng Y, Fu Y, Sun C, Deng C. Investigational drugs with dual activity against HBV and HIV (Review). Exp Ther Med 2020; 21:35. [PMID: 33262821 PMCID: PMC7690342 DOI: 10.3892/etm.2020.9467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/14/2020] [Indexed: 12/17/2022] Open
Abstract
Chronic hepatitis B (CHB) and acquired immunodeficiency syndrome (AIDS) are global public health problems that pose a significant health burden. Human immunodeficiency virus (HIV) and hepatitis B virus (HBV) coinfection is common, as these viruses have similar transmission routes, such as blood transmission, sexual transmission and mother-to-child transmission. Coinfection frequently leads to accelerated disease progression. For individuals coinfected with HIV/HBV, combination antiretroviral therapy containing dual anti-HBV drugs is recommended. Certain studies have also indicated the benefits of antiretroviral drugs with anti-HBV activity in patients with coinfection. A total of four Food and Drug Administration-approved HIV drugs also have anti-HBV activity; namely, emtricitabine, lamivudine, tenofovir disoproxil fumarate and tenofovir alafenamide, which are all nucleoside reverse transcriptase inhibitors. However, various issues, including drug resistance and side effects, limit their application. Therefore, it is necessary to develop more drugs with dual activity against HBV and HIV. The present review outlines the mechanisms, safety and efficacy of certain drugs that have been investigated for this purpose.
Collapse
Affiliation(s)
- Shiyu Sun
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Department of Tuberculosis, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Department of Infection and Immunity Laboratory, Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Qing Yang
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Department of Tuberculosis, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Department of Infection and Immunity Laboratory, Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yunjian Sheng
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Department of Tuberculosis, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Department of Infection and Immunity Laboratory, Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yi Fu
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Department of Gastrointestinal Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Changfeng Sun
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Department of Tuberculosis, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Department of Infection and Immunity Laboratory, Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Cunliang Deng
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Department of Tuberculosis, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Department of Infection and Immunity Laboratory, Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
3
|
Gomez-Lucia E, Collado VM, Miró G, Martín S, Benítez L, Doménech A. Follow-Up of Viral Parameters in FeLV- or FIV-Naturally Infected Cats Treated Orally with Low Doses of Human Interferon Alpha. Viruses 2019; 11:E845. [PMID: 31514435 PMCID: PMC6783854 DOI: 10.3390/v11090845] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/23/2019] [Accepted: 09/08/2019] [Indexed: 01/05/2023] Open
Abstract
Specific treatments for the long-life infections by feline leukemia virus (FeLV) and feline immunodeficiency virus (FIV) are either toxic, expensive or not too effective. Interferon α (IFN-α) is an immunomodulatory molecule which has been shown in vitro to decrease the release of infective particles. The aim of this study was to follow the progress of the clinical score and viral parameters of FeLV- and FIV-naturally infected privately owned cats treated with recombinant human IFN-α (rHuIFN-α, Roferon-A). Twenty-seven FeLV-infected cats (FeLV+) and 31 FIV-infected cats (FIV+) were enrolled in the study. Owners were instructed to orally administer 1 mL/day of 60 IU rHuIFN-α/mL in alternating weeks for four months. Blood samples were taken at the beginning of the study (M0), mid-treatment (M2), end of treatment (M4), and 6-10 months later (M10). Clinical status at these time points improved notably with rHuIFN-α treatment, regardless of the initial severity of the disease, an effect which lasted throughout the study in most animals (15 of the 16 FeLV+ symptomatic cats; 20 of the 22 FIV+ symptomatic cats) improved markedly their clinical situation. In FeLV+ cats plasma antigenemia (p27CA), reverse transcriptase (RT) activity, and proviral load decreased at M2 and M4 but increased again at M10 ("rebound effect"). The level of antigenemia or RT activity was below the detection limits in FIV+ cats, and the effect on proviral load was less marked than in FeLV+ cats. Taken together, these results indicate that rHuIFN-α is a good candidate for treating FeLV+ cats, but the "rebound effect" seen when treatment was discontinued suggests that additional studies should be conducted to clarify its effect on progression of the infection in cats.
Collapse
Affiliation(s)
- Esperanza Gomez-Lucia
- Department of Animal Health, Veterinary Faculty, Complutense University of Madrid, 28040 Madrid, Spain.
| | - Victorio M Collado
- Department of Animal Health, Veterinary Faculty, Complutense University of Madrid, 28040 Madrid, Spain
| | - Guadalupe Miró
- Department of Animal Health, Veterinary Faculty, Complutense University of Madrid, 28040 Madrid, Spain
| | - Sonsoles Martín
- Department of Animal Medicine and Surgery, Veterinary Faculty, Complutense University of Madrid, 28040 Madrid, Spain
| | - Laura Benítez
- Department of Genetics, Physiology and Microbiology, Faculty of Biology, Complutense University of Madrid, José Antonio Novais, 12, 28040 Madrid, Spain
| | - Ana Doménech
- Department of Animal Health, Veterinary Faculty, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
4
|
Papasavvas E, Azzoni L, Kossenkov AV, Dawany N, Morales KH, Fair M, Ross BN, Lynn K, Mackiewicz A, Mounzer K, Tebas P, Jacobson JM, Kostman JR, Showe L, Montaner LJ. NK Response Correlates with HIV Decrease in Pegylated IFN-α2a-Treated Antiretroviral Therapy-Suppressed Subjects. THE JOURNAL OF IMMUNOLOGY 2019; 203:705-717. [PMID: 31253727 DOI: 10.4049/jimmunol.1801511] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 06/03/2019] [Indexed: 01/27/2023]
Abstract
We previously reported that pegylated IFN-α2a (Peg-IFN-α2a) added to antiretroviral therapy (ART)-suppressed, HIV-infected subjects resulted in plasma HIV control and integrated HIV DNA decrease. We now evaluated whether innate NK cell activity or PBMC transcriptional profiles were associated with decreases in HIV measures. Human peripheral blood was analyzed prior to Peg-IFN-α2a administration (ART, baseline), after 5 wk of ART+Peg-IFN-α2a, and after 12 wk of Peg-IFN-α2a monotherapy (primary endpoint). After 5 wk of ART+Peg-IFN-α2a, immune subset frequencies were preserved, and induction of IFN-stimulated genes was noted in all subjects except for a subset in which the lack of IFN-stimulated gene induction was associated with increased expression of microRNAs. Viral control during Peg-IFN-α2a monotherapy was associated with 1) higher levels of NK cell activity and IFN-γ-induced protein 10 (IP-10) on ART (preimmunotherapy) and 2) downmodulation of NK cell KIR2DL1 and KIR2DL2/DL3 expression, transcriptional enrichment of expression of genes associated with NK cells in HIV controller subjects, and higher ex vivo IFN-α-induced NK cytotoxicity after 5 wk of ART+Peg-IFN-α2a. Integrated HIV DNA decline after immunotherapy was also associated with gene expression patterns indicative of cell-mediated activation and NK cytotoxicity. Overall, an increase in innate activity and NK cell cytotoxicity were identified as correlates of Peg-IFN-α2a-mediated HIV control.
Collapse
Affiliation(s)
| | | | | | - Noor Dawany
- The Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Knashawn H Morales
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | | | | | - Kenneth Lynn
- Presbyterian Hospital-University of Pennsylvania Hospital, Philadelphia, PA 19104
| | | | - Karam Mounzer
- Jonathan Lax Immune Disorders Treatment Center, Philadelphia Field Initiating Group for HIV-1 Trials, Philadelphia, PA 19107
| | - Pablo Tebas
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Jeffrey M Jacobson
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140; and
| | - Jay R Kostman
- John Bell Health Center, Philadelphia Field Initiating Group for HIV-1 Trials, Philadelphia, PA 19107
| | | | | |
Collapse
|
5
|
Zhang Y, Ozono S, Yao W, Tobiume M, Yamaoka S, Kishigami S, Fujita H, Tokunaga K. CRISPR-mediated activation of endogenous BST-2/tetherin expression inhibits wild-type HIV-1 production. Sci Rep 2019; 9:3134. [PMID: 30816279 PMCID: PMC6395588 DOI: 10.1038/s41598-019-40003-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 02/07/2019] [Indexed: 12/21/2022] Open
Abstract
The CRISPR technology not only can knock out target genes by using the RNA-guided Cas9 nuclease but also can activate their expression when a nuclease-deficient Cas9 (dCas9) is employed. Using the latter function, we here show the effect of the CRISPR-mediated pinpoint activation of endogenous expression of BST-2 (also known as tetherin), a virus restriction factor with a broad antiviral spectrum. Single-guide RNA (sgRNA) sequences targeting the BST-2 promoter were selected by promoter assays. Potential sgRNAs and dCas9 fused to the VP64 transactivation domain, along with an accessory transcriptional activator complex, were introduced into cells by lentiviral transduction. Increased expression of BST-2 mRNA in transduced cells was confirmed by real-time RT-PCR. Cells in which BST-2 expression was highly enhanced showed the effective inhibition of HIV-1 production and replication even in the presence of the viral antagonist Vpu against BST-2. These findings confirm that the physiological stoichiometry between host restriction factors and viral antagonists may determine the outcome of the battle with viruses.
Collapse
Affiliation(s)
- Yanzhao Zhang
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Seiya Ozono
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi, 400-8510, Japan
| | - Weitong Yao
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
- Department of Molecular Virology, Tokyo Medical and Dental University, Tokyo, 113-8519, Japan
| | - Minoru Tobiume
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Shoji Yamaoka
- Department of Molecular Virology, Tokyo Medical and Dental University, Tokyo, 113-8519, Japan
| | - Satoshi Kishigami
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi, 400-8510, Japan
| | - Hideaki Fujita
- Faculty of Pharmaceutical Sciences, Nagasaki International University, Nagasaki, 859-3298, Japan
| | - Kenzo Tokunaga
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan.
| |
Collapse
|
6
|
Short-Term Pegylated Interferon α2a Treatment Does Not Significantly Reduce the Viral Reservoir of Simian Immunodeficiency Virus-Infected, Antiretroviral Therapy-Treated Rhesus Macaques. J Virol 2018; 92:JVI.00279-18. [PMID: 29720521 DOI: 10.1128/jvi.00279-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/26/2018] [Indexed: 01/05/2023] Open
Abstract
The major obstacle to human immunodeficiency type 1 (HIV-1) eradication is a reservoir of latently infected cells that persists despite long-term antiretroviral therapy (ART) and causes rapid viral rebound if treatment is interrupted. Type I interferons are immunomodulatory cytokines that induce antiviral factors and have been evaluated for the treatment of HIV-infected individuals, resulting in moderate reduction of viremia and inconclusive data about their effect on reservoir size. Here, we assessed the potential of pegylated IFN-α2a (pIFN-α2a) to reduce the viral reservoir in simian immunodeficiency virus (SIV)-infected, ART-treated rhesus macaques (RMs). We found that pIFN-α2a treatment of animals in which virus replication is effectively suppressed with ART is safe and well tolerated, as no major clinical side effects were observed. By monitoring the cellular immune response during this intervention, we established that pIFN-α2a administration is not associated with either CD4+ T cell depletion or increased immune activation. Importantly, we found that interferon-stimulated genes (ISGs) were significantly upregulated in IFN-treated RMs compared to control animals, confirming that pIFN-α2a is bioactive in vivo To evaluate the effect of pIFN-α2a administration on the viral reservoir in CD4+ T cells, we performed cell-associated proviral SIV DNA measurements in multiple tissues and assessed levels of replication-competent virus by a quantitative viral outgrowth assay (QVOA). These analyses failed to reveal any significant difference in reservoir size between IFN-treated and control animals. In summary, our data suggest that short-term type I interferon treatment in combination with suppressive ART is not sufficient to induce a significant reduction of the viral reservoir in SIV-infected RMs.IMPORTANCE The potential of type I interferons to reduce the viral reservoir has been recently studied in clinical trials in HIV-infected humans. However, given the lack of mechanistic data and the potential for safety concerns, a more comprehensive testing of IFN treatment in vivo in SIV-infected RMs is critical to provide rationale for further development of this intervention in humans. Utilizing the SIV/RM model in which virus replication is suppressed with ART, we addressed experimental limitations of previous human studies, in particular the lack of a control group and specimen sampling limited to blood. Here, we show by rigorous testing of blood and lymphoid tissues that virus replication and reservoir size were not significantly affected by pIFN-α2a treatment in SIV-infected, ART-treated RMs. This suggests that intensified and/or prolonged IFN treatment regimens, possibly in combination with other antilatency agents, are necessary to effectively purge the HIV/SIV reservoir under ART.
Collapse
|
7
|
Raftery N, Stevenson NJ. Advances in anti-viral immune defence: revealing the importance of the IFN JAK/STAT pathway. Cell Mol Life Sci 2017; 74:2525-2535. [PMID: 28432378 PMCID: PMC7079803 DOI: 10.1007/s00018-017-2520-2] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/31/2017] [Accepted: 04/04/2017] [Indexed: 02/06/2023]
Abstract
Interferon-alpha (IFN-α) is a potent anti-viral cytokine, critical to the host immune response against viruses. IFN-α is first produced upon viral detection by pathogen recognition receptors. Following its expression, IFN-α embarks upon a complex downstream signalling cascade called the JAK/STAT pathway. This signalling pathway results in the expression of hundreds of effector genes known as interferon stimulated genes (ISGs). These genes are the basis for an elaborate effector mechanism and ultimately, the clearance of viral infection. ISGs mark an elegant mechanism of anti-viral host defence that warrants renewed research focus in our global efforts to treat existing and emerging viruses. By understanding the mechanistic role of individual ISGs we anticipate the discovery of a new "treasure trove" of anti-viral mediators that may pave the way for more effective, targeted and less toxic anti-viral therapies. Therefore, with the aim of highlighting the value of the innate type 1 IFN response in our battle against viral infection, this review outlines both historic and recent advances in understanding the IFN-α JAK/STAT pathway, with a focus on new research discoveries relating to specific ISGs and their potential role in curing existing and future emergent viral infections.
Collapse
Affiliation(s)
- Nicola Raftery
- School of Medicine, Trinity College Dublin, Dublin 2, Ireland
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland
| | - Nigel J Stevenson
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
8
|
Riley JL, Montaner LJ. Cell-Mediated Immunity to Target the Persistent Human Immunodeficiency Virus Reservoir. J Infect Dis 2017; 215:S160-S171. [PMID: 28520969 PMCID: PMC5853458 DOI: 10.1093/infdis/jix002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Effective clearance of virally infected cells requires the sequential activity of innate and adaptive immunity effectors. In human immunodeficiency virus (HIV) infection, naturally induced cell-mediated immune responses rarely eradicate infection. However, optimized immune responses could potentially be leveraged in HIV cure efforts if epitope escape and lack of sustained effector memory responses were to be addressed. Here we review leading HIV cure strategies that harness cell-mediated control against HIV in stably suppressed antiretroviral-treated subjects. We focus on strategies that may maximize target recognition and eradication by the sequential activation of a reconstituted immune system, together with delivery of optimal T-cell responses that can eliminate the reservoir and serve as means to maintain control of HIV spread in the absence of antiretroviral therapy (ART). As evidenced by the evolution of ART, we argue that a combination of immune-based strategies will be a superior path to cell-mediated HIV control and eradication. Available data from several human pilot trials already identify target strategies that may maximize antiviral pressure by joining innate and engineered T cell responses toward testing for sustained HIV remission and/or cure.
Collapse
Affiliation(s)
- James L Riley
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, and
| | - Luis J Montaner
- HIV-1 Immunopathogenesis Laboratory, Wistar Institute, Philadelphia, Pennsylvania
| |
Collapse
|
9
|
Interferon Alpha Subtype-Specific Suppression of HIV-1 Infection In Vivo. J Virol 2016; 90:6001-6013. [PMID: 27099312 DOI: 10.1128/jvi.00451-16] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 04/13/2016] [Indexed: 01/29/2023] Open
Abstract
UNLABELLED Although all 12 subtypes of human interferon alpha (IFN-α) bind the same receptor, recent results have demonstrated that they elicit unique host responses and display distinct efficacies in the control of different viral infections. The IFN-α2 subtype is currently in HIV-1 clinical trials, but it has not consistently reduced viral loads in HIV-1 patients and is not the most effective subtype against HIV-1 in vitro We now demonstrate in humanized mice that, when delivered at the same high clinical dose, the human IFN-α14 subtype has very potent anti-HIV-1 activity whereas IFN-α2 does not. In both postexposure prophylaxis and treatment of acute infections, IFN-α14, but not IFN-α2, significantly suppressed HIV-1 replication and proviral loads. Furthermore, HIV-1-induced immune hyperactivation, which is a prognosticator of disease progression, was reduced by IFN-α14 but not IFN-α2. Whereas ineffective IFN-α2 therapy was associated with CD8(+) T cell activation, successful IFN-α14 therapy was associated with increased intrinsic and innate immunity, including significantly higher induction of tetherin and MX2, increased APOBEC3G signature mutations in HIV-1 proviral DNA, and higher frequencies of TRAIL(+) NK cells. These results identify IFN-α14 as a potent new therapeutic that operates via mechanisms distinct from those of antiretroviral drugs. The ability of IFN-α14 to reduce both viremia and proviral loads in vivo suggests that it has strong potential as a component of a cure strategy for HIV-1 infections. The broad implication of these results is that the antiviral efficacy of each individual IFN-α subtype should be evaluated against the specific virus being treated. IMPORTANCE The naturally occurring antiviral protein IFN-α2 is used to treat hepatitis viruses but has proven rather ineffective against HIV in comparison to triple therapy with the antiretroviral (ARV) drugs. Although ARVs suppress the replication of HIV, they fail to completely clear infections. Since IFN-α acts by different mechanism than ARVs and has been shown to reduce HIV proviral loads, clinical trials are under way to test whether IFN-α2 combined with ARVs might eradicate HIV-1 infections. IFN-α is actually a family of 12 distinct proteins, and each IFN-α subtype has different efficacies toward different viruses. Here, we use mice that contain a human immune system, so they can be infected with HIV. With this model, we demonstrate that while IFN-α2 is only weakly effective against HIV, IFN-α14 is extremely potent. This discovery identifies IFN-α14 as a more powerful IFN-α subtype for use in combination therapy trials aimed toward an HIV cure.
Collapse
|
10
|
O’Brien M, Manches O, Wilen C, Gopal R, Huq R, Wu V, Sunseri N, Bhardwaj N. CD4 Receptor is a Key Determinant of Divergent HIV-1 Sensing by Plasmacytoid Dendritic Cells. PLoS Pathog 2016; 12:e1005553. [PMID: 27082754 PMCID: PMC4833349 DOI: 10.1371/journal.ppat.1005553] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 03/14/2016] [Indexed: 11/26/2022] Open
Abstract
Plasmacytoid dendritic cells (pDC) are innate immune cells that sense viral nucleic acids through endosomal Toll-like receptor (TLR) 7/9 to produce type I interferon (IFN) and to differentiate into potent antigen presenting cells (APC). Engagement of TLR7/9 in early endosomes appears to trigger the IRF7 pathway for IFN production whereas engagement in lysosomes seems to trigger the NF-κB pathway for maturation into APC. We showed previously that HIV-1 (HIV) localizes predominantly to early endosomes, not lysosomes, and mainly stimulate IRF7 rather than NF-κB signaling pathways in pDC. This divergent signaling may contribute to disease progression through production of pro-apoptotic and pro-inflammatory IFN and inadequate maturation of pDCs. We now demonstrate that HIV virions may be re-directed to lysosomes for NF-κB signaling by either pseudotyping HIV with influenza hemagglutinin envelope or modification of CD4 mediated-intracellular trafficking. These data suggest that HIV envelope-CD4 receptor interactions drive pDC activation toward an immature IFN producing phenotype rather than differentiation into a mature dendritic cell phenotype. Plasmacytoid dendritic cells (pDC) are innate immune cells that are specialized to produce type I interferon (IFN) and to activate adaptive immune responses. Although IFN is an anti-viral cytokine, it may contribute more to pathogenesis than to protection during chronic viral infections, including chronic HIV infection. pDC sense HIV to produce abundant IFN but minimal NF- κB–dependent production of TNFα and minimal up-regulation of co-stimulatory molecules, suggesting that HIV promotes pDC to become interferon producing cells (IPC) rather than antigen presenting cells (APC). Here, we use florescent HIV virions pseudotyped with influenza hemagglutinin (HA) envelope and a cell system expressing CD4 molecules with modified intracellular trafficking. We found that HIV virions pseudotyped with HA stimulate pDC to mature, similar to influenza-stimulated pDC, and traffic intracellularly similarly to influenza. We also find that CD4-mediated intracellular trafficking guides HIV trafficking and downstream signaling. Our study presents new and important findings which demonstrate that divergent HIV sensing by pDC to produce IFN, rather than to become mature antigen presenting cells, is mediated specifically by CD4-HIV envelope interactions.
Collapse
Affiliation(s)
- Meagan O’Brien
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- * E-mail: Meagan.O’ (MO); (OM)
| | - Olivier Manches
- Division of Hematology and Oncology, Hess Center for Science and Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- * E-mail: Meagan.O’ (MO); (OM)
| | - Craig Wilen
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ramya Gopal
- Division of Hematology and Oncology, Hess Center for Science and Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Rumana Huq
- Microscopy Shared Resource Facility, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Vernon Wu
- Division of Hematology and Oncology, Hess Center for Science and Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Nicole Sunseri
- Department of Pediatrics, the University of Chicago, Chicago, Illinois, United States of America
| | - Nina Bhardwaj
- Division of Hematology and Oncology, Hess Center for Science and Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
11
|
Bosinger SE, Utay NS. Type I interferon: understanding its role in HIV pathogenesis and therapy. Curr HIV/AIDS Rep 2016; 12:41-53. [PMID: 25662992 DOI: 10.1007/s11904-014-0244-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite over 30 years of research, the contribution of type I interferons (IFN-Is) to both the control of HIV replication and initiation of immunologic damage remains debated. In acute infection, IFN-Is, likely from plasmacytoid dendritic cells (pDCs), activate NK cells and upregulate restriction factors targeting virtually the entire HIV life cycle. In chronic infection, IFN-Is may also contribute to CD4 T cell loss and immune exhaustion. pDCs subsequently infiltrate lymphoid and mucosal tissues, and their circulating populations wane in chronic infection; IFN-I may be produced by other cells. Data from nonhuman primates indicate prompt IFN-I signaling is critical in acute infection. Whereas some studies showed IFN-I administration without combination antiretroviral therapy (cART) is beneficial, others suggest that stimulating or blocking IFN-I signaling in chronic ART-suppressed HIV infection has had positive results. Here, we describe the history of HIV and IFN-I, IFN-I's sources, IFN-I's effects on HIV control and host defense, and recent interventional studies in SIV and HIV infection.
Collapse
Affiliation(s)
- Steven E Bosinger
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory Vaccine Center Bldg. 3028, Atlanta, GA, 30322, USA,
| | | |
Collapse
|
12
|
Sandler NG, Bosinger SE, Estes JD, Zhu RTR, Tharp GK, Boritz E, Levin D, Wijeyesinghe S, Makamdop KN, del Prete GQ, Hill BJ, Timmer JK, Reiss E, Yarden G, Darko S, Contijoch E, Todd JP, Silvestri G, Nason M, Norgren RB, Keele BF, Rao S, Langer JA, Lifson JD, Schreiber G, Douek DC. Type I interferon responses in rhesus macaques prevent SIV infection and slow disease progression. Nature 2014; 511:601-5. [PMID: 25043006 PMCID: PMC4418221 DOI: 10.1038/nature13554] [Citation(s) in RCA: 391] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 06/04/2014] [Indexed: 12/26/2022]
Abstract
Inflammation in HIV infection is predictive of non-AIDS morbidity and death, higher set point plasma virus load and virus acquisition; thus, therapeutic agents are in development to reduce its causes and consequences. However, inflammation may simultaneously confer both detrimental and beneficial effects. This dichotomy is particularly applicable to type I interferons (IFN-I) which, while contributing to innate control of infection, also provide target cells for the virus during acute infection, impair CD4 T-cell recovery, and are associated with disease progression. Here we manipulated IFN-I signalling in rhesus macaques (Macaca mulatta) during simian immunodeficiency virus (SIV) transmission and acute infection with two complementary in vivo interventions. We show that blockade of the IFN-I receptor caused reduced antiviral gene expression, increased SIV reservoir size and accelerated CD4 T-cell depletion with progression to AIDS despite decreased T-cell activation. In contrast, IFN-α2a administration initially upregulated expression of antiviral genes and prevented systemic infection. However, continued IFN-α2a treatment induced IFN-I desensitization and decreased antiviral gene expression, enabling infection with increased SIV reservoir size and accelerated CD4 T-cell loss. Thus, the timing of IFN-induced innate responses in acute SIV infection profoundly affects overall disease course and outweighs the detrimental consequences of increased immune activation. Yet, the clinical consequences of manipulation of IFN signalling are difficult to predict in vivo and therapeutic interventions in human studies should be approached with caution.
Collapse
Affiliation(s)
- Netanya G Sandler
- 1] Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892, USA [2] Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch at Galveston, Galveston, Texas 77555, USA
| | - Steven E Bosinger
- 1] Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, Georgia 30322, USA [2] Non-Human Primate Genomics Core, Yerkes National Primate Research Center, Robert W. Woodruff Health Sciences Center, Emory University, Atlanta, Georgia 30322, USA
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Richard T R Zhu
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Gregory K Tharp
- 1] Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, Georgia 30322, USA [2] Non-Human Primate Genomics Core, Yerkes National Primate Research Center, Robert W. Woodruff Health Sciences Center, Emory University, Atlanta, Georgia 30322, USA
| | - Eli Boritz
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Doron Levin
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Sathi Wijeyesinghe
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Krystelle Nganou Makamdop
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Gregory Q del Prete
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Brenna J Hill
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - J Katherina Timmer
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Emma Reiss
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Ganit Yarden
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Samuel Darko
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Eduardo Contijoch
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - John Paul Todd
- Laboratory of Animal Medicine, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Guido Silvestri
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, Georgia 30322, USA
| | - Martha Nason
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Robert B Norgren
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Srinivas Rao
- Laboratory of Animal Medicine, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jerome A Langer
- Department of Pharmacology, Rutgers - Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Gideon Schreiber
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Daniel C Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
13
|
Streeck H, Rockstroh JK. Challenges in the treatment of HIV and HCV coinfection. Expert Rev Clin Immunol 2014; 2:811-22. [DOI: 10.1586/1744666x.2.5.811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
14
|
Abstract
Since the introduction of highly active antiretroviral therapy (HAART), HIV-related deaths have declined dramatically in the developed world. However, HAART is neither able to eradicate the virus nor are its immunomodulatory effects sufficient to effect complete control of the virus. In addition, the long-term use of HAART is complicated by drug-related toxicities and compliance issues, both of which impact upon the development of viral resistance. The failure of structured treatment interruption strategies in those with chronic HIV-infection combined with the above limitations, has prompted renewed interest in immunotherapy. Cytokines and therapeutic vaccination have been proposed as HAART-adjunctive and HAART-sparing treatments in HIV-infection, and the current and future role of cytokine therapy in this disease will be the subject of this review.
Collapse
Affiliation(s)
- Sarah L Pett
- University of New South Wales, National Centre in HIV Epidemiology and Clinical Research, Level 2, 376, Victoria Street, Darlinghurst, NSW 2010, Australia.
| | | |
Collapse
|
15
|
Chereshnev VA, Bocharov G, Bazhan S, Bachmetyev B, Gainova I, Likhoshvai V, Argilaguet JM, Martinez JP, Rump JA, Mothe B, Brander C, Meyerhans A. Pathogenesis and treatment of HIV infection: the cellular, the immune system and the neuroendocrine systems perspective. Int Rev Immunol 2013; 32:282-306. [PMID: 23617796 DOI: 10.3109/08830185.2013.779375] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Infections with HIV represent a great challenge for the development of strategies for an effective cure. The spectrum of diseases associated with HIV ranges from opportunistic infections and cancers to systemic physiological disorders like encephalopathy and neurocognitive impairment. A major progress in controlling HIV infection has been achieved by highly active antiretroviral therapy (HAART). However, HAART does neither eliminate the virus reservoirs in form of latently infected cells nor does it completely reconstitute immune reactivity and physiological status. Furthermore, the failure of the STEP vaccine trial and the only marginal efficacies of the RV144 trial together suggest that the causal relationships between the complex sets of viral and immunological processes that contribute to protection or disease pathogenesis are still poorly understood. Here, we provide an up-to-date overview of HIV-host interactions at the cellular, the immune system and the neuroendocrine systems level. Only by integrating this multi-level knowledge one will be able to handle the systems complexity and develop new methodologies of analysis and prediction for a functional restoration of the immune system and the health of the infected host.
Collapse
Affiliation(s)
- V A Chereshnev
- Institute of Immunology and Physiology, Ural Branch RAS, Ekaterinburg, Russia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Wonderlich ER, Wijewardana V, Liu X, Barratt-Boyes SM. Virus-encoded TLR ligands reveal divergent functional responses of mononuclear phagocytes in pathogenic simian immunodeficiency virus infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:2188-98. [PMID: 23338235 PMCID: PMC3577972 DOI: 10.4049/jimmunol.1201645] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The role of mononuclear phagocytes in the pathogenesis or control of HIV infection is unclear. In this study, we monitored the dynamics and function of dendritic cells (DC) and monocytes/macrophages in rhesus macaques acutely infected with pathogenic SIVmac251 with and without antiretroviral therapy (ART). SIV infection was associated with monocyte mobilization and recruitment of plasmacytoid DC (pDC) and macrophages to lymph nodes, which did not occur with ART treatment. SIVmac251 single-stranded RNA encoded several uridine-rich sequences that were potent TLR7/8 ligands in mononuclear phagocytes of naive animals, stimulating myeloid DC (mDC) and monocytes to produce TNF-α and pDC and macrophages to produce both TNF-α and IFN-α. Following SIV infection, pDC and monocytes/macrophages rapidly became hyporesponsive to stimulation with SIV-encoded TLR ligands and influenza virus, a condition that was reversed by ART. The loss of pDC and macrophage function was associated with a profound but transient block in the capacity of lymph node cells to secrete IFN-α upon stimulation. In contrast to pDC and monocytes/macrophages, mDC increased TNF-α production in response to stimulation following acute infection. Moreover, SIV-infected rhesus macaques with stable infection had increased mDC responsiveness to SIV-encoded TLR ligands and influenza virus at set point, whereas animals that progressed rapidly to AIDS had reduced mDC responsiveness. These findings indicate that SIV encodes immunostimulatory TLR ligands and that pDC, mDC, and monocytes/macrophages respond to these ligands differently as a function of SIV infection. The data also suggest that increased responsiveness of mDC to stimulation following SIV infection may be beneficial to the host.
Collapse
Affiliation(s)
- Elizabeth R. Wonderlich
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261
| | - Viskam Wijewardana
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261
| | - Xiangdong Liu
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261
| | - Simon M. Barratt-Boyes
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261
| |
Collapse
|
17
|
Moens B, Pannecouque C, López G, Talledo M, Gotuzzo E, Khouri R, Bittencourt A, Farré L, Galvão-Castro B, Vandamme AM, Van Weyenbergh J. Simultaneous RNA quantification of human and retroviral genomes reveals intact interferon signaling in HTLV-1-infected CD4+ T cell lines. Virol J 2012; 9:171. [PMID: 22917064 PMCID: PMC3492208 DOI: 10.1186/1743-422x-9-171] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 08/17/2012] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND IFN-α contributes extensively to host immune response upon viral infection through antiviral, pro-apoptotic, antiproliferative and immunomodulatory activities. Although extensively documented in various types of human cancers and viral infections, controversy exists in the exact mechanism of action of IFN-α in human immunodeficiency virus type 1 (HIV-1) and human T-lymphotropic virus type 1 (HTLV-1) retroviral infections. RESULTS IFN-α displayed strong anti-HIV-1 effects in HIV-1/HTLV-1 co-infected MT-4 cells in vitro, demonstrated by the dose-dependent inhibition of the HIV-1-induced cytopathic effect (IC50 = 83.5 IU/ml, p < 0.0001) and p24 levels in cell-free supernatant (IC50 = 1.2 IU/ml, p < 0.0001). In contrast, IFN-α treatment did not affect cell viability or HTLV-1 viral mRNA levels in HTLV-1 mono-infected cell lines, based on flow cytometry and nCounter analysis, respectively. However, we were able to confirm the previously described post-transcriptional inhibition of HTLV-1 p19 secretion by IFN-α in cell lines (p = 0.0045), and extend this finding to primary Adult T cell Leukemia patient samples (p = 0.031). In addition, through microarray and nCounter analysis, we performed the first genome-wide simultaneous quantification of complete human and retroviral transciptomes, demonstrating significant transcriptional activation of interferon-stimulated genes without concomitant decrease of HTLV-1 mRNA levels. CONCLUSIONS Taken together, our results indicate that both the absence of in vitro antiproliferative and pro-apoptotic activity as well as the modest post-transcriptional antiviral activity of IFN-α against HTLV-1, were not due to a cell-intrinsic defect in IFN-α signalisation, but rather represents a retrovirus-specific phenomenon, considering the strong HIV-1 inhibition in co-infected cells.
Collapse
Affiliation(s)
- Britta Moens
- Laboratory of Clinical and Epidemiological Virology, Rega Institute for Medical Research, KULeuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Keating SM, Jacobs ES, Norris PJ. Soluble mediators of inflammation in HIV and their implications for therapeutics and vaccine development. Cytokine Growth Factor Rev 2012; 23:193-206. [PMID: 22743035 PMCID: PMC3418433 DOI: 10.1016/j.cytogfr.2012.05.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
From early in the HIV epidemic it was appreciated that many inflammatory markers such as neopterin and TNF-α were elevated in patients with AIDS. With the advent of modern technology able to measure a broad array of cytokines, we now know that from the earliest points of infection HIV induces a cytokine storm. This review will focus on how cytokines are disturbed in HIV infection and will explore potential therapeutic uses of cytokines. These factors can be used directly as therapy during HIV infection, either to suppress viral replication or prevent deleterious immune effects of infection, such as CD4+ T cell depletion. Cytokines also show great promise as adjuvants in the development of HIV vaccines, which would be critical for the eventual control of the epidemic.
Collapse
Affiliation(s)
- Sheila M Keating
- Blood Systems Research Institute, 270 Masonic Avenue, San Francisco, CA 94118, USA.
| | | | | |
Collapse
|
19
|
Interferon-alpha administration enhances CD8+ T cell activation in HIV infection. PLoS One 2012; 7:e30306. [PMID: 22291932 PMCID: PMC3265460 DOI: 10.1371/journal.pone.0030306] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 12/13/2011] [Indexed: 12/11/2022] Open
Abstract
Background Type I interferons play important roles in innate immune defense. In HIV infection, type I interferons may delay disease progression by inhibiting viral replication while at the same time accelerating disease progression by contributing to chronic immune activation. Methods To investigate the effects of type I interferons in HIV-infection, we obtained cryopreserved peripheral blood mononuclear cell samples from 10 subjects who participated in AIDS Clinical Trials Group Study 5192, a trial investigating the activity of systemic administration of IFNα for twelve weeks to patients with untreated HIV infection. Using flow cytometry, we examined changes in cell cycle status and expression of activation antigens by circulating T cells and their maturation subsets before, during and after IFNα treatment. Results The proportion of CD38+HLA-DR+CD8+ T cells increased from a mean of 11.7% at baseline to 24.1% after twelve weeks of interferon treatment (p = 0.006). These frequencies dropped to an average of 20.1% six weeks after the end of treatment. In contrast to CD8+ T cells, the frequencies of activated CD4+ T cells did not change with administration of type I interferon (mean percentage of CD38+DR+ cells = 2.62% at baseline and 2.17% after 12 weeks of interferon therapy). As plasma HIV levels fell with interferon therapy, this was correlated with a “paradoxical” increase in CD8+ T cell activation (p<0.001). Conclusion Administration of type I interferon increased expression of the activation markers CD38 and HLA DR on CD8+ T cells but not on CD4+ T cells of HIV+ persons. These observations suggest that type I interferons may contribute to the high levels of CD8+ T cell activation that occur during HIV infection.
Collapse
|
20
|
Tavel JA, Huang CY, Shen J, Metcalf JA, Dewar R, Shah A, Vasudevachari MB, Follmann DA, Herpin B, Davey RT, Polis MA, Kovacs J, Masur H, Lane HC. Interferon-alpha produces significant decreases in HIV load. J Interferon Cytokine Res 2011; 30:461-4. [PMID: 20235638 DOI: 10.1089/jir.2009.0090] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A randomized, controlled clinical trial was started in the pre-HAART era to compare the efficacy of zidovudine (AZT) and interferon-alpha (IFN-alpha) either alone or in combination to reduce HIV viremia, maintain CD4(+) cell count, and decrease time to AIDS progression and death. The purpose of the current study was to compare the effects of AZT and IFN on HIV load using modern technology. One hundred and eighty patients with CD4(+) counts above 500 cells/mm(3) were randomized to receive AZT alone, IFN-alpha alone, or AZT and IFN-alpha in combination. CD4(+) cell count and HIV load at baseline and at the last follow-up visit were compared, and time to AIDS or death was calculated by treatment group. At a mean follow-up of 45 weeks, the mean change in log HIV RNA was -0.06 for the AZT alone group, -0.47 for the AZT plus IFN-alpha group (P = 0.01 versus AZT group), and -0.35 for the IFN-alpha alone group (P = 0.02 versus AZT group). There was no significant difference among groups in change in total CD4(+) count or in time to AIDS or death. Since treatment with IFN-alpha produces significant decreases in HIV load, additional studies of IFN-alpha as part of a combination regimen are warranted.
Collapse
Affiliation(s)
- Jorge A Tavel
- Division of Clinical Research, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Saba NS, George TJ, Boulmay BC. Adjuvant high-dose interferon-{alpha} for resected melanoma in a patient with HIV infection. Oncologist 2010; 15:695-8. [PMID: 20555019 DOI: 10.1634/theoncologist.2009-0335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Adjuvant interferon (IFN)-alpha remains the standard adjuvant therapy for intermediate and high-risk melanoma after definitive surgical resection. Data addressing the role and safety of adjuvant immunotherapy in HIV-infected patients with melanoma are lacking. We report on an HIV(+) patient who received IFN-alpha as adjuvant treatment for high-risk melanoma. To our knowledge, this is the first reported case of such an approach.
Collapse
Affiliation(s)
- Nakhle S Saba
- Section of Hematology and Medical Oncology, Department of Medicine, Louisiana State University Health Sciences Center, 1542 Tulane Avenue, Box T4M-2, New Orleans, Louisiana 70112, USA
| | | | | |
Collapse
|
22
|
Abstract
OBJECTIVE The primary objectives of this study were to evaluate the safety, tolerability, and antiviral activity of pegylated interferon-alpha (PegIntron) in HIV-1 treatment-experienced patients failing their current antiretroviral regimen. DESIGN This was a phase II, multicenter, randomized, double-blind, placebo-controlled study. METHODS Patients were randomized to receive either weekly subcutaneous PegIntron 0.5, 1.0, 1.5, or 3 microg/kg or placebo added to their failing antiretroviral regimen for the first 4 weeks of study. Individuals who achieved more than 0.5 log10 reduction in HIV RNA at week 4 were allowed to continue study medication with optimization of their antiretroviral therapy for an additional 24 weeks. RESULTS In the 259 patients included in the intent-to-treat analysis, changes in plasma HIV RNA from baseline to week 4 were -0.25 (P > 0.5), -0.46 (P = 0.024), -0.39 (P = 0.008), -0.53 (P < 0.001), and -0.17 (P > 0.5) log10 copies/ml in the 0.5, 1.0, 1.5, and 3.0 microg/kg and placebo arms, respectively. No significant changes were seen in CD4 T-cell parameters in any of the treatment or control arms. Adverse events (most commonly fever, flu-like symptoms, other constitutional symptoms, and psychiatric symptoms) resulted in discontinuation of study medication in 13, 17, 16, 28, and 2% of patients in the 0.5, 1.0, 1.5, 3.0 microg/kg, and placebo group, respectively. CONCLUSION The demonstration of significant antiviral activity in a heavily pretreated patient population with acceptable toxicity and only weekly dosing makes PegIntron a potentially valuable therapy for patients with HIV infection that warrants further investigation in a broader population of patients.
Collapse
|
23
|
Asmuth DM, Abel K, George MD, Dandekar S, Pollard RB, Miller CJ. Pegylated interferon-alpha 2a treatment of chronic SIV-infected macaques. J Med Primatol 2008; 37:26-30. [PMID: 18199069 DOI: 10.1111/j.1600-0684.2007.00221.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND In vitro and clinical observations in HIV-infected patients receiving interferon alpha therapy have shown a reduction in HIV loads. Limited investigations have explored the innate or adaptive immune responses of IFN-alpha on SIV replication in vivo. METHODS Seven chronically infected rhesus macaques were given pegylated IFN-alpha 2a (n = four) or saline (n = three) injections once weekly for 14 weeks. Weekly peripheral blood samples were taken for safety parameters, viral load determinations, and measurements of innate and adaptive immune responses. RESULTS Pharmacokinetic measurements demonstrated therapeutic peg-IFN-alpha levels for the initial period of therapy and IFN-alpha inducible antiviral molecules were increased sporadically in the PBMC mRNA of the treatment group. Despite the demonstrable effect of the IFN-alpha injections, the treatment had no effect on plasma viral RNA levels. CONCLUSIONS This work demonstrates that while short term IFN-alpha therapy induces innate antiviral immunity, it does not dramatically enhance or suppress viral replication. However, studies in the SIV model to determine therapeutic potential of chronic IFN-alpha therapy for the treatment of HIV will require macaque specific cytokines.
Collapse
Affiliation(s)
- D M Asmuth
- Division of Infectious Diseases, Department of Internal Medicine, University of California-Davis Medical Center, 4150 V Street, Sacramento, CA 95817, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Sakuma R, Mael AA, Ikeda Y. Alpha interferon enhances TRIM5alpha-mediated antiviral activities in human and rhesus monkey cells. J Virol 2007; 81:10201-6. [PMID: 17609277 PMCID: PMC2045407 DOI: 10.1128/jvi.00419-07] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Accepted: 06/25/2007] [Indexed: 02/07/2023] Open
Abstract
Dominant, constitutively expressed antiretroviral factors, including TRIM5alpha and APOBEC3 proteins, are distinguished from the conventional innate immune systems and are classified as intrinsic immunity factors. Here, we demonstrate that interferon alpha (IFN-alpha) treatment upregulates TRIM5alpha mRNA in rhesus monkey cells, which correlates with the enhanced TRIM5alpha-mediated pre- and postintegration blocks of human immunodeficiency virus replication. In human cells, IFN-alpha increases the levels of TRIM5alpha mRNA, resulting in enhanced antiviral activity against N-tropic murine leukemia virus infection. These observations indicate that the TRIM5alpha-mediated antiviral effects can be orchestrated by the conventional innate immune response. It is conceivable that TRIM5alpha plays an essential role in controlling both the initial retroviral exposure and the subsequent viral dissemination in vivo.
Collapse
Affiliation(s)
- Ryuta Sakuma
- Molecular Medicine Program, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | | | | |
Collapse
|
25
|
Rodriguez B, Lederman MM, Jiang W, Bazdar DA, Gàrate K, Harding CV, Sieg SF. Interferon-alpha differentially rescues CD4 and CD8 T cells from apoptosis in HIV infection. AIDS 2006; 20:1379-89. [PMID: 16791012 DOI: 10.1097/01.aids.0000233571.51899.ab] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To examine the effects of interferon-alpha (IFN-alpha) on T cell survival and activation in HIV infection. DESIGN The effects of IFN-alpha on spontaneous apoptosis and CD38 expression among T cell subsets were determined in vitro and studied in relation to CD4 cell counts, plasma HIV RNA levels and the age of the subjects. METHODS Peripheral blood mononuclear cells from 48 HIV-infected persons and 17 healthy donors were incubated in vitro overnight with or without the addition of IFN-alpha. Percentages of apoptotic cells (positive for annexin V) and CD38 cells were determined among T cell subsets. RESULTS IFN-alpha inhibited spontaneous apoptosis of CD4 and CD8 T lymphocytes. This protective activity was impaired in CD4 T cells from HIV-infected persons. The reduced protection of IFN-alpha among CD4 cells from HIV-infected persons was not related to the percentages of activated (CD38 or CD45RO+CD38+) cells. Surprisingly, IFN-alpha induced CD38 expression among CD8 T cells from HIV-infected persons, and the magnitude of this effect was directly related to circulating CD4 T cell count. The CD8 T cell subset that expressed CD38 in response to IFN-alpha was defined as CD28 negative, CD62 ligand (CD62L) intermediate/negative. CONCLUSIONS Heightened expression of IFN-alpha in HIV infection may contribute to the phenotypic activation state that characterizes chronic infection while a diminished responsiveness of CD4 T cells to the protective effect of this cytokine may contribute to differential survival of CD4 and CD8 T cells in HIV disease.
Collapse
Affiliation(s)
- Benigno Rodriguez
- Division of Infectious Diseases, Case Western Reserve University, Center foe AIDS Research, University Hospitals of Cleveland, Cleveland, Ohio, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Alfano M, Poli G. Role of cytokines and chemokines in the regulation of innate immunity and HIV infection. Mol Immunol 2005; 42:161-82. [PMID: 15488606 DOI: 10.1016/j.molimm.2004.06.016] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The earliest defense against microbial infection is represented by the responses of the innate (or natural) immune system, that also profoundly regulates the adaptive (or acquired) T- and B-cell immune responses. Activation of the innate immune system is primed by microbial invasion in response to conserved structures present in large groups of microorganisms (LPS, peptidoglycan, double-stranded RNA), and is finely tuned by different cell types (including dendritic cells, macrophages, natural killer cells, natural killer T cells, and gammadelta T cells). In addition, several soluble factors (complement components, defensins, mannose-binding lectins, interferons, cytokines and chemokines) can play a major role in the regulation of both the innate and adaptive immunity. In this review, we will briefly overview the regulation of some cellular subsets of the innate immune system particularly involved in human immunodeficiency virus (HIV) infection and then focus our attention on those cytokines and chemokines whose levels of expression are more profoundly affected by HIV infection and that, conversely, can modulate virus infection and replication.
Collapse
Affiliation(s)
- Massimo Alfano
- AIDS Immunopathogenesis Unit, San Raffaele Scientific Institute, P2-P3 Laboratories, DIBIT, Via Olgettina no. 58, 20132 Milano, Italy
| | | |
Collapse
|
27
|
Audigé A, Schlaepfer E, Bonanomi A, Joller H, Knuchel MC, Weber M, Nadal D, Speck RF. HIV-1 does not provoke alteration of cytokine gene expression in lymphoid tissue after acute infection ex vivo. THE JOURNAL OF IMMUNOLOGY 2004; 172:2687-96. [PMID: 14764744 DOI: 10.4049/jimmunol.172.4.2687] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The cytokine response to invading microorganisms is critical for priming the adaptive immune response. During acute HIV infection, the response is disrupted, but the mechanism is poorly understood. We examined the cytokine response in human lymphoid tissue, acutely infected ex vivo with HIV. Lymphoid tissue was cultured either as blocks or as human lymphocyte aggregate cultures (HLAC) of tonsils and lymph nodes. This approach allowed us to examine the effects of HIV on cytokines using distinct culture techniques. In contrast to HLAC, mock-infected tissue blocks displayed a 50- to 100-fold up-regulation of mRNAs for IL-1beta, -6, and -8 in the first 6 days of culture. Parallel increases were also noted at the protein level in the supernatants. Although IL-1beta, -6, and -8 are known to synergistically enhance HIV replication, peak HIV replication (measured as p24 Ag) was similar in tissue blocks and HLAC. Surprisingly, vigorous HIV replication of CXCR4- and CCR5-tropic HIV strains did not result in characteristic mRNA profiles for IL-1beta, -2, -4, -6, -8, -10, -12, -15, IFN-gamma, TNF-alpha, TGF-beta, and beta-chemokines in tissue blocks or HLAC. The increased expression of IL-1beta, -6, and -8 in tissue blocks may approximate clinical situations with heightened immune activation; neutralization of these cytokines resulted in inhibition of HIV replication, suggesting that these cytokines may contribute to HIV replication in certain clinical settings. These results also indicate that different molecular mechanisms govern HIV replication in tissue blocks and HLAC. Prevention of effective cytokine responses may be an important mechanism that HIV uses during acute infection.
Collapse
Affiliation(s)
- Annette Audigé
- Division of Infectious Diseases and Hospital Epidemiology, Institute of Clinical Immunology, and Clinic of Visceral and Transplantation Surgery, University Hospital of Zurich, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
|
29
|
Jaworowski A, Maslin CLV, Wesselingh SL. The use of growth factors and cytokines to treat opportunistic infections in HIV-1 disease. Sex Health 2004; 1:161-74. [PMID: 16335304 DOI: 10.1071/sh03022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The success of highly active antiretroviral therapy (HAART) in reducing AIDS-related mortality means that in regions where HAART is available, HIV infection may now be regarded as a chronic disease. However the inability of HAART to eliminate HIV-1 from various anatomical and cellular reservoirs within the body means that HIV-infected individuals require life-long treatment with therapy that can have significant side effects. Management of HIV disease is therefore increasingly focused on drug-related toxicities and the improvement of current HAART regimens. Here we review the potential use of immunomodulatory cytokines to directly or indirectly stimulate the mononuclear phagocyte system as adjuncts to current HIV treatment as well as their use in the management of opportunistic infections in individuals who develop immunodeficiency. We argue that cytokines, which stimulate mononuclear phagocyte activity against opportunistic pathogens, may be useful for the treatment of individuals who develop recurrent opportunistic infections. Cytokines may act synergistically with antimicrobial agents to improve outcomes, which is of particular importance since recurrent infections frequently result in resistance to standard antimicrobial treatments. Before their use can be advocated however, given their toxicity and significant cost, the potential benefits of cytokines must be demonstrated in larger clinical trials.
Collapse
Affiliation(s)
- Anthony Jaworowski
- AIDS Pathogenesis and Clinical Research Program, The Burnet Institute for Medical Research and Public Health, Melbourne, Australia.
| | | | | |
Collapse
|
30
|
Kramer I, Shearer G. Quantifying the strength and durability of induced immunity to HIV infection in women engaging in unprotected sexual contacts with infected men. ACTA ACUST UNITED AC 2002. [DOI: 10.1016/s0895-7177(02)00299-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
31
|
Stylianou E, Yndestad A, Sikkeland LI, Bjerkeli V, Damås JK, Haug T, Eiken HG, Aukrust P, Frøland SS. Effects of interferon-alpha on gene expression of chemokines and members of the tumour necrosis factor superfamily in HIV-infected patients. Clin Exp Immunol 2002; 130:279-85. [PMID: 12390316 PMCID: PMC1906525 DOI: 10.1046/j.1365-2249.2002.01980.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We examined the effect of interferon (IFN)-alpha on the expression of 375 genes relevant to inflammatory and immunological reactions in peripheral blood mononuclear cells (PBMC) from HIV-infected patients by cDNA expression array and real-time quantitative RT-PCR. Our main findings were: (i) IFN-alpha induced up-regulation of several genes in the tumour necrosis factor (TNF) superfamily including the ligands APRIL, FasL, TNF-alpha and TRAIL, with particularly enhancing effects on the latter in HIV-infected patients. (ii) While IFN-alpha markedly up-regulated the expression of anti-angionetic ELR- CXC-chemokines (e.g. MIG and IP-10), it suppressed the expression of angiogenic ELR+ CXC-chemokines (e.g. GRO-alpha, IL-8 and ENA-78), with similar patterns in both patients and controls. (iii) IFN-alpha induced a marked increase in gene expression of the HIV co-receptor CCR5 in both patients and controls. We suggest that these effects may contribute to both the therapeutic and toxic effects of IFN-alpha. Moreover, our findings underscore that the biological effects of IFN-alpha in HIV infection are complex and that the clinical net effects of IFN-alpha treatment may be difficult to predict. However, the potent enhancing effect of IFN-alpha on several pro-apoptotic genes in the TNF superfamily and the enhancing effect on CCR5 expression suggest a possible pathogenic role of IFN-alpha in the progression of HIV-related immunodeficiency and suggests caution in the therapeutic use of IFN-alpha in HIV-infected -individuals.
Collapse
Affiliation(s)
- E Stylianou
- Section of Clinical Immunology and Infectious Diseases, Research Institute for Internal Medicine, Rikshospitalet, Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Immune reconstitution in HIV-infected patients remains a potential mechanism to explain delayed disease progression and increased survival following suppressive therapy. Many discrepancies remain to be studied. Is an immune response to HIV protective? Why are anti-HIV CD4 responses lost so quickly in progressors and how can they be restored? What is the damage to the immune system that occurs early in disease and why can it not be overcome by simply controlling viral replication? Will management of immune reconstitution be used in future adjunct treatment strategies (vaccine or STI)? Because HAART is not the answer to long-term management of HIV throughout the world, the recovery of immune function and it's potential to control viral replication remains a key goal in the long-term management of HIV-infected persons.
Collapse
Affiliation(s)
- Drew Weissman
- Division of Infectious Diseases, University of Pennsylvania, 522B Johnson Pavilion, Philadelphia, PA 19104, USA
| | | |
Collapse
|
33
|
Abel K, Alegria-Hartman MJ, Rothaeusler K, Marthas M, Miller CJ. The relationship between simian immunodeficiency virus RNA levels and the mRNA levels of alpha/beta interferons (IFN-alpha/beta) and IFN-alpha/beta-inducible Mx in lymphoid tissues of rhesus macaques during acute and chronic infection. J Virol 2002; 76:8433-45. [PMID: 12134046 PMCID: PMC155165 DOI: 10.1128/jvi.76.16.8433-8445.2002] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
To define the role of alpha/beta interferons (IFN-alpha/beta) in simian immunodeficiency virus (SIV) infection, IFN-alpha and IFN-beta mRNA levels and mRNA levels of Mx, an antiviral effector molecule, were determined in lymphoid tissues of rhesus macaques infected with pathogenic SIV. IFN-alpha/beta responses were induced during the acute phase and persisted in various lymphoid tissues throughout the chronic phase of infection. IFN-alpha/beta responses were most consistent in tissues with high viral RNA levels; thus, IFN-alpha/beta responses were not generally associated with effective control of SIV replication. IFN-alpha/beta responses were differentially regulated in different lymphoid tissues and at different stages of infection. The most consistent IFN-alpha/beta responses in acute and chronic SIV infection were observed in peripheral lymph nodes. In the spleen, only a transient increase in IFN-alpha/beta mRNA levels during acute SIV infection was observed. Further, IFN-alpha and IFN-beta mRNA levels showed a tissue-specific expression pattern during the chronic, but not the acute, phase of infection. In the acute phase of infection, SIV RNA levels in lymphoid tissues of rhesus macaques correlated with mRNA levels of both IFN-alpha and IFN-beta, whereas during chronic SIV infection only increased IFN-alpha mRNA levels correlated with the level of virus replication in the same tissues. In lymphoid tissues of all SIV-infected monkeys, higher viral RNA levels were associated with increased Mx mRNA levels. We found no evidence that monkeys with increased Mx mRNA levels in lymphoid tissues had enhanced control of virus replication. In fact, Mx mRNA levels were associated with high viral RNA levels in lymphoid tissues of chronically infected animals.
Collapse
Affiliation(s)
- Kristina Abel
- Center for Comparative Medicine, University of California-Davis, 95616, USA.
| | | | | | | | | |
Collapse
|
34
|
Affiliation(s)
- M C Allende
- Clinical and Molecular Retrovirology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
35
|
Talal AH, Canchis PW, Jacobson I. The HCV and HIV coinfected patient: what have we learned about pathophysiology? Curr Gastroenterol Rep 2002; 4:15-22. [PMID: 11825537 DOI: 10.1007/s11894-002-0033-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Hepatitis C virus (HCV) infection is an important problem in individuals who are also infected with HIV. HCV infection is very common in HIV-infected individuals, occurring in approximately one quarter to one third of this group, presumably as a consequence of shared routes of transmission related to virologic and pathogenic aspects of the viral infections. Although both are single-stranded RNA viruses and share similar epidemiologic properties, there are many important differences. Although the quantity of HIV RNA in plasma is an important prognostic determinant of HIV infection, this has not been shown with HCV. A direct relationship is apparent between HIV-related destruction of CD4 cells and the clinical consequences of the disease resulting from immunodeficiency. The pathogenesis of HCV, which occurs as a consequence of hepatic fibrosis, is much more complex. The hepatic stellate cell, the major producer of the extracellular matrix protein, is the main contributor to hepatic fibrosis, but the mechanism by which HCV induces hepatic fibrosis remains unclear. Treatment of HCV is increasingly important in HIV-infected patients due to improved HIV-associated morbidity and mortality and due to the frequency with which HCV occurs in patients with HIV-HCV coinfection. Timing of treatment initiation, management of side effects, and possible effects of anti-HCV therapy on HIV are among the issues that need consideration. Also, because several issues concerning HCV are unique to coinfected patients, further research is needed to determine optimal management of HCV in this setting.
Collapse
Affiliation(s)
- Andrew H Talal
- Weill Medical College of Cornell University, 525 East 68th Street, A354, New York, NY 10021, USA.
| | | | | |
Collapse
|
36
|
Ansari AA, Mayne AE, Sundstrom JB, Bostik P, Grimm B, Altman JD, Villinger F. Administration of recombinant rhesus interleukin-12 during acute simian immunodeficiency virus (SIV) infection leads to decreased viral loads associated with prolonged survival in SIVmac251-infected rhesus macaques. J Virol 2002; 76:1731-43. [PMID: 11799168 PMCID: PMC135900 DOI: 10.1128/jvi.76.4.1731-1743.2002] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ability of recombinant rhesus interleukin-12 (rMamu-IL-12) administration during acute simian immunodeficiency virus SIVmac251 infection to influence the quality of the antiviral immune responses was assessed in rhesus macaques. Group I (n = 4) was the virus-only control group. Group II and III received a conditioning regimen of rMamu-IL-12 (10 and 20 microg/kg, respectively, subcutaneously [s.c.]) on days -2 and 0. Thereafter, group II received 2 microg of IL-12 per kg and group III received 10 microg/kg s.c. twice a week for 8 weeks. On day 0 all animals were infected with SIVmac251 intravenously. While all four group I animals and three of four group II animals died by 8 and 10 months post infection (p.i.), all four group III animals remained alive for >20 months p.i. The higher IL-12 dose led to lower plasma viral loads and markedly lower peripheral blood mononuclear cell and lymph node proviral DNA loads. During the acute viremia phase, the high-IL-12-dose monkeys showed an increase in CD3(-) CD8 alpha/alpha(+) and CD3(+) CD8 alpha/alpha(+) cells and, unlike the control and low-IL-12-dose animals, did not demonstrate an increase in CD4(+) CD45RA(+) CD62L(+) naive cells. The high-IL-12-dose animals also demonstrated that both CD8 alpha/alpha(+) and CD8 alpha/beta(+) cells produced antiviral factors early p.i., whereas only CD8 alpha/beta(+) cells retained this function late p.i. Long-term survival correlated with sustained high levels of SIV gag/pol and SIV env cytotoxic T lymphocytes and retention of high memory responses against nominal antigens. This is the first study to demonstrate the capacity of IL-12 to significantly protect macaques from SIV-induced disease, and it provides a useful model to more precisely identify correlates of virus-specific disease-protective responses.
Collapse
Affiliation(s)
- A A Ansari
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
A variety of medications are used in treating patients infected with the human immunodeficiency virus (HIV). These medications are used to control viremia and to prevent and treat opportunistic infections. An individual is often required to take numerous drugs at the same time and thus clinicians are confronted with potential drug interactions, some of which are significant. Three different groups of anti-HIV drugs are used to treat patients. These groups include nucleoside reverse transcription inhibitors, non-nucleoside reverse transcription inhibitors, and protease inhibitors. This article reviews the most relevant drug interactions that occur during the treatment of HIV-infected patients with traditional and also alternative drugs. The role of therapeutic drug monitoring in the routine management of HIV-infected patients is discussed.
Collapse
Affiliation(s)
- A Dasgupta
- Department of Pathology and Laboratory Medicine and Internal Medicine, Division of Infectious Disease, University of Texas-Houston Medical School, Houston, Texas 77030, USA.
| | | |
Collapse
|
38
|
Abstract
Hepatitis C coinfection is common in patients with HIV, particularly in injection-drug users. Hepatitis C virus levels tend to be higher in coinfected patients, and histologic progression is more rapid than in patients with HCV alone. The efficacy of interferon monotherapy in HIV patients with an adequate CD4 cell count is comparable to that observed in patients without HIV. The combination of interferon plus ribavirin and pegylated interferon will further improve response rates. Interferon therapy is associated with leukopenia and a decrease in absolute CD4 cell count. Some concern remains that ribavirin might reduce the activity of pyrimidine analogues such as zidovudine and stavudine, and HIV-RNA levels should be followed when these medications are given concurrently. It is hoped that in time, new drug development will make the multiple-drug therapeutic strategy that has been highly successful in the management of HIV feasible for the treatment of HCV.
Collapse
Affiliation(s)
- S J Cotler
- Section of Hepatology, RUSH-Presbyterian-St. Luke's Medical Center, Chicago, Illinois, USA
| | | |
Collapse
|
39
|
Hatzakis A, Gargalianos P, Kiosses V, Lazanas M, Sypsa V, Anastassopoulou C, Vigklis V, Sambatakou H, Botsi C, Paraskevis D, Stalgis C. Low-dose IFN-alpha monotherapy in treatment-naive individuals with HIV-1 infection: evidence of potent suppression of viral replication. J Interferon Cytokine Res 2001; 21:861-9. [PMID: 11710999 DOI: 10.1089/107999001753238114] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
To evaluate the safety and antiviral action of interferon-alpha (IFN-alpha) in HIV-1 infection, we undertook a proof of concept study in 27 treatment-naive patients. Eligible patients comprised two groups: the IFN-alphaT group (n = 17), which received 5 MIU IFN-alpha s.c. daily for 32 consecutive days, and the IFN-alphaNT group (n = 10), which did not receive IFN-alpha prior to highly active antiretroviral therapy (HAART), which was commenced on day 28 in both groups. IFN-alphaTreatment was well tolerated in 14 of the 17 patients of the IFN-alphaT group who completed the study. The mean HIV RNA reduction in the IFN-alphaT group on day 14 was 1.1 log(10). Viral load suppression was inversely associated with baseline viral load (p = 0.031). Four weeks after initiation of HAART, IFN-alphaT and IFN-alphaNT group patients had 2.40 and 1.82 log(10) HIV RNA reduction from baseline, respectively (p < 0.001). There was no evidence of cross-resistance with existing antiretrovirals in patients with HIV-RNA rebound after initial plasma viral load decline > or = 1 log(10) during IFN-alpha monotherapy. Thus, low daily IFN-alpha exhibits potent anti-HIV-1 activity in vivo without serious adverse effects. These properties render IFN-alpha an attractive candidate for further assessment as a constituent of HAART.
Collapse
Affiliation(s)
- A Hatzakis
- Department of Hygiene and Epidemiology, Athens University Medical School, Athens, Greece.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Xiang J, Wünschmann S, Diekema DJ, Klinzman D, Patrick KD, George SL, Stapleton JT. Effect of coinfection with GB virus C on survival among patients with HIV infection. N Engl J Med 2001; 345:707-14. [PMID: 11547739 DOI: 10.1056/nejmoa003364] [Citation(s) in RCA: 258] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Previous studies have suggested that people with human immunodeficiency virus (HIV) infection who are coinfected with GB virus C (GBV-C, or hepatitis G virus) have delayed progression of HIV disease. GBV-C is related to hepatitis C virus but does not appear to cause liver disease. METHODS We examined the effect of coinfection with GBV-C on the survival of patients with HIV infection. We also evaluated cultures of peripheral-blood mononuclear cells infected with both viruses to determine whether GBV-C infection alters replication in vitro. RESULTS Of 362 HIV-infected patients, 144 (39.8 percent) had GBV-C viremia in two tests. Forty-one of the patients with GBV-C viremia (28.5 percent) died during the follow-up period, as compared with 123 of the 218 patients who tested negative for GBV-C RNA (56.4 percent; P<0.001). The mean duration of follow-up for the entire cohort was 4.1 years. In a Cox regression analysis adjusted for HIV treatment, baseline CD4+ T-cell count, age, sex, race, and mode of transmission of HIV, the mortality rate among the 218 HIV-infected patients without GBV-C coinfection was significantly higher than that among the 144 patients with GBV-C coinfection (relative risk, 3.7; 95 percent confidence interval, 2.5 to 5.4). HIV replication, as measured by the detection of p24 antigen in culture supernatants, was reproducibly inhibited in cultures of peripheral-blood mononuclear cells by GBV-C coinfection. Coinfection did not alter the surface expression of HIV cellular receptors on peripheral-blood mononuclear cells, as determined by flow cytometry. CONCLUSIONS GBV-C infection is common in people with HIV infection and is associated with significantly improved survival.
Collapse
Affiliation(s)
- J Xiang
- Department of Internal Medicine and Research, Iowa City Veterans Affairs Medical Center, and the University of Iowa College of Medicine, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Darcissac EC, Vidal V, Guillaume M, Thebault JJ, Bahr GM. Clinical tolerance and profile of cytokine induction in healthy volunteers following the simultaneous administration of ifn-alpha and the synthetic immunomodulator murabutide. J Interferon Cytokine Res 2001; 21:655-61. [PMID: 11576459 DOI: 10.1089/107999001753124381] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
As the therapeutic use of interferon-alpha (IFN-alpha) is limited by a dose-dependent toxicity and variable efficacy, ways of improving the therapeutic index of the cytokine are being sought. Murabutide (N-acetyl muramyl-L-alanyl-D-glutamine-O-n-butyl-ester) (ISTAC Biotechnology, Lille, France) is a safe synthetic and clinically acceptable immunomodulator that enhances the biologic activities of IFN-alpha in different experimental models. We evaluated in healthy human volunteers tolerance of the coadministration of Murabutide with increasing doses of IFN-alpha. The simultaneous administration of the two drugs was well tolerated without any increased or prohibiting toxicity, and all recipients experienced side effects that were similar to those observed after the administration of IFN-alpha alone. We also profiled the serum levels of cytokines induced following coinjection of the two drugs. We mostly detected an induction of anti-inflammatory cytokines and of human immunodeficiency virus type 1 (HIV-1)-suppressive beta-chemokines, in the absence of release of key proinflammatory cytokines. Therefore, the simultaneous administration of Murabutide and IFN-alpha is well tolerated and does not lead to increased toxicity. In addition, the selectivity in the profile of cytokines and chemokines induced following the coadministration of Murabutide and IFN-alpha points to the potential use of this combination in the treatment of inflammatory diseases and chronic viral infections.
Collapse
|
42
|
Torre-Cisneros J, Rivero A. [Management of chronic hepatitis C infection in patients infected with the human immunodeficiency virus: from what we know to what we ignore]. Med Clin (Barc) 2001; 116:775-8. [PMID: 11440683 DOI: 10.1016/s0025-7753(01)71984-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
43
|
Abstract
Cytokines play an important role in controlling the homoeostasis of the immune system. Infection with HIV results in dysregulation of the cytokine profile in vivo and in vitro. During the course of HIV-1 infection secretion of T-helper type 1 (Th1) cytokines, such as interleukin (IL)-2, and antiviral interferon (IFN)-gamma, is generally decreased, whereas production of T helper type 2 (Th2) cytokines, IL-4, IL-10, proinflammatory cytokines (IL-1, IL-6, IL-8) and tumour necrosis factor (TNF)-alpha, is increased. Such abnormal cytokine production contributes to the pathogenesis of the disease by impairing cell-mediated immunity. A number of cytokines have been shown to modulate in vitro HIV-1 infection and replication in both CD4 T lymphocytes and cells of macrophage lineage. HIV-inductive cytokines include: TNF-alpha, TNF-beta, IL-1 and IL-6, which stimulate HIV-1 replication in T cells and monocyte-derived macrophages (MDM), IL-2, IL-7 and IL-15, which upregulate HIV-1 in T cells, and macrophage-colony stimulating factor, which stimulates HIV-1 in MDM. HIV-suppressive cytokines include: IFN-alpha, IFN-beta and IL-16, which inhibit HIV-1 replication in T cells and MDM, and IL-10 and IL-13, which inhibit HIV-1 in MDM. Bifunctional cytokines such as IFN-gamma, IL-4 and granulocyte-macrophage colony-stimulating factor have been shown to have both inhibitory and stimulatory effects on HIV-1. The beta-chemokines, macrophage-inflammatory protein (MIP)-1alpha, MIP-1beta and RANTES are important inhibitors of macrophage-tropic strains of HIV-1, whereas the alpha-chemokine stromal-derived factor-1 suppresses infection of T-tropic strains of HIV-1. This review outlines the interactions between cytokines and HIV-1, and presents clinical applications of cytokine therapy combined with highly active antiretroviral therapy or vaccines.
Collapse
Affiliation(s)
- K Kedzierska
- AIDS Pathogenesis Research Unit, Macfarlane Burnet Centre for Medical Research, Fairfield, Australia
| | | |
Collapse
|