101
|
Mikulak J, Di Vito C, Zaghi E, Mavilio D. Host Immune Responses in HIV-1 Infection: The Emerging Pathogenic Role of Siglecs and Their Clinical Correlates. Front Immunol 2017; 8:314. [PMID: 28386256 PMCID: PMC5362603 DOI: 10.3389/fimmu.2017.00314] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/06/2017] [Indexed: 11/25/2022] Open
Abstract
A better understanding of the mechanisms employed by HIV-1 to escape immune responses still represents one of the major tasks required for the development of novel therapeutic approaches targeting a disease still lacking a definitive cure. Host innate immune responses against HIV-1 are key in the early phases of the infection as they could prevent the development and the establishment of two hallmarks of the infection: chronic inflammation and viral reservoirs. Sialic acid-binding immunoglobulin-like lectins (Siglecs) belong to a family of transmembrane proteins able to dampen host immune responses and set appropriate immune activation thresholds upon ligation with their natural ligands, the sialylated carbohydrates. This immune-modulatory function is also targeted by many pathogens that have evolved to express sialic acids on their surface in order to escape host immune responses. HIV-1 envelope glycoprotein 120 (gp120) is extensively covered by carbohydrates playing active roles in life cycle of the virus. Indeed, besides forming a protecting shield from antibody recognition, this coat of N-linked glycans interferes with the folding of viral glycoproteins and enhances virus infectivity. In particular, the sialic acid residues present on gp120 can bind Siglec-7 on natural killer and monocytes/macrophages and Siglec-1 on monocytes/macrophages and dendritic cells. The interactions between these two members of the Siglec family and the sialylated glycans present on HIV-1 envelope either induce or increase HIV-1 entry in conventional and unconventional target cells, thus contributing to viral dissemination and disease progression. In this review, we address the impact of Siglecs in the pathogenesis of HIV-1 infection and discuss how they could be employed as clinic and therapeutic targets.
Collapse
Affiliation(s)
- Joanna Mikulak
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Italy; Istituto di Ricerca Genetica e Biomedica, UOS di Milano, Consiglio Nazionale delle Ricerche (UOS/IRGB/CNR), Rozzano, Italy
| | - Clara Di Vito
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center , Rozzano , Italy
| | - Elisa Zaghi
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center , Rozzano , Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Italy; Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| |
Collapse
|
102
|
Araínga M, Edagwa B, Mosley RL, Poluektova LY, Gorantla S, Gendelman HE. A mature macrophage is a principal HIV-1 cellular reservoir in humanized mice after treatment with long acting antiretroviral therapy. Retrovirology 2017; 14:17. [PMID: 28279181 PMCID: PMC5345240 DOI: 10.1186/s12977-017-0344-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/06/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Despite improved clinical outcomes seen following antiretroviral therapy (ART), resting CD4+ T cells continue to harbor latent human immunodeficiency virus type one (HIV-1). However, such cells are not likely the solitary viral reservoir and as such defining where and how others harbor virus is imperative for eradication measures. To such ends, we used HIV-1ADA-infected NOD.Cg-Prkdc scid Il2rg tm1Wjl /SzJ mice reconstituted with a human immune system to explore two long-acting ART regimens investigating their abilities to affect viral cell infection and latency. At 6 weeks of infection animals were divided into four groups. One received long-acting (LA) cabotegravir (CAB) and rilpivirine (RVP) (2ART), a second received LA CAB, lamivudine, abacavir and RVP (4ART), a third were left untreated and a fourth served as an uninfected control. After 4 weeks of LA ART treatment, blood, spleen and bone marrow (BM) cells were collected then phenotypically characterized. CD4+ T cell subsets, macrophages and hematopoietic progenitor cells were analyzed for HIV-1 nucleic acids by droplet digital PCR. RESULTS Plasma viral loads were reduced by two log10 or to undetectable levels in the 2 and 4ART regimens, respectively. Numbers and distributions of CD4+ memory and regulatory T cells, macrophages and hematopoietic progenitor cells were significantly altered by HIV-1 infection and by both ART regimens. ART reduced viral DNA and RNA in all cell and tissue compartments. While memory cells were the dominant T cell reservoir, integrated HIV-1 DNA was also detected in the BM and spleen macrophages in both regimen-treated mice. CONCLUSION Despite vigorous ART regimens, HIV-1 DNA and RNA were easily detected in mature macrophages supporting their potential role as an infectious viral reservoir.
Collapse
Affiliation(s)
- Mariluz Araínga
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Benson Edagwa
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Larisa Y Poluektova
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA.
| |
Collapse
|
103
|
Feline Immunodeficiency Virus Neuropathogenesis: A Model for HIV-Induced CNS Inflammation and Neurodegeneration. Vet Sci 2017; 4:vetsci4010014. [PMID: 29056673 PMCID: PMC5606611 DOI: 10.3390/vetsci4010014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/22/2017] [Accepted: 03/01/2017] [Indexed: 12/11/2022] Open
Abstract
Feline Immunodeficiency virus (FIV), similar to its human analog human immunodeficiency virus (HIV), enters the central nervous system (CNS) soon after infection and establishes a protected viral reservoir. The ensuing inflammation and damage give rise to varying degrees of cognitive decline collectively known as HIV-associated neurocognitive disorders (HAND). Because of the similarities to HIV infection and disease, FIV has provided a useful model for both in vitro and in vivo studies of CNS infection, inflammation and pathology. This mini review summarizes insights gained from studies of early infection, immune cell trafficking, inflammation and the mechanisms of neuropathogenesis. Advances in our understanding of these processes have contributed to the development of therapeutic interventions designed to protect neurons and regulate inflammatory activity.
Collapse
|
104
|
Sanchez AB, Kaul M. Neuronal Stress and Injury Caused by HIV-1, cART and Drug Abuse: Converging Contributions to HAND. Brain Sci 2017; 7:brainsci7030025. [PMID: 28241493 PMCID: PMC5366824 DOI: 10.3390/brainsci7030025] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/15/2017] [Accepted: 02/20/2017] [Indexed: 12/21/2022] Open
Abstract
Multiple mechanisms appear to contribute to neuronal stress and injury underlying HIV-associated neurocognitive disorders (HAND), which occur despite the successful introduction of combination antiretroviral therapy (cART). Evidence is accumulating that components of cART can itself be neurotoxic upon long-term exposure. In addition, abuse of psychostimulants, such as methamphetamine (METH), seems to compromise antiretroviral therapy and aggravate HAND. However, the combined effect of virus and recreational and therapeutic drugs on the brain is still incompletely understood. However, several lines of evidence suggest a shared critical role of oxidative stress, compromised neuronal energy homeostasis and autophagy in promotion and prevention of neuronal dysfunction associated with HIV-1 infection, cART and psychostimulant use. In this review, we present a synopsis of recent work related to neuronal stress and injury induced by HIV infection, antiretrovirals (ARVs) and the highly addictive psychostimulant METH.
Collapse
Affiliation(s)
- Ana B Sanchez
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| | - Marcus Kaul
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
- Department of Psychiatry, University of California San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
105
|
DiNapoli SR, Ortiz AM, Wu F, Matsuda K, Twigg HL, Hirsch VM, Knox K, Brenchley JM. Tissue-resident macrophages can contain replication-competent virus in antiretroviral-naive, SIV-infected Asian macaques. JCI Insight 2017; 2:e91214. [PMID: 28239657 DOI: 10.1172/jci.insight.91214] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
SIV DNA can be detected in lymphoid tissue-resident macrophages of chronically SIV-infected Asian macaques. These macrophages also contain evidence of recently phagocytosed SIV-infected CD4+ T cells. Here, we examine whether these macrophages contain replication-competent virus, whether viral DNA can be detected in tissue-resident macrophages from antiretroviral (ARV) therapy-treated animals and humans, and how the viral sequences amplified from macrophages and contemporaneous CD4+ T cells compare. In ARV-naive animals, we find that lymphoid tissue-resident macrophages contain replication-competent virus if they also contain viral DNA in ARV-naive Asian macaques. The genetic sequence of the virus within these macrophages is similar to those within CD4+ T cells from the same anatomic sites. In ARV-treated animals, we find that viral DNA can be amplified from lymphoid tissue-resident macrophages of SIV-infected Asian macaques that were treated with ARVs for at least 5 months, but we could not detect replication-competent virus from macrophages of animals treated with ARVs. Finally, we could not detect viral DNA in alveolar macrophages from HIV-infected individuals who received ARVs for 3 years and had undetectable viral loads. These data demonstrate that macrophages can contain replication-competent virus, but may not represent a significant reservoir for HIV in vivo.
Collapse
Affiliation(s)
| | | | - Fan Wu
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Kenta Matsuda
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Homer L Twigg
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Vanessa M Hirsch
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Kenneth Knox
- Department of Medicine, University of Arizona, Tucson, Arizona, USA
| | | |
Collapse
|
106
|
Affiliation(s)
- Alex A. Compton
- Virus & Immunity Unit, Institut Pasteur, Paris, France
- * E-mail: (AAC); (OS)
| | - Olivier Schwartz
- Virus & Immunity Unit, Institut Pasteur, Paris, France
- CNRS-URA 3015, Paris, France
- Vaccine Research Institute, Creteil, France
- * E-mail: (AAC); (OS)
| |
Collapse
|
107
|
Agosto LM, Hirnet JB, Michaels DH, Shaik-Dasthagirisaheb YB, Gibson FC, Viglianti G, Henderson AJ. Porphyromonas gingivalis-mediated signaling through TLR4 mediates persistent HIV infection of primary macrophages. Virology 2016; 499:72-81. [PMID: 27639573 PMCID: PMC5126732 DOI: 10.1016/j.virol.2016.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/01/2016] [Accepted: 09/07/2016] [Indexed: 12/22/2022]
Abstract
Periodontal infections contribute to HIV-associated co-morbidities in the oral cavity and provide a model to interrogate the dysregulation of macrophage function, inflammatory disease progression, and HIV replication during co-infections. We investigated the effect of Porphyromonas gingivalis on the establishment of HIV infection in monocyte-derived macrophages. HIV replication in macrophages was significantly repressed in the presence of P. gingivalis. This diminished viral replication was due partly to a decrease in the expression of integrated HIV provirus. HIV repression depended upon signaling through TLR4 as knock-down of TLR4 with siRNA rescued HIV expression. Importantly, HIV expression was reactivated upon removal of P. gingivalis. Our observations suggest that exposure of macrophages to Gram-negative bacteria influence the establishment and maintenance of HIV persistence in macrophages through a TLR4-dependent mechanism.
Collapse
Affiliation(s)
- Luis M Agosto
- Department of Medicine, Section of Infectious Diseases, Boston Medical Center, Boston, MA, USA.
| | - Juliane B Hirnet
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| | - Daniel H Michaels
- Department of Medicine, Section of Infectious Diseases, Boston Medical Center, Boston, MA, USA
| | | | - Frank C Gibson
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610-0424, USA
| | - Gregory Viglianti
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| | - Andrew J Henderson
- Department of Medicine, Section of Infectious Diseases, Boston Medical Center, Boston, MA, USA; Department of Microbiology, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
108
|
Li W, Tong HI, Gorantla S, Poluektova LY, Gendelman HE, Lu Y. Neuropharmacologic Approaches to Restore the Brain's Microenvironment. J Neuroimmune Pharmacol 2016; 11:484-94. [PMID: 27352074 PMCID: PMC4985494 DOI: 10.1007/s11481-016-9686-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/25/2016] [Indexed: 12/15/2022]
Abstract
Maintaining the central nervous system microenvironment after injury, infection, inflammatory and degenerative diseases is contingent upon adequate control of glial homeostatic functions. Disease is caused by microbial, environmental and endogenous factors that compromise ongoing nervous system functions. The final result is neuronal injury, dropout and nerve connection loss, and these underlie the pathobiology of Alzheimer's and Parkinson's disease, amyotrophic lateral sclerosis, stroke, and bacterial, parasitic and viral infections. However, what promotes disease are homeostatic changes in the brain's microenvironment affected by innate glial immune pro-inflammatory and adaptive immune responses. These events disturb the brain's metabolic activities and communication abilities. How the process affects the brain's regulatory functions that can be harnessed for therapeutic gain is the subject at hand. Specific examples are provided that serve to modulate inflammation and improve disease outcomes specifically for HIV-associated neurocognitive disorders.
Collapse
Affiliation(s)
- Weizhe Li
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Hsin-I Tong
- Department of Public Health Sciences, Environmental Health Laboratory, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Larisa Y Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Yuanan Lu
- Department of Public Health Sciences, Environmental Health Laboratory, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
109
|
Ekholm S, Simon JH. Magnetic Resonance Imaging and the Acquired Immunodeficiency Syndrome Dementia Complex. Acta Radiol 2016. [DOI: 10.1177/028418518802900218] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
It is now recognized that patients infected by the virus linked to the acquired immunodeficiency syndrome (AIDS) can develop dementia symptoms as the initial and sometimes only symptomatology for AIDS. This appears to be a syndrome whose origin is independent of secondary non-viral infection or malignancy. Magnetic resonance imaging (MRI) in three patients with well documented AIDS dementia revealed high signal periventricular white matter lesions. In one case, large lesions were not apparent on computed tomography and gross inspection of the fixed brain prior to autopsy. In two cases in which serial in vivo MR studies were obtained, there was a progressive increase in lesion volume over a short (several months) period of observation. Periventricular white matter lesions may be an early sign accompanying AIDS dementia, and the degree of changes correlated well with the clinical picture in our patients.
Collapse
|
110
|
Salomon H, Gu Z, Gao Q, Nagai K, Hiscott J, Wainberg MA. Host Cell Dependence of Human Immunodeficiency Virus Type-1 Drug Resistance Profiles and Tissue Culture Selection Patterns. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/095632029500600404] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Clinical isolates of the human immunodeficiency virus type 1 (HIV-1) displayed differential sensitivity to antiviral nucleosides depending on the type of host cell employed for viral propagation. Viruses derived from the peripheral blood mononuclear cells (PBMC) of subjects on prolonged 3′-azido-3′-deoxythymidine (AZT) therapy behaved as AZT-resistant when tested in either cord blood mononuclear cells or MT-4 cells but as relatively drug-sensitive in the U-937 monocytic cell line. Viruses derived from monocytes/ macrophages of the same individuals behaved as drug-sensitive in all cells tested. It was also shown that cloned recombinant viruses, which contained defined resistance-conferring mutations at either position 65 or 184 in the HIV pol gene, were generally less susceptible to each of 2′-3′-dideoxyinosine (ddl), 2′,3′-dideoxycytidine (ddC) and the (-)enantiomer of 2′,3′-dideoxy-3′thiacytidine (3TC) in MT-4 cells than in any of PBMC, cord blood mononuclear cells (CBMC) or Jurkat cells. Finally, resistance against each of AZT, ddl and ddC could be selected for more easily using MT-4 cells than CBMC or Jurkat lymphocytes and not at all with the U-937 monocytic cell line.
Collapse
Affiliation(s)
- H. Salomon
- Lady Davis Institute and McGill University AIDS Centre, Jewish General Hospital, 3755 Cote Ste-Catherine Road, Montreal, Quebec H3T 1E2, Canada
| | - Z. Gu
- Lady Davis Institute and McGill University AIDS Centre, Jewish General Hospital, 3755 Cote Ste-Catherine Road, Montreal, Quebec H3T 1E2, Canada
| | - Q. Gao
- Lady Davis Institute and McGill University AIDS Centre, Jewish General Hospital, 3755 Cote Ste-Catherine Road, Montreal, Quebec H3T 1E2, Canada
| | - K. Nagai
- Lady Davis Institute and McGill University AIDS Centre, Jewish General Hospital, 3755 Cote Ste-Catherine Road, Montreal, Quebec H3T 1E2, Canada
| | - J. Hiscott
- Lady Davis Institute and McGill University AIDS Centre, Jewish General Hospital, 3755 Cote Ste-Catherine Road, Montreal, Quebec H3T 1E2, Canada
| | - M. A. Wainberg
- Lady Davis Institute and McGill University AIDS Centre, Jewish General Hospital, 3755 Cote Ste-Catherine Road, Montreal, Quebec H3T 1E2, Canada
| |
Collapse
|
111
|
Pollicita M, Ruff MR, Pert CB, Polianova MT, Schols D, Ranazzi A, Perno CF, Aquaro S. Profound Anti-HIV-1 Activity of DAPTA in Monocytes/macrophages and Inhibition of CCR5-mediated Apoptosis in Neuronal Cells. ACTA ACUST UNITED AC 2016; 18:285-95. [DOI: 10.1177/095632020701800504] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Monocytes/macrophages (M/M) are strategic reservoirs of HIV-1, spreading the virus to other cells and inducing apoptosis in T-lymphocytes, astrocytes and neurons. M/M are commonly infected by R5 HIV-1 strains, which use the chemokine receptor CCR5. D-Ala-peptide T-amide (DAPTA), or Peptide T, named for its high threonine content (ASTTTNYT), is a synthetic peptide comprised of eight amino acids (185–192) of the gp120 V2 region and functions as a viral entry inhibitor by targeting selectively CCR5. The anti-HIV-1 activity of DAPTA was evaluated in M/M infected with R5 HIV-1 strains. DAPTA at 10−9M inhibited HIV-1 replication in M/M by >90%. PCR analysis of viral cDNA in M/M showed that DAPTA blocks HIV entry and in this way prevents HIV-1 infection. Moreover, DAPTA acts as a strong inhibitor and was more active than the non-peptidic CCR5 antagonist TAK-779 in inhibiting apoptosis (mediated by R5 HIV-1 strains produced and released by infected M/M) on a neuroblastoma cell line. Our results suggest that antiviral compounds which interfere with receptor mechanisms such as CCR5 could be important, either alone or in combination with other antiretroviral treatments, in preventing HIV infection in the central nervous system and the consequential neuronal damage that leads to neuronal AIDS.
Collapse
Affiliation(s)
- Michela Pollicita
- Department of Experimental Medicine and Biochemical Science, University of Rome Tor Vergata, Rome, Italy
| | | | | | | | - Dominique Schols
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Alessandro Ranazzi
- Department of Experimental Medicine and Biochemical Science, University of Rome Tor Vergata, Rome, Italy
| | - Carlo-Federico Perno
- Department of Experimental Medicine and Biochemical Science, University of Rome Tor Vergata, Rome, Italy
| | - Stefano Aquaro
- Department of Experimental Medicine and Biochemical Science, University of Rome Tor Vergata, Rome, Italy
- Department of Pharmaco-Biology, University of Calabria, Rende, Italy
| |
Collapse
|
112
|
Craig JC, Grief C, Mills JS, Hockley D, Duncan IB, Roberts NA. Effects of a Specific Inhibitor of HIV Proteinase (Ro 31-8959) on Virus Maturation in a Chronically Infected Promonocytic Cell Line (U1). ACTA ACUST UNITED AC 2016. [DOI: 10.1177/095632029100200308] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The human immunodeficiency virus (HIV) proteinase inhibitor Ro 31-8959 prevents the maturation of virus in phorbol 12-myristate 13-acetate (PMA)-stimulated U1 cells, a chronically infected promonocytic cell line. Inhibition of both the morphological maturation of virions and the enzymic processing of gag polyprotein (p56) to produce capsid protein p24 was demonstrated at nanomolar concentrations of the compound. Furthermore, prolonged inhibition of the processing of p56 antigen was confirmed in pulse-chase experiments. The conclusion is that Ro 31-8959 can inhibit production of mature virions in a promonocyte cell line which is infected chronically/latently with HIV.
Collapse
Affiliation(s)
- J. C. Craig
- Roche Products Limited, PO Box 8, Welwyn Garden City, Herts AL7 3AY, UK
| | - C. Grief
- Department of Electron Microscopy and AIDS Collaborative Centre, National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Herts, UK
| | - J. S. Mills
- Roche Products Limited, PO Box 8, Welwyn Garden City, Herts AL7 3AY, UK
| | - D. Hockley
- Department of Electron Microscopy and AIDS Collaborative Centre, National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Herts, UK
| | - I. B. Duncan
- Roche Products Limited, PO Box 8, Welwyn Garden City, Herts AL7 3AY, UK
| | - N. A. Roberts
- Roche Products Limited, PO Box 8, Welwyn Garden City, Herts AL7 3AY, UK
| |
Collapse
|
113
|
Williams DW, Engle EL, Shirk EN, Queen SE, Gama L, Mankowski JL, Zink MC, Clements JE. Splenic Damage during SIV Infection: Role of T-Cell Depletion and Macrophage Polarization and Infection. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2068-2087. [PMID: 27322772 DOI: 10.1016/j.ajpath.2016.03.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 03/04/2016] [Accepted: 03/25/2016] [Indexed: 12/31/2022]
Abstract
The effects of HIV infection on spleen and its cellular subsets have not been fully characterized, particularly for macrophages in which diverse populations exist. We used an accelerated SIV-infected macaque model to examine longitudinal effects on T-cell and macrophage populations and their susceptibilities to infection. Substantial lymphoid depletion occurred, characterized by follicular burn out and a loss of CD3 T lymphocytes, which was associated with cellular activation and transient dysregulations in CD4/CD8 ratios and memory effector populations. In contrast, the loss of CD68 and CD163(+)CD68(+) macrophages and increase in CD163 cells was irreversible, which began during acute infection and persisted until terminal disease. Mac387 macrophages and monocytes were transiently recruited into spleen, but were not sufficient to mitigate the changes in macrophage subsets. Type I interferon, M2 polarizing genes, and chemokine-chemokine receptor signaling were up-regulated in spleen and drove macrophage alterations. SIV-infected T cells were numerous within the white pulp during acute infection, but were rarely observed thereafter. CD68, CD163, and Mac387 macrophages were highly infected, which primarily occurred in the red pulp independent of T cells. Few macrophages underwent apoptosis, indicating that they are a long-lasting target for HIV/SIV. Our results identify macrophages as an important contributor to HIV/SIV infection in spleen and in promoting morphologic changes through the loss of specific macrophage subsets that mediate splenic organization.
Collapse
Affiliation(s)
- Dionna W Williams
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth L Engle
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Erin N Shirk
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Suzanne E Queen
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lucio Gama
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joseph L Mankowski
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - M Christine Zink
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Janice E Clements
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
114
|
Matsuda K, Chen CY, Whitted S, Chertova E, Roser DJ, Wu F, Plishka RJ, Ourmanov I, Buckler-White A, Lifson JD, Strebel K, Hirsch VM. Enhanced antagonism of BST-2 by a neurovirulent SIV envelope. J Clin Invest 2016; 126:2295-307. [PMID: 27159392 PMCID: PMC4887162 DOI: 10.1172/jci83725] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 03/22/2016] [Indexed: 11/17/2022] Open
Abstract
Current antiretroviral therapy (ART) is not sufficient to completely suppress disease progression in the CNS, as indicated by the rising incidence of HIV-1-associated neurocognitive disorders (HAND) among infected individuals on ART. It is not clear why some HIV-1-infected patients develop HAND, despite effective repression of viral replication in the circulation. SIV-infected nonhuman primate models are widely used to dissect the mechanisms of viral pathogenesis in the CNS. Here, we identified 4 amino acid substitutions in the cytoplasmic tail of viral envelope glycoprotein gp41 of the neurovirulent virus SIVsm804E that enhance replication in macrophages and associate with enhanced antagonism of the host restriction factor BM stromal cell antigen 2 (BST-2). Rhesus macaques were inoculated with a variant of the parental virus SIVsmE543-3 that had been engineered to contain the 4 amino acid substitutions present in gp41 of SIVsm804E. Compared with WT virus-infected controls, animals infected with mutant virus exhibited higher viral load in cerebrospinal fluid. Together, these results are consistent with a potential role for BST-2 in the CNS microenvironment and suggest that BST-2 antagonists may serve as a possible target for countermeasures against HAND.
Collapse
Affiliation(s)
- Kenta Matsuda
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Chia-Yen Chen
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Sonya Whitted
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Elena Chertova
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - David J. Roser
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Fan Wu
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Ronald J. Plishka
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Ilnour Ourmanov
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Alicia Buckler-White
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Klaus Strebel
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Vanessa M. Hirsch
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| |
Collapse
|
115
|
Meeker R, English R, Tompkins M. Enhanced Excitotoxicity in Primary Feline Neural Cultures Exposed to Feline Immunodeficiency Virus (FIV). ACTA ACUST UNITED AC 2016; 1:1-27. [PMID: 16873168 DOI: 10.1300/j128v01n03_01] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The ability of feline immunodeficiency virus (FIV) to induce neurodegenerative changes in vitro similar to those due to HIV was examined as a potential model to examine the mechanisms underlying AIDS dementia. Primary cultures of feline neural tissue (neurons, astrocytes and microglia) were established from E40-E57 fetal cat cortex and challenged by inoculation with the NCSU<sub>1</sub> strain of FIV. Proviral FIV was detected in the cultures and correlated with the presence of microglia. No direct toxicity of FIV was seen. Stimulation of FIV-inoculated cortical cultures with 20 uM glutamate resulted in a greatly enhanced acute swelling response in approximately 14-24% of the neurons and an increase in the number of dead cells after 24 h relative to control cultures. The enhanced responses were due to an increase in the sensitivity of the cells to glutamate and were dependent on the presence of a soluble factor in the medium. The similarity of the indirect excitoxic effects of FIV to current models of HIV-gp120 neurotoxicity and the versatility of the in vitro cultures, indicate that FIV should provide a valuable model for the investigation of the mechanisms of neurodegeneration in AIDS dementia.
Collapse
|
116
|
Kok YL, Vongrad V, Shilaih M, Di Giallonardo F, Kuster H, Kouyos R, Günthard HF, Metzner KJ. Monocyte-derived macrophages exhibit distinct and more restricted HIV-1 integration site repertoire than CD4(+) T cells. Sci Rep 2016; 6:24157. [PMID: 27067385 PMCID: PMC4828718 DOI: 10.1038/srep24157] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/21/2016] [Indexed: 11/09/2022] Open
Abstract
The host genetic landscape surrounding integrated HIV-1 has an impact on the fate of the provirus. Studies analysing HIV-1 integration sites in macrophages are scarce. We studied HIV-1 integration site patterns in monocyte-derived macrophages (MDMs) and activated CD4(+) T cells derived from seven antiretroviral therapy (ART)-treated HIV-1-infected individuals whose cells were infected ex vivo with autologous HIV-1 isolated during the acute phase of infection. A total of 1,484 unique HIV-1 integration sites were analysed. Their distribution in the human genome and genetic features, and the effects of HIV-1 integrase polymorphisms on the nucleotide selection specificity at these sites were indistinguishable between the two cell types, and among HIV-1 isolates. However, the repertoires of HIV-1-hosting gene clusters overlapped to a higher extent in MDMs than in CD4(+) T cells. The frequencies of HIV-1 integration events in genes encoding HIV-1-interacting proteins were also different between the two cell types. Lastly, HIV-1-hosting genes linked to clonal expansion of latently HIV-1-infected CD4(+) T cells were over-represented in gene hotspots identified in CD4(+) T cells but not in those identified in MDMs. Taken together, the repertoire of genes targeted by HIV-1 in MDMs is distinct from and more restricted than that of CD4(+) T cells.
Collapse
Affiliation(s)
- Yik Lim Kok
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Valentina Vongrad
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Mohaned Shilaih
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Francesca Di Giallonardo
- Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, School of Biological Sciences and Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Herbert Kuster
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Roger Kouyos
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Karin J Metzner
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
117
|
Graziano F, Vicenzi E, Poli G. Immuno-Pharmacological Targeting of Virus-Containing Compartments in HIV-1-Infected Macrophages. Trends Microbiol 2016; 24:558-567. [PMID: 27012511 DOI: 10.1016/j.tim.2016.02.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/23/2016] [Accepted: 02/26/2016] [Indexed: 01/26/2023]
Abstract
In addition to CD4 T lymphocytes, HIV-1 infects tissue macrophages that can actively accumulate infectious virions in vacuolar subcellular structures mostly connected to the plasma membrane and recently termed virus-containing compartments (VCCs). The VCC-associated HIV-1 reservoir of infected macrophages can be either increased or depleted by immunologic and pharmacologic agents, at least in vitro, thus suggesting that these factors (or related molecules) could be effective in curtailing the macrophage-associated HIV-1 reservoir in infected individuals receiving combination antiretroviral therapy (cART). Here we review evidence on the pathogenic role of tissue macrophages as long-term viral reservoirs in vivo and upon in vitro infection with a particular emphasis on the immuno-pharmacological modulation of VCCs.
Collapse
Affiliation(s)
- Francesca Graziano
- AIDS Immunopathogenesis Unit, San Raffaele Scientific Institute, Milano, Italy
| | - Elisa Vicenzi
- Viral Pathogens and Biosafety Unit, San Raffaele Scientific Institute, Milano, Italy
| | - Guido Poli
- AIDS Immunopathogenesis Unit, San Raffaele Scientific Institute, Milano, Italy; Vita-Salute San Raffaele University, School of Medicine, Milano, Italy; Institute of Human Virology, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
118
|
Fujita M. Commentary: MARCH8 Inhibits HIV-1 Infection by Reducing Virion Incorporation of Envelope Glycoproteins. Front Microbiol 2016; 7:254. [PMID: 26941737 PMCID: PMC4764732 DOI: 10.3389/fmicb.2016.00254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 02/15/2016] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mikako Fujita
- Research Institute for Drug Discovery, School of Pharmacy, Kumamoto University Kumamoto, Japan
| |
Collapse
|
119
|
Abstract
CNS infection is a nearly constant facet of systemic CNS infection and is generally well controlled by suppressive systemic antiretroviral therapy (ART). However, there are instances when HIV can be detected in the cerebrospinal fluid (CSF) despite suppression of plasma viruses below the clinical limits of measurement. We review three types of CSF viral escape: asymptomatic, neuro-symptomatic, and secondary. The first, asymptomatic CSF escape, is seemingly benign and characterized by lack of discernable neurological deterioration or subsequent CNS disease progression. Neuro-symptomatic CSF escape is an uncommon, but important, entity characterized by new or progressive CNS disease that is critical to recognize clinically because of its management implications. Finally, secondary CSF escape, which may be even more uncommon, is defined by an increase of CSF HIV replication in association with a concomitant non-HIV infection, as a consequence of the local inflammatory response. Understanding these CSF escape settings not only is important for clinical diagnosis and management but also may provide insight into the CNS HIV reservoir.
Collapse
|
120
|
Abstract
The persistence of human immunodeficiency virus type 1 (HIV-1) in latent reservoirs is a major barrier to HIV cure. Reservoir establishment depends on low viral expression that may be related to provirus integration sites (IS). In vitro, in cell lines and primary T cells, latency is associated with specific IS through reduced viral expression mediated by transcriptional interference by host cellular promoters, reverse orientation, and the presence of specific epigenetic modifiers. In primary T cell models of latency, specific IS are associated with intracellular viral antigen expression that is not directly related to cell activation. In contrast, in patient CD4+ T cells, there is enrichment for IS in genes controlling cell cycle and survival and in some clonally expanded T cell subpopulations. Multiple insertion sites within some specific genes may suggest that integrated HIV can increase the host’s T cell survival.
Collapse
Affiliation(s)
- Simin D. Rezaei
- Faculty of Medicine, Dentistry and Health Sciences, Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, The University of Melbourne, 4th Floor, 786-798 Elizabeth St, Melbourne, 3010 Australia
| | - Paul U. Cameron
- Faculty of Medicine, Dentistry and Health Sciences, Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, The University of Melbourne, 4th Floor, 786-798 Elizabeth St, Melbourne, 3010 Australia
- Infectious Diseases Unit, Department of Infectious Diseases, Alfred Hospital, 85 Commercial Rd, Melbourne, Victoria 3004 Australia
| |
Collapse
|
121
|
Sen S, Kaminiski R, Deshmane S, Langford D, Khalili K, Amini S, Datta PK. Role of hexokinase-1 in the survival of HIV-1-infected macrophages. Cell Cycle 2015; 14:980-9. [PMID: 25602755 DOI: 10.1080/15384101.2015.1006971] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Viruses have developed various strategies to protect infected cells from apoptosis. HIV-1 infected macrophages are long-lived and considered reservoirs for HIV-1. One significant deciding factor between cell survival and cell death is glucose metabolism. We hypothesized that HIV-1 protects infected macrophages from apoptosis in part by modulating the host glycolytic pathway specifically by regulating hexokinase-1 (HK-1) an enzyme that converts glucose to glucose-6-phosphate. Therefore, we analyzed the regulation of HK-1 in HIV-1 infected PBMCs, and in a chronically HIV-1 infected monocyte-like cell line, U1. Our results demonstrate that HIV-1 induces a robust increase in HK-1 expression. Surprisingly, hexokinase enzymatic activity was significantly inhibited in HIV-1 infected PBMCs and in PMA differentiated U1 cells. Interestingly, we observed increased levels of mitochondria-bound HK-1 in PMA induced U1 cells and in the HIV-1 accessory protein, viral protein R (Vpr) transduced U937 cell derived macrophages. Dissociation of HK-1 from mitochondria in U1 cells using a pharmacological agent, clotrimazole (CTZ) induced mitochondrial membrane depolarization and caspase-3/7 mediated apoptosis. Dissociation of HK-1 from mitochondria in Vpr transduced U937 also activated caspase-3/7 activity. These observations indicate that HK-1 plays a non-metabolic role in HIV-1 infected macrophages by binding to mitochondria thereby maintaining mitochondrial integrity. These results suggest that targeting the interaction of HK-1 with the mitochondria to induce apoptosis in persistently infected macrophages may prove beneficial in purging the macrophage HIV reservoir.
Collapse
Key Words
- COXIV, Cytochrome c oxidase subunit IV
- CTZ, Clotrimazole
- G-6-P, glucose-6-phosphate
- G6PD, glucose-6-phosphate dehydrogenase
- HIV-1
- HK-1, Hexokinase-1
- M-CSF, macrophage colony-stimulating factor
- OMM, outer mitochondrial membrane
- VDAC, voltage-dependent anion channel
- Vpr, viral protein R
- apoptosis, glucose metabolism
- cART, combination antiretroviral therapy
- hexokinase
- macrophage
- mitochondria
Collapse
Affiliation(s)
- Satarupa Sen
- a Department of Biology ; College of Science and Technology ; Temple University ; Philadelphia , PA USA
| | | | | | | | | | | | | |
Collapse
|
122
|
Wang MQ, Huang YL, Huang J, Zheng JL, Qian GX. RIG-I detects HIV-1 infection and mediates type I interferon response in human macrophages from patients with HIV-1-associated neurocognitive disorders. GENETICS AND MOLECULAR RESEARCH 2015; 14:13799-811. [PMID: 26535695 PMCID: PMC4864023 DOI: 10.4238/2015.october.28.42] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The aim of this study was to explore the precise role of retinoic acid-inducible gene-I (RIG-I) signaling in human immunodeficiency virus type 1 (HIV-1)-infected macrophages from patients with HIV-1-associated neurocognitive disorders (HAND). Postmortem brain tissues were collected from patients with HIV-1-associated dementia and were compared to samples collected from HIV serum-positive patients without dementia and HIV serum-negative patients. A human monocyte-derived macrophage (MDM) primary culture system was established to evaluate the expression of RIG-I in these samples. Knockdown of RIG-I pathways genes was employed and STAT1 expression and phosphorylation levels were examined to explore the molecular mechanisms of HAND. The expression of RIG-I in postmortem brain tissue from HAND patients was significantly higher than in patients who were HIV serum-positive without dementia or HIV serum-negative. Moreover, we demonstrated that HIV-1 infection could result in a significant increase in the level of RIG-I in human MDMs. Moreover, a correlation was found between the increase in RIG-I expression and STAT1 expression and phosphorylation. Accordingly, knockdown of RIG-I decreased the phosphorylation of STAT1 and downregulated interferon-related genes. These observations highlight the importance of RIG-I signaling in anti-HIV innate immunity in macrophages, which may be beneficial for the treatment of HIV and aid in the understanding of the neuropathogenesis of HAND.
Collapse
Affiliation(s)
- M Q Wang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Y L Huang
- Laboratory of Neuroimmunology and Regenerative Therapy, University of Nebraska Medical Center, Omaha, NE, USA
| | - J Huang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - J L Zheng
- Laboratory of Neuroimmunology and Regenerative Therapy, University of Nebraska Medical Center, Omaha, NE, USA
| | - G X Qian
- Department of Biochemistry and Molecular Biology, ShanghaiJiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
123
|
Fitting S, Zou S, El-Hage N, Suzuki M, Paris JJ, Schier CJ, Rodríguez JW, Rodriguez M, Knapp PE, Hauser KF. Opiate addiction therapies and HIV-1 Tat: interactive effects on glial [Ca²⁺]i, oxyradical and neuroinflammatory chemokine production and correlative neurotoxicity. Curr HIV Res 2015; 12:424-34. [PMID: 25760046 PMCID: PMC4475822 DOI: 10.2174/1570162x1206150311161147] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 12/13/2014] [Accepted: 02/12/2015] [Indexed: 01/05/2023]
Abstract
Few preclinical studies have compared the relative therapeutic efficacy of medications used to treat opiate addiction in relation to neuroAIDS. Here we compare the ability of methadone and buprenorphine, and the prototypic opiate morphine, to potentiate the neurotoxic and proinflammatory ([Ca2+]i, ROS, H2O2, chemokines) effects of HIV-1 Tat in neuronal and/or mixed-glial co-cultures. Repeated observations of neurons during 48 h exposure to combinations of Tat, equimolar concentrations (500 nM) of morphine, methadone, or buprenorphine exacerbated neurotoxicity significantly above levels seen with Tat alone. Buprenorphine alone displayed marked neurotoxicity at 500 nM, prompting additional studies of its neurotoxic effects at 5 nM and 50 nM concentrations ± Tat. In combination with Tat, buprenorphine displayed paradoxical, concentration-dependent, neurotoxic and neuroprotective actions. Buprenorphine neurotoxicity coincided with marked elevations in [Ca2+]i, but not increases in glial ROS or chemokine release. Tat by itself elevated the production of CCL5/RANTES, CCL4/MIP-1β, and CCL2/MCP-1. Methadone and buprenorphine alone had no effect, but methadone interacted with Tat to further increase production of CCL5/RANTES. In combination with Tat, all drugs significantly increased glial [Ca2+]i, but ROS was only significantly increased by co-exposure with morphine. Taken together, the increases in glial [Ca2+]i, ROS, and neuroinflammatory chemokines were not especially accurate predictors of neurotoxicity. Despite similarities, opiates displayed differences in their neurotoxic and neuroinflammatory interactions with Tat. Buprenorphine, in particular, was partially neuroprotective at a low concentration, which may result from its unique pharmacological profile at multiple opioid receptors. Overall, the results reveal differences among addiction medications that may impact neuroAIDS.
Collapse
|
124
|
Cobos Jiménez V, Martinez FO, Booiman T, van Dort KA, van de Klundert MAA, Gordon S, Geijtenbeek TBH, Kootstra NA. G3BP1 restricts HIV-1 replication in macrophages and T-cells by sequestering viral RNA. Virology 2015; 486:94-104. [PMID: 26432022 DOI: 10.1016/j.virol.2015.09.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/11/2015] [Accepted: 09/13/2015] [Indexed: 11/16/2022]
Abstract
HIV-1 exploits the cellular machinery for replication and therefore several interactions with cellular factors take place, some of which are yet unknown. We identified GTPase-activating protein-(SH3 domain)-binding protein 1 (G3BP1) as a cellular factor that restricts HIV-1, by analyzing transcriptome profiles of in vitro-cytokine-activated macrophages that are non-permissive to HIV-1 replication. Silencing of G3BP1 by RNA interference resulted in increased HIV-1 replication in primary T-cells and macrophages, but did not affect replication of other retroviruses. G3BP1 specifically interacted with HIV-1 RNA in the cytoplasm, suggesting that it sequesters viral transcripts, thus preventing translation or packaging. G3BP1 was highly expressed in resting naïve or memory T-cells from healthy donors and HIV-1 infected patients, but significantly lower in IL-2-activated T-cells. These results strongly suggest that G3BP1 captures HIV-1 RNA transcripts and thereby restricts mRNA translation, viral protein production and virus particle formation.
Collapse
Affiliation(s)
- Viviana Cobos Jiménez
- Department of Experimental Immunology, Sanquin Research, Landsteiner Laboratory, and Center for Infectious Diseases and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| | - Fernando O Martinez
- Kennedy Rheumatology Institute, University of Oxford, Oxford OX3 7LD, United Kingdom
| | - Thijs Booiman
- Department of Experimental Immunology, Sanquin Research, Landsteiner Laboratory, and Center for Infectious Diseases and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| | - Karel A van Dort
- Department of Experimental Immunology, Sanquin Research, Landsteiner Laboratory, and Center for Infectious Diseases and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| | - Maarten A A van de Klundert
- Department of Experimental Immunology, Sanquin Research, Landsteiner Laboratory, and Center for Infectious Diseases and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| | - Siamon Gordon
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Sanquin Research, Landsteiner Laboratory, and Center for Infectious Diseases and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| | - Neeltje A Kootstra
- Department of Experimental Immunology, Sanquin Research, Landsteiner Laboratory, and Center for Infectious Diseases and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam 1105AZ, The Netherlands.
| |
Collapse
|
125
|
Alteri C, Surdo M, Bellocchi MC, Saccomandi P, Continenza F, Armenia D, Parrotta L, Carioti L, Costa G, Fourati S, Di Santo F, Scutari R, Barbaliscia S, Fedele V, Carta S, Balestra E, Alcaro S, Marcelin AG, Calvez V, Ceccherini-Silberstein F, Artese A, Perno CF, Svicher V. Incomplete APOBEC3G/F Neutralization by HIV-1 Vif Mutants Facilitates the Genetic Evolution from CCR5 to CXCR4 Usage. Antimicrob Agents Chemother 2015; 59:4870-81. [PMID: 26055363 PMCID: PMC4505216 DOI: 10.1128/aac.00137-15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 05/30/2015] [Indexed: 11/20/2022] Open
Abstract
Incomplete APOBEC3G/F neutralization by a defective HIV-1Vif protein can promote genetic diversification by inducing G-to-A mutations in the HIV-1 genome. The HIV-1 Env V3 loop, critical for coreceptor usage, contains several putative APOBEC3G/F target sites. Here, we determined if APOBEC3G/F, in the presence of Vif-defective HIV-1 virus, can induce G-to-A mutations at V3 positions critical to modulation of CXCR4 usage. Peripheral blood mononuclear cells (PBMC) and monocyte-derived macrophages (MDM) from 2 HIV-1-negative donors were infected with CCR5-using 81.A-VifWT virus (i.e., with wild-type [WT] Vif protein), 81.A-VifE45G, or 81.A-VifK22E (known to incompletely/partially neutralize APOBEC3G/F). The rate of G-toA mutations was zero or extremely low in 81.A-VifWT- and 81.A-VifE45G-infected PBMC from both donors. Conversely, G-to-A enrichment was detected in 81.A-VifK22E-infected PBMC (prevalence ranging from 2.18% at 7 days postinfection [dpi] to 3.07% at 21 dpi in donor 1 and from 10.49% at 7 dpi to 8.69% at 21 dpi in donor 2). A similar scenario was found in MDM. G-to-A mutations occurred at 8 V3 positions, resulting in nonsynonymous amino acid substitutions. Of them, G24E and E25K strongly correlated with phenotypically/genotypically defined CXCR4-using viruses (P = 0.04 and 5.5e-7, respectively) and increased the CXCR4 N-terminal binding affinity for V3 (WT, -40.1 kcal/mol; G24E, -510 kcal/mol; E25K, -522 kcal/mol). The analysis of paired V3 and Vif DNA sequences from 84 HIV-1-infected patients showed that the presence of a Vif-defective virus correlated with CXCR4 usage in proviral DNA (P = 0.04). In conclusion, incomplete APOBEC3G/F neutralization by a single Vif amino acid substitution seeds a CXCR4-using proviral reservoir. This can have implications for the success of CCR5 antagonist-based therapy, as well as for the risk of disease progression.
Collapse
Affiliation(s)
- Claudia Alteri
- University of Rome "Tor Vergata," Department of Experimental Medicine and Surgery, Rome, Italy
| | - Matteo Surdo
- University of Rome "Tor Vergata," Department of Experimental Medicine and Surgery, Rome, Italy
| | | | - Patrizia Saccomandi
- University of Rome "Tor Vergata," Department of Experimental Medicine and Surgery, Rome, Italy
| | | | - Daniele Armenia
- University of Rome "Tor Vergata," Department of Experimental Medicine and Surgery, Rome, Italy
| | - Lucia Parrotta
- Università Magna Graecia di Catanzaro, Dipartimento di Scienze della Salute, Campus Universitario, Catanzaro, Italy
| | - Luca Carioti
- University of Rome "Tor Vergata," Department of Experimental Medicine and Surgery, Rome, Italy
| | - Giosuè Costa
- Università Magna Graecia di Catanzaro, Dipartimento di Scienze della Salute, Campus Universitario, Catanzaro, Italy
| | - Slim Fourati
- Department of Virology, Hospital "Pitie Salpietrere," Paris, France
| | - Fabiola Di Santo
- University of Rome "Tor Vergata," Department of Experimental Medicine and Surgery, Rome, Italy
| | - Rossana Scutari
- University of Rome "Tor Vergata," Department of Experimental Medicine and Surgery, Rome, Italy
| | - Silvia Barbaliscia
- University of Rome "Tor Vergata," Department of Experimental Medicine and Surgery, Rome, Italy
| | | | | | - Emanuela Balestra
- University of Rome "Tor Vergata," Department of Experimental Medicine and Surgery, Rome, Italy
| | - Stefano Alcaro
- Università Magna Graecia di Catanzaro, Dipartimento di Scienze della Salute, Campus Universitario, Catanzaro, Italy
| | | | - Vincent Calvez
- Department of Virology, Hospital "Pitie Salpietrere," Paris, France
| | | | - Anna Artese
- Università Magna Graecia di Catanzaro, Dipartimento di Scienze della Salute, Campus Universitario, Catanzaro, Italy
| | - Carlo Federico Perno
- University of Rome "Tor Vergata," Department of Experimental Medicine and Surgery, Rome, Italy INMI L. Spallanzani, Rome, Italy
| | - Valentina Svicher
- University of Rome "Tor Vergata," Department of Experimental Medicine and Surgery, Rome, Italy
| |
Collapse
|
126
|
Jeong JJ, Kim DH. 5,7-Dihydroxy-6-Methoxy-Flavonoids Eliminate HIV-1 D3-transfected Cytoprotective Macrophages by Inhibiting the PI3K/Akt Signaling Pathway. Phytother Res 2015; 29:1355-1365. [DOI: 10.1002/ptr.5388] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 04/25/2015] [Accepted: 05/16/2015] [Indexed: 11/07/2022]
Affiliation(s)
- Jin-Ju Jeong
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, College of Pharmacy; Kyung Hee University; Seoul South Korea
| | - Dong-Hyun Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, College of Pharmacy; Kyung Hee University; Seoul South Korea
| |
Collapse
|
127
|
Rosiglitazone suppresses HIV-1 Tat-induced vascular inflammation via Akt signaling. Mol Cell Biochem 2015; 407:173-9. [PMID: 26048716 DOI: 10.1007/s11010-015-2467-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 05/29/2015] [Indexed: 01/20/2023]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARƔ) contributes to human immunodeficiency virus (HIV)-1-induced dysfunction of brain endothelial cells. The aim of the present study was to evaluate the protection mechanism of PPARƔ against Tat-induced responses of adhesion molecules. We measured the protein expressions of intercellular adhesion molecule (ICAM)-1 and vascular cell adhesion molecule (VCAM)-1 in human brain microvascular endothelial cells (hCMEC/D3) and C57BL/6J mouse brain microvessels with Western blotting and immunofluorescent labeling. The mRNA levels of ICAM-1 and VCAM-1 were determined by real-time reverse-transcriptase polymerase chain reaction. HIV-1 Tat induced overexpression of ICAM-1 but not VCAM-1 in both hCMEC/D3 and brain microvessels, this response was attenuated by treatment with the PPARƔ agonist rosiglitazone. Tat-mediated upregulation of ICAM-1 and VCAM-1 levels were abolished by the addition of PPARƔ antagonist GW9662 and the Akt inhibitor KP3721, indicating that Akt signaling is involved in the PPARƔ-mediated protection of Tat-induced adhesion molecule upregulation. These results show that Akt signaling plays a key role in PPARƔ's vascular inflammatory effects that contribute to blood-brain barrier damage.
Collapse
|
128
|
Abstract
HIV infects the central nervous system (CNS) during primary infection and persists in resident macrophages. CNS infection initiates a strong local immune response that fails to control the virus but is responsible for by-stander lesions involved in neurocognitive disorders. Although highly active anti-retroviral therapy now offers an almost complete control of CNS viral proliferation, low-grade CNS inflammation persists. This review focuses on HIV-induced intrathecal immunoglobulin (Ig) synthesis. Intrathecal Ig synthesis early occurs in more than three-quarters of patients in response to viral infection of the CNS and persists throughout the course of the disease. Viral antigens are targeted but this specific response accounts for <5% of the whole intrathecal synthesis. Although the nature and mechanisms leading to non-specific synthesis are unknown, this prominent proportion is comparable to that observed in various CNS viral infections. Cerebrospinal fluid-floating antibody-secreting cells account for a minority of the whole synthesis, which mainly takes place in perivascular inflammatory infiltrates of the CNS parenchyma. B-cell traffic and lineage across the blood-brain-barrier have not yet been described. We review common technical pitfalls and update the pending questions in the field. Moreover, since HIV infection is associated with an intrathecal chronic oligoclonal (and mostly non-specific) Ig synthesis and associates with low-grade axonal lesions, this could be an interesting model of the chronic intrathecal synthesis occurring during multiple sclerosis.
Collapse
|
129
|
Yang FC, Kuang WD, Li C, Sun WW, Qu D, Wang JH. Toll-Interacting Protein Suppresses HIV-1 Long-Terminal-Repeat-Driven Gene Expression and Silences the Post-Integrational Transcription of Viral Proviral DNA. PLoS One 2015; 10:e0125563. [PMID: 25915421 PMCID: PMC4411168 DOI: 10.1371/journal.pone.0125563] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/24/2015] [Indexed: 12/13/2022] Open
Abstract
Toll-interacting protein (Tollip) is a host adaptor protein for negatively regulating Toll-like receptor 2-, 4-, and IL-1R (interleukin-1 receptor)-mediated signaling. We found that Tollip expression could be induced in MDDCs (monocyte-derived dendritic cells) by HIV-1 particles and recombinant gp120 glycoprotein. Hence, we investigated the role of Tollip in modulating HIV-1 infection. We found that Tollip expression suppressed NF-κB-dependent HIV-1 long terminal repeat (LTR)-driven transcription and thus inhibited HIV-1 infection. Our protein truncation experiments proved that the intact C-terminus of Tollip was required for inhibition of both NF-κB activity and HIV-1 LTR-driven gene expression. Intriguingly, Tollip silenced the post-integrational transcription of HIV-1 proviral DNA, indicating the potential role of Tollip in maintaining viral persistence. Our results reveal the novel role of host factor Tollip in modulating HIV-1 infection, and may suggest the hijacking of Tollip as the negative regulator of the TLR pathway and even the downstream signaling, by HIV-1 for maintaining persistent infection. Further elucidation of the mechanisms by which HIV-1 induces Tollip expression and identification of the role of Tollip in modulating HIV-1 latency will facilitate the understanding of host regulation in viral replication and benefit the exploration of novel strategies for combating HIV-1 infection.
Collapse
Affiliation(s)
- Fu-Chun Yang
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Wen-Dong Kuang
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Chuan Li
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Wei-Wei Sun
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Di Qu
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Jian-Hua Wang
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
- * E-mail:
| |
Collapse
|
130
|
Brown A. Understanding the MIND phenotype: macrophage/microglia inflammation in neurocognitive disorders related to human immunodeficiency virus infection. Clin Transl Med 2015; 4:7. [PMID: 25852823 PMCID: PMC4385031 DOI: 10.1186/s40169-015-0049-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/27/2015] [Indexed: 02/07/2023] Open
Abstract
Tissue macrophages play important roles in maintaining homeostasis in most organs of the body including the brain where microglia represent the resident phagocytic cells of this compartment. The possibility of one day harnessing macrophage plasticity to treat or ameliorate disorders including obesity, cancer, organ damage, intestinal disorders, neurodegeneration, and cardiovascular disease in which these cells play a role, is a very exciting prospect. Inflammatory signaling is required for regenerative repair, healing, and pathogen clearance functions. However, when the inflammatory response persists in a chronic fashion over an extended period of time, damage to neurons is followed by neuronal injury and dysfunction. Macrophages in the brain are heterogeneous arising from tissues during embryogenesis, and in the adult, from bone marrow derived monocytes that enter through the blood-brain-barrier. While much of our insight regarding macrophage functional subtypes has been garnered through elegant studies in mice, which are amenable to genetic manipulation, far less is known about such cells in human tissues, and particularly in the brain under normal, disease, or injurious conditions. In this regard, non-human primate models for human immunodeficiency virus have been extremely useful for understanding the contribution of bone marrow-derived monocytes in neurological disease and their interaction and impact on the activation state of resident microglia in the brain. This review will focus on what has been learned from the rhesus macaque models about the types of macrophages present in the brains of animals with encephalitis. In vitro studies, which have used human blood monocytes differentiated into macrophages to address the question of macrophage subsets in HIV infection will be highlighted. Recent insights on macrophage phenotype and persistent inflammation in the brain in HIV-associated neurocognitive disorder from immunohistochemical studies on human autopsy tissue will be examined.
Collapse
Affiliation(s)
- Amanda Brown
- Johns Hopkins University School of Medicine, 600 North Wolfe Street/Meyer 6-181, Baltimore, MD 21287 USA
| |
Collapse
|
131
|
Carvallo L, Lopez L, Che FY, Lim J, Eugenin EA, Williams DW, Nieves E, Calderon TM, Madrid-Aliste C, Fiser A, Weiss L, Angeletti RH, Berman JW. Buprenorphine decreases the CCL2-mediated chemotactic response of monocytes. THE JOURNAL OF IMMUNOLOGY 2015; 194:3246-58. [PMID: 25716997 DOI: 10.4049/jimmunol.1302647] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Despite successful combined antiretroviral therapy, ∼ 60% of HIV-infected people exhibit HIV-associated neurocognitive disorders (HAND). CCL2 is elevated in the CNS of infected people with HAND and mediates monocyte influx into the CNS, which is critical in neuroAIDS. Many HIV-infected opiate abusers have increased neuroinflammation that may augment HAND. Buprenorphine is used to treat opiate addiction. However, there are few studies that examine its impact on HIV neuropathogenesis. We show that buprenorphine reduces the chemotactic phenotype of monocytes. Buprenorphine decreases the formation of membrane projections in response to CCL2. It also decreases CCL2-induced chemotaxis and mediates a delay in reinsertion of the CCL2 receptor, CCR2, into the cell membrane after CCL2-mediated receptor internalization, suggesting a mechanism of action of buprenorphine. Signaling pathways in CCL2-induced migration include increased phosphorylation of p38 MAPK and of the junctional protein JAM-A. We show that buprenorphine decreases these phosphorylations in CCL2-treated monocytes. Using DAMGO, CTAP, and Nor-BNI, we demonstrate that the effect of buprenorphine on CCL2 signaling is opioid receptor mediated. To identify additional potential mechanisms by which buprenorphine inhibits CCL2-induced monocyte migration, we performed proteomic analyses to characterize additional proteins in monocytes whose phosphorylation after CCL2 treatment was inhibited by buprenorphine. Leukosialin and S100A9 were identified and had not been shown previously to be involved in monocyte migration. We propose that buprenorphine limits CCL2-mediated monocyte transmigration into the CNS, thereby reducing neuroinflammation characteristic of HAND. Our findings underscore the use of buprenorphine as a therapeutic for neuroinflammation as well as for addiction.
Collapse
Affiliation(s)
- Loreto Carvallo
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Lillie Lopez
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Fa-Yun Che
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Jihyeon Lim
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Eliseo A Eugenin
- Public Health Research Institute, Newark, NJ 07103; Department of Microbiology and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers The State University of New Jersey, Newark, NJ 07103
| | - Dionna W Williams
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Edward Nieves
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Tina M Calderon
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Carlos Madrid-Aliste
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY 10461; and
| | - Andras Fiser
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY 10461; and
| | - Louis Weiss
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Ruth Hogue Angeletti
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Joan W Berman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461; Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
132
|
Costiniuk CT, Jenabian MA. HIV reservoir dynamics in the face of highly active antiretroviral therapy. AIDS Patient Care STDS 2015; 29:55-68. [PMID: 25412339 DOI: 10.1089/apc.2014.0173] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Upon discontinuation of highly active antiretroviral therapy (HAART), human immunodeficiency virus (HIV)-infected individuals experience a brisk rebound in blood plasma viremia due to the exodus of HIV from various body reservoirs. Assessment of HIV dynamics during HAART and following treatment discontinuation is essential to better understand HIV persistence. Here we will first provide a brief overview of the molecular mechanisms involved in HIV reservoir formation and persistence. After a summary of HAART-mediated HIV decay within peripheral blood, we discuss findings from clinical studies examining the effects of HAART initiation and interruption on HIV reservoir dynamics in major anatomical compartments, including lymph nodes and spleen, gut associated lymphoid tissue, reproductive organs, the central nervous system, and the lungs. Features contributing to these reservoirs as distinct compartments, including anatomical features, the presence of drug transporters, and the effect of co-infection, are also discussed.
Collapse
Affiliation(s)
- Cecilia T. Costiniuk
- Department of Medicine, Divisions of Infectious Diseases/Chronic Viral Illness Service and Lachine Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - Mohammad-Ali Jenabian
- Département des Sciences Biologiques et Centre de recherche BioMed, Université du Québec à Montréal (UQAM), Montreal, Quebec, Canada
| |
Collapse
|
133
|
Silva K, Hope-Lucas C, White T, Hairston TK, Rameau T, Brown A. Cortical neurons are a prominent source of the proinflammatory cytokine osteopontin in HIV-associated neurocognitive disorders. J Neurovirol 2015; 21:174-85. [PMID: 25636782 PMCID: PMC4372685 DOI: 10.1007/s13365-015-0317-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/30/2014] [Accepted: 01/09/2015] [Indexed: 12/13/2022]
Abstract
The proinflammatory cytokine osteopontin (OPN) is elevated in the cerebrospinal fluid (CSF) in individuals with HIV-associated neurocognitive disorders (HAND) and remains so in those on suppressive antiretroviral therapy. To understand the pathophysiological significance of elevated OPN in the CNS, we sought to determine the cellular source of this cytokine. As HIV-1 replicates productively in macrophages/microglia, we tested whether these cells are the predominant producers of OPN in the brain. Stringent patient selection criteria, which excluded brain tissues from those with evidence of drug abuse and dependence, were used. Uninfected normal controls, amyotrophic lateral sclerosis (ALS), HIV+ asymptomatic neurocognitive impairment (ANI), and HIV+ mild neurocognitive disorder (MND)/HIV-associated dementia (HAD) groups were included. Double-label immunohistochemistry for CNS cells and OPN was used to quantify OPN expression in astrocytes, macrophages/microglia, and neurons. While resident macrophages/microglia expressed OPN, astrocytes and unexpectedly neurons were also a major source of OPN. OPN levels in ionized Ca(2+)-binding adapter 1 (Iba1)/allograft inflammatory factor-1 (AIF-1)+ microglia in HIV+ ANI and MND/HAD exceeded those of HIV-negative controls and were comparable to expression seen in ALS. Moreover, in neurons, OPN was expressed at the highest levels in the HIV+ ANI group. These findings suggest that while infiltrating HIV-infected macrophages are most likely the initial source of OPN, resident CNS cells become activated and also express this inflammatory cytokine at significant levels. Moreover, as OPN levels are elevated compared to uninfected individuals and increases with the severity of impairment, it appears that the expression of OPN is persistent and sustained within the brain parenchyma in those that progress to HAND.
Collapse
Affiliation(s)
- Katie Silva
- Department of Neurology, Johns Hopkins University School of Medicine, 600 North Wolfe Street/Meyer 6-181, Baltimore, MD, 21287-7131, USA
| | | | | | | | | | | |
Collapse
|
134
|
Sabbatucci M, Covino DA, Purificato C, Mallano A, Federico M, Lu J, Rinaldi AO, Pellegrini M, Bona R, Michelini Z, Cara A, Vella S, Gessani S, Andreotti M, Fantuzzi L. Endogenous CCL2 neutralization restricts HIV-1 replication in primary human macrophages by inhibiting viral DNA accumulation. Retrovirology 2015; 12:4. [PMID: 25608886 PMCID: PMC4314729 DOI: 10.1186/s12977-014-0132-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 12/19/2014] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Macrophages are key targets of HIV-1 infection. We have previously described that the expression of CC chemokine ligand 2 (CCL2) increases during monocyte differentiation to macrophages and it is further up-modulated by HIV-1 exposure. Moreover, CCL2 acts as an autocrine factor that promotes viral replication in infected macrophages. In this study, we dissected the molecular mechanisms by which CCL2 neutralization inhibits HIV-1 replication in monocyte-derived macrophages (MDM), and the potential involvement of the innate restriction factors protein sterile alpha motif (SAM) histidine/aspartic acid (HD) domain containing 1 (SAMHD1) and apolipoprotein B mRNA-editing, enzyme-catalytic, polypeptide-like 3 (APOBEC3) family members. RESULTS CCL2 neutralization potently reduced the number of p24 Gag+ cells during the course of either productive or single cycle infection with HIV-1. In contrast, CCL2 blocking did not modify entry of HIV-1 based Virus Like Particles, thus demonstrating that the restriction involves post-entry steps of the viral life cycle. Notably, the accumulation of viral DNA, both total, integrated and 2-LTR circles, was strongly impaired by neutralization of CCL2. Looking for correlates of HIV-1 DNA accumulation inhibition, we found that the antiviral effect of CCL2 neutralization was independent of the modulation of SAMHD1 expression or function. Conversely, a strong and selective induction of APOBEC3A expression, to levels comparable to those of freshly isolated monocytes, was associated with the inhibition of HIV-1 replication mediated by CCL2 blocking. Interestingly, the CCL2 neutralization mediated increase of APOBEC3A expression was type I IFN independent. Moreover, the transcriptome analysis of the effect of CCL2 blocking on global gene expression revealed that the neutralization of this chemokine resulted in the upmodulation of additional genes involved in the defence response to viruses. CONCLUSIONS Neutralization of endogenous CCL2 determines a profound restriction of HIV-1 replication in primary MDM affecting post-entry steps of the viral life cycle with a mechanism independent of SAMHD1. In addition, CCL2 blocking is associated with induction of APOBEC3A expression, thus unravelling a novel mechanism which might contribute to regulate the expression of innate intracellular viral antagonists in vivo. Thus, our study may potentially lead to the development of new therapeutic strategies for enhancing innate cellular defences against HIV-1 and protecting macrophages from infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Laura Fantuzzi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
135
|
Ragin AB, Wu Y, Gao Y, Keating S, Du H, Sammet C, Kettering CS, Epstein LG. Brain alterations within the first 100 days of HIV infection. Ann Clin Transl Neurol 2014; 2:12-21. [PMID: 25642430 PMCID: PMC4301670 DOI: 10.1002/acn3.136] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 09/27/2014] [Accepted: 09/29/2014] [Indexed: 01/12/2023] Open
Abstract
Objective Brain involvement is a serious complication of HIV infection. The earliest changes in the brain, which represents an anatomic site for viral persistence, are largely unknown. Methods This investigation used quantitative Magnetic Resonance methodologies, including high resolution and diffusion tensor (DTI) imaging, to evaluate the brain in 15 HIV and 20 seronegative subjects. All HIV subjects were antibody nonreactive with assay-estimated infection duration of less than 100 days. Results Brain volumetric analysis revealed reduced parenchyma with enlargement of the third ventricle and brainstem. DTI quantified loss of white matter integrity in the corpus callosum and diffusion alterations in caudate. Cognitive differences were indicated in psychomotor speed and visual recall. There were no differences between antiretroviral-initiated and naïve HIV subgroups. Interpretation These findings, quantified within 100 days of infection, shed light on the earliest brain changes in HIV infection. Onset of neural injury may date to initial viral invasion and the transient early period of unchecked viremia and marked immunosuppression of the seroconversion period.
Collapse
Affiliation(s)
- Ann B Ragin
- Department of Radiology, Feinberg School of Medicine, Northwestern University Chicago, Illinois
| | - Ying Wu
- Center for Advanced Imaging, NorthShore University Hospital Evanston, Illinois
| | - Yi Gao
- Department of Statistics, Northwestern University Evanston, Illinois
| | - Sheila Keating
- Blood Systems Research Institute San Francisco, California
| | - Hongyan Du
- Clinical & Research Informatics, NorthShore University Hospital Evanston, Illinois
| | - Christina Sammet
- Department of Radiology, Feinberg School of Medicine, Northwestern University Chicago, Illinois ; Department of Medical Imaging, Ann and Robert H. Lurie Children's Hospital of Chicago Chicago, Illinois
| | - Casey S Kettering
- Department of Radiology, Feinberg School of Medicine, Northwestern University Chicago, Illinois
| | - Leon G Epstein
- Department of Neurology, Ann and Robert H. Lurie Children's Hospital of Chicago Chicago, Illinois ; Department of Pediatrics & Neurology, Feinberg School of Medicine, Northwestern University Chicago, Illinois
| |
Collapse
|
136
|
HIV Tat acts on endogenous retroviruses of the W family and this occurs via Toll-like receptor 4: inference for neuroAIDS. AIDS 2014; 28:2659-70. [PMID: 25250834 DOI: 10.1097/qad.0000000000000477] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The objective of this study is to verify whether HIV activates two endogenous retroviruses of the human endogenous retrovirus (HERV)-W family, multiple sclerosis-associated retrovirus (MSRV) and Syncytin-1, whose neuropathogenic and immunopathogenic properties could contribute to HIV-related neurodegeneration. DESIGN AND METHODS Peripheral blood mononuclear cells, monocyte-macrophages and astrocytes were either infected by HIV or exposed to HIV-Tat, and/or other treatments. The expression of transcripts and proteins of interest was evaluated by real-time RT-PCR and western blotting assays, respectively. RESULTS HIV and Tat increase the levels of MSRVenv mRNAs and HERV-Wenv proteins in astrocytes and in blood cells. In monocyte-macrophages, Tat also induces high levels of CCR2, CD16 and Toll-like receptor 4 (TLR4) molecules. Syncytin-1 response to Tat depends on the cell context: in monocytes, Tat stimulates MSRVenv and inhibits Syncytin-1, while in differentiated macrophages, it stimulates both elements. In primary astrocytes, Tat stimulates MSRV and Syncytin-1 indirectly, through interaction with TLR4 and induction of tumour necrosis factor-alpha (TNFα), without internalization. CONCLUSION In-vivo consequence of the study could be that, through increase of CD16 and CCR2, Tat promotes neuroinvasion not only by HIV-infected monocytes/macrophages but also by the HERV-Ws, with their neuropathogenic potential. Also, the novel finding of TLR4 stimulation by Tat may be of relevance, as TLR4 is critical in neuroinflammation. Within central nervous system (CNS), Tat-induced TNFα could induce high levels of the HERV-Ws, in both macrophages and astrocytes, also without HIV replication. The indirect mechanism by which Tat activates the HERV-Ws through induction of TNFα could add a new piece to the puzzle of CNS pathogenesis, that is the HERV-Wenv contribute to the HIV-related neurodegeneration.
Collapse
|
137
|
Calantone N, Wu F, Klase Z, Deleage C, Perkins M, Matsuda K, Thompson EA, Ortiz AM, Vinton CL, Ourmanov I, Loré K, Douek DC, Estes JD, Hirsch VM, Brenchley JM. Tissue myeloid cells in SIV-infected primates acquire viral DNA through phagocytosis of infected T cells. Immunity 2014; 41:493-502. [PMID: 25238099 DOI: 10.1016/j.immuni.2014.08.014] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 08/25/2014] [Indexed: 10/24/2022]
Abstract
The viral accessory protein Vpx, expressed by certain simian and human immunodeficiency viruses (SIVs and HIVs), is thought to improve viral infectivity of myeloid cells. We infected 35 Asian macaques and African green monkeys with viruses that do or do not express Vpx and examined viral targeting of cells in vivo. While lack of Vpx expression affected viral dynamics in vivo, with decreased viral loads and infection of CD4⁺ T cells, Vpx expression had no detectable effect on infectivity of myeloid cells. Moreover, viral DNA was observed only within myeloid cells in tissues not massively depleted of CD4⁺ T cells. Myeloid cells containing viral DNA also showed evidence of T cell phagocytosis in vivo, suggesting that their viral DNA may be attributed to phagocytosis of SIV-infected T cells. These data suggest that myeloid cells are not a major source of SIV in vivo, irrespective of Vpx expression.
Collapse
Affiliation(s)
- Nina Calantone
- Lab of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Fan Wu
- Lab of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Zachary Klase
- Lab of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Molly Perkins
- Lab of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Kenta Matsuda
- Lab of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Elizabeth A Thompson
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA; Department of Medicine, Karolinska Institutet, Stockholm 171, Sweden
| | | | - Carol L Vinton
- Lab of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Ilnour Ourmanov
- Lab of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Karin Loré
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA; Department of Medicine, Karolinska Institutet, Stockholm 171, Sweden
| | - Daniel C Douek
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Vanessa M Hirsch
- Lab of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | | |
Collapse
|
138
|
Chauhan A, Khandkar M. Endocytosis of human immunodeficiency virus 1 (HIV-1) in astrocytes: a fiery path to its destination. Microb Pathog 2014; 78:1-6. [PMID: 25448132 DOI: 10.1016/j.micpath.2014.11.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/24/2014] [Accepted: 11/03/2014] [Indexed: 11/27/2022]
Abstract
Despite successful suppression of peripheral HIV-1 infection by combination antiretroviral therapy, immune activation by residual virus in the brain leads to HIV-associated neurocognitive disorders (HAND). In the brain, several types of cells, including microglia, perivascular macrophage, and astrocytes have been reported to be infected by HIV-1. Astrocytes, the most abundant cells in the brain, maintain homeostasis. The general consensus on HIV-1 infection in astrocytes is that it produces unproductive viral infection. HIV-1 enters astrocytes by pH-dependent endocytosis, leading to degradation of the virus in endosomes, but barely succeeds in infection. Here, we have discussed endocytosis-mediated HIV-1 entry and viral programming in astrocytes.
Collapse
Affiliation(s)
- Ashok Chauhan
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, 6439 Garners Ferry Road, Columbia, SC 29209, USA.
| | - Mehrab Khandkar
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, 6439 Garners Ferry Road, Columbia, SC 29209, USA
| |
Collapse
|
139
|
Archin NM, Sung JM, Garrido C, Soriano-Sarabia N, Margolis DM. Eradicating HIV-1 infection: seeking to clear a persistent pathogen. Nat Rev Microbiol 2014; 12:750-64. [PMID: 25402363 PMCID: PMC4383747 DOI: 10.1038/nrmicro3352] [Citation(s) in RCA: 219] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Effective antiretroviral therapy (ART) blunts viraemia, which enables HIV-1-infected individuals to control infection and live long, productive lives. However, HIV-1 infection remains incurable owing to the persistence of a viral reservoir that harbours integrated provirus within host cellular DNA. This latent infection is unaffected by ART and hidden from the immune system. Recent studies have focused on the development of therapies to disrupt latency. These efforts unmasked residual viral genomes and highlighted the need to enable the clearance of latently infected cells, perhaps via old and new strategies that improve the HIV-1-specific immune response. In this Review, we explore new approaches to eradicate established HIV-1 infection and avoid the burden of lifelong ART.
Collapse
Affiliation(s)
- Nancie M Archin
- Department of Medicine, University of North Carolina at Chapel Hill
| | - Julia Marsh Sung
- Department of Medicine, University of North Carolina at Chapel Hill
| | - Carolina Garrido
- Department of Medicine, University of North Carolina at Chapel Hill
| | | | - David M Margolis
- 1] Department of Medicine, University of North Carolina at Chapel Hill. [2] Department of Microbiology and Immunology, University of North Carolina at Chapel Hill. [3] Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
140
|
Gaskill PJ, Yano HH, Kalpana GV, Javitch JA, Berman JW. Dopamine receptor activation increases HIV entry into primary human macrophages. PLoS One 2014; 9:e108232. [PMID: 25268786 PMCID: PMC4182469 DOI: 10.1371/journal.pone.0108232] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 08/25/2014] [Indexed: 01/11/2023] Open
Abstract
Macrophages are the primary cell type infected with HIV in the central nervous system, and infection of these cells is a major component in the development of neuropathogenesis and HIV-associated neurocognitive disorders. Within the brains of drug abusers, macrophages are exposed to increased levels of dopamine, a neurotransmitter that mediates the addictive and reinforcing effects of drugs of abuse such as cocaine and methamphetamine. In this study we examined the effects of dopamine on HIV entry into primary human macrophages. Exposure to dopamine during infection increased the entry of R5 tropic HIV into macrophages, irrespective of the concentration of the viral inoculum. The entry pathway affected was CCR5 dependent, as antagonizing CCR5 with the small molecule inhibitor TAK779 completely blocked entry. The effect was dose-dependent and had a steep threshold, only occurring above 108 M dopamine. The dopamine-mediated increase in entry required dopamine receptor activation, as it was abrogated by the pan-dopamine receptor antagonist flupenthixol, and could be mediated through both subtypes of dopamine receptors. These findings indicate that the effects of dopamine on macrophages may have a significant impact on HIV pathogenesis. They also suggest that drug-induced increases in CNS dopamine may be a common mechanism by which drugs of abuse with distinct modes of action exacerbate neuroinflammation and contribute to HIV-associated neurocognitive disorders in infected drug abusers.
Collapse
Affiliation(s)
- Peter J. Gaskill
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| | - Hideaki H. Yano
- Department of Psychiatry and Pharmacology, Columbia University, New York, New York, United States of America
| | - Ganjam V. Kalpana
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Jonathan A. Javitch
- Department of Psychiatry and Pharmacology, Columbia University, New York, New York, United States of America
| | - Joan W. Berman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
141
|
Jadhav VS, Krause KH, Singh SK. HIV-1 Tat C modulates NOX2 and NOX4 expressions through miR-17 in a human microglial cell line. J Neurochem 2014; 131:803-15. [PMID: 25146963 DOI: 10.1111/jnc.12933] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/12/2014] [Accepted: 08/14/2014] [Indexed: 12/22/2022]
Abstract
HIV-1 invades CNS in the early course of infection, which can lead to the cascade of neuroinflammation. NADPH oxidases (NOXs) are the major producers of reactive oxygen species (ROS), which play important roles during pathogenic insults. The molecular mechanism of ROS generation via microRNA-mediated pathway in human microglial cells in response to HIV-1 Tat protein has been demonstrated in this study. Over-expression and knockdown of microRNAs, luciferase reporter assay, and site-directed mutagenesis are main molecular techniques used in this study. A significant reduction in miR-17 levels and increased NOX2, NOX4 expression levels along with ROS production were observed in human microglial cells upon HIV-1 Tat C exposure. The validation of NOX2 and NOX4 as direct targets of miR-17 was done by luciferase reporter assay. The over-expression and knockdown of miR-17 in human microglial cells showed the direct role of miR-17 in regulation of NOX2, NOX4 expression and intracellular ROS generation. We demonstrated the regulatory role of cellular miR-17 in ROS generation through over-expression and knockdown of miR-17 in human microglial cells exposed to HIV-1 Tat C protein. Activated microglial cells mediated neuroinflammatory events are observed in HIV-associated neurological disorders. The reduction in miR-17 levels was observed in microglial cells exposed to HIV-1 Tat C protein. miR-17 regulated the expression of NOX2 and NOX4, which in turn regulated the reactive oxygen species (ROS) production in microglial cells. Increased ROS production led to the activation of microglial cells and increased cytokine production. This study thus demonstrated a novel miR-17-mediated regulatory pathway of ROS production in microglial cells. HMC3 = human microglia clone 3 cell lines.
Collapse
Affiliation(s)
- Vaishnavi Sunil Jadhav
- Laboratory of Neurovirology and Inflammation Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad, India
| | | | | |
Collapse
|
142
|
Treatment intensification with maraviroc (CCR5 antagonist) leads to declines in CD16-expressing monocytes in cART-suppressed chronic HIV-infected subjects and is associated with improvements in neurocognitive test performance: implications for HIV-associated neurocognitive disease (HAND). J Neurovirol 2014; 20:571-82. [PMID: 25227930 DOI: 10.1007/s13365-014-0279-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 08/05/2014] [Accepted: 08/06/2014] [Indexed: 01/21/2023]
Abstract
HIV-associated neurocognitive disorders (HAND) continues to be prevalent (30-50%) despite plasma HIV-RNA suppression with combination antiretroviral therapy (cART). There is no proven therapy for individuals on suppressive cART with HAND. We have shown that the degree of HIV reservoir burden (HIV DNA) in monocytes appear to be linked to cognitive outcomes. HIV infection of monocytes may therefore be critical in the pathogenesis of HAND. A single arm, open-labeled trial was conducted to examine the effect of maraviroc (MVC) intensification on monocyte inflammation and neuropsychological (NP) performance in 15 HIV subjects on stable 6-month cART with undetectable plasma HIV RNA (<48 copies/ml) and detectable monocyte HIV DNA (>10 copies/10(6) cells). MVC was added to their existing cART regimen for 24 weeks. Post-intensification change in monocytes was assessed using multiparametric flow cytometry, monocyte HIV DNA content by PCR, soluble CD163 (sCD163) by an ELISA, and NP performance over 24 weeks. In 12 evaluable subjects, MVC intensification resulted in a decreased proportion of circulating intermediate (median; 3.06% (1.93, 6.45) to 1.05% (0.77, 2.26)) and nonclassical (5.2% (3.8, 7.9) to 3.2% (1.8, 4.8)) CD16-expressing monocytes, a reduction in monocyte HIV DNA content to zero log10 copies/10(6) cells and in levels of sCD163 of 43% by 24 weeks. This was associated with significant improvement in NP performance among six subjects who entered the study with evidence of mild to moderate cognitive impairment. The results of this study suggest that antiretroviral therapy with potency against monocytes may have efficacy against HAND.
Collapse
|
143
|
Chauhan A, Tikoo A, Patel J, Abdullah AM. HIV-1 endocytosis in astrocytes: a kiss of death or survival of the fittest? Neurosci Res 2014; 88:16-22. [PMID: 25219546 DOI: 10.1016/j.neures.2014.08.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 08/14/2014] [Accepted: 08/18/2014] [Indexed: 12/23/2022]
Abstract
The brain is a target of HIV-1 and serves as an important viral reservoir. Astrocytes, the most abundant glial cell in the human brain, are involved in brain plasticity and neuroprotection. Several studies have reported HIV-1 infection of astrocytes in cell cultures and infected brain tissues. The prevailing concept is that HIV-1 infection of astrocytes leads to latent infection. Here, we provide our perspective on endocytosis-mediated HIV-1 entry and its fate in astrocytes. Natural entry of HIV-1 into astrocytes occurs via endocytosis. However, endocytosis of HIV-1 in astrocytes is a natural death trap where the majority of virus particles are degraded in endosomes and a few which escape intact lead to successful infection. Thus, regardless of artificial fine-tuning (treatment with cytokines or proinflammatory products) done to astrocytes, HIV-1 does not infect them efficiently unless the viral entry route or the endosomal enzymatic machinery has been manipulated.
Collapse
Affiliation(s)
- Ashok Chauhan
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, United States; Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29209, United States.
| | - Akshay Tikoo
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, United States
| | - Jankiben Patel
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, United States
| | - Arwa Mujahid Abdullah
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, United States
| |
Collapse
|
144
|
Glisoni RJ, Sosnik A. Novel Poly(Ethylene Oxide)-b-Poly(Propylene Oxide) Copolymer-Glucose Conjugate by the Microwave-Assisted Ring Opening of a Sugar Lactone. Macromol Biosci 2014; 14:1639-51. [DOI: 10.1002/mabi.201400235] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 07/30/2014] [Indexed: 12/30/2022]
Affiliation(s)
- Romina J. Glisoni
- The Group of Biomaterials and Nanotechnology for Improved Medicines (BIONIMED), Department of Pharmaceutical Technology, Faculty of Pharmacy and Biochemistry; University of Buenos Aires; Buenos Aires CP1113 Argentina
- National Science Research Council (CONICET); Buenos Aires Argentina
| | - Alejandro Sosnik
- Group of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering; Technion-Israel Institute of Technology; Technion City 32000 Haifa Israel
- Department of Materials Science and Engineering, De-Jur Building, Office 607; Technion-Israel Institute of Technology; Technion City 32000 Haifa Israel
| |
Collapse
|
145
|
Evering TH, Kamau E, St Bernard L, Farmer CB, Kong XP, Markowitz M. Single genome analysis reveals genetic characteristics of Neuroadaptation across HIV-1 envelope. Retrovirology 2014; 11:65. [PMID: 25125210 PMCID: PMC4145222 DOI: 10.1186/s12977-014-0065-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 07/24/2014] [Indexed: 01/25/2023] Open
Abstract
Background The widespread use of highly effective, combination antiretroviral therapy (cART) has led to a significant reduction in the incidence of HIV-associated dementia (HAD). Despite these advances, the prevalence of HIV-1 associated neurocognitive disorders (HANDs) has been estimated at approximately 40%-50%. In the cART era, the majority of this disease burden is represented by asymptomatic neurocognitive impairment and mild neurocognitive disorder (ANI and MND respectively). Although less severe than HAD, these diagnoses carry with them substantial morbidity. Results In this cross-sectional study, single genome amplification (SGA) was used to sequence 717 full-length HIV-1 envelope (env) clade B variants from the paired cerebrospinal fluid (CSF) and blood plasma samples of fifteen chronically infected HIV-positive individuals with normal neurocognitive performance (NCN), ANI and MND. Various degrees of compartmentalization were found across disease states and history of cART utilization. In individuals with compartmentalized virus, mean HIV-1 env population diversity was lower in the CSF than plasma-derived variants. Overall, mean V1V2 loop length was shorter in CSF-derived quasispecies when compared to contemporaneous plasma populations, and this was found to correlate with a lower mean number of N-linked glycosylation sites in this region. A number of discrete amino acid positions that correlate strongly with compartmentalization in the CSF were identified in both variable and constant regions of gp120 as well as in gp41. Correlated mutation analyses further identified that a subset of amino acid residues in these compartmentalization “hot spot” positions were strongly correlated with one another, suggesting they may play an important, definable role in the adaptation of viral variants to the CSF. Analysis of these hot spots in the context of a well-supported crystal structure of HIV-1 gp120 suggests mechanisms through which amino acid differences at the identified residues might contribute to viral compartmentalization in the CSF. Conclusions The detailed analyses of SGA-derived full length HIV-1 env from subjects with both normal neurocognitive performance and the most common HAND diagnoses in the cART era allow us to identify novel and confirm previously described HIV-1 env genetic determinants of neuroadaptation and relate potential motifs to HIV-1 env structure and function. Electronic supplementary material The online version of this article (doi:10.1186/s12977-014-0065-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Teresa H Evering
- Aaron Diamond AIDS Research Center, an affiliate of the Rockefeller University, New York, USA.
| | | | | | | | | | | |
Collapse
|
146
|
Abstract
Monocytes are blood-derived mononuclear phagocytic cells that traffic throughout the body and can provide rapid innate immune effector responses in response to microbial pathogen infections. Among blood monocytes, the most abundant subset in mice is represented by inflammatory Ly6C(+) CCR2(+) monocytes and is the functional equivalent of the CD14(+) monocytes in humans. Herein we focus on published evidence describing the exquisite functional plasticity of these cells, and we extend this overview to their multiples roles in vivo during host immune defenses against microbial pathogen infections, as antigen-presenting cells, inflammatory cells or Trojan horse cells.
Collapse
|
147
|
Maung R, Hoefer MM, Sanchez AB, Sejbuk NE, Medders KE, Desai MK, Catalan IC, Dowling CC, de Rozieres CM, Garden GA, Russo R, Roberts AJ, Williams R, Kaul M. CCR5 knockout prevents neuronal injury and behavioral impairment induced in a transgenic mouse model by a CXCR4-using HIV-1 glycoprotein 120. THE JOURNAL OF IMMUNOLOGY 2014; 193:1895-910. [PMID: 25031461 DOI: 10.4049/jimmunol.1302915] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The innate immune system has been implicated in several neurodegenerative diseases, including HIV-1-associated dementia. In this study, we show that genetic ablation of CCR5 prevents microglial activation and neuronal damage in a transgenic model of HIV-associated brain injury induced by a CXCR4-using viral envelope gp120. The CCR5 knockout (KO) also rescues spatial learning and memory in gp120-transgenic mice. However, the CCR5KO does not abrogate astrocytosis, indicating it can occur independently from neuronal injury and behavioral impairment. To characterize further the neuroprotective effect of CCR5 deficiency we performed a genome-wide gene expression analysis of brains from HIVgp120tg mice expressing or lacking CCR5 and nontransgenic controls. A comparison with a human brain microarray study reveals that brains of HIVgp120tg mice and HIV patients with neurocognitive impairment share numerous differentially regulated genes. Furthermore, brains of CCR5 wild-type and CCR5KO gp120tg mice express markers of an innate immune response. One of the most significantly upregulated factors is the acute phase protein lipocalin-2 (LCN2). Using cerebrocortical cell cultures, we find that LCN2 is neurotoxic in a CCR5-dependent fashion, whereas inhibition of CCR5 alone is not sufficient to abrogate neurotoxicity of a CXCR4-using gp120. However, the combination of pharmacologic CCR5 blockade and LCN2 protects neurons from toxicity of a CXCR4-using gp120, thus recapitulating the finding in CCR5-deficient gp120tg mouse brain. Our study provides evidence for an indirect pathologic role of CCR5 and a novel protective effect of LCN2 in combination with inhibition of CCR5 in HIV-associated brain injury.
Collapse
Affiliation(s)
- Ricky Maung
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Melanie M Hoefer
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Ana B Sanchez
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Natalia E Sejbuk
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Kathryn E Medders
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037; Neuroscience, Aging and Stem Cell Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Maya K Desai
- Neuroscience, Aging and Stem Cell Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Irene C Catalan
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Cari C Dowling
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Cyrus M de Rozieres
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Gwenn A Garden
- Department of Neurology, University of Washington, Seattle, WA 98195
| | - Rossella Russo
- Neuroscience, Aging and Stem Cell Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037; Department of Pharmacobiology, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Amanda J Roberts
- Molecular and Cellular Neurosciences Department, The Scripps Research Institute, La Jolla, CA 92037
| | - Roy Williams
- Bioinformatics Shared Resource, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037; and
| | - Marcus Kaul
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037; Neuroscience, Aging and Stem Cell Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037; Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
148
|
Jeong JJ, Kim B, Kim DH. Ginsenoside Rb1 eliminates HIV-1 (D3)-transduced cytoprotective human macrophages by inhibiting the AKT pathway. J Med Food 2014; 17:849-54. [PMID: 24983400 DOI: 10.1089/jmf.2013.3020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Abstract Acquired immunodeficiency syndrome patients treated with red ginseng, which contains protopanxadiol and protopanaxatriol ginsenosides as its main constituents, have been reported to remain healthy for >20 years in the absence of highly active antiretroviral therapy. Of these ginsenosides, ginsenoside Rh1, a protopanaxatriol ginsenoside, is known to eliminate cytoprotective HIV-1-infected macrophages by inhibiting pyruvate dehydrogenase lipoamide kinase isozyme 1 (PDK-1) phosphorylation. In this study, we investigated the capacity of ginsenoside Rb1, a protopanaxadiol ginsenoside, to eliminate cytoprotective primary human macrophages. We found that ginsenoside Rb1 could also eliminate cytoprotective primary human macrophages infected with HIV-1 D3. Ginsenoside Rb1 inhibited lipopolysaccharide/cycloheximide-induced AKT and glycogen synthase kinase-3β phosphorylation in the D3-transduced macrophages, but not the phosphorylation of PDK-1 and phosphoinositide-3-kinase (PI3K). Furthermore, we also observed that a combined treatment with ginsenoside Rb1 and miltefosine synergistically abolished the cytoprotective CHME5 cells expressing HIV-1 tat. Based on these findings, we can conclude that ginsenoside Rb1 can eliminate cytoprotective macrophages infected with HIV-1 by inhibiting the AKT pathway.
Collapse
Affiliation(s)
- Jin-Ju Jeong
- 1 Department of Life and Nanopharmaceutical Sciences, Kyung Hee University , Seoul, South Korea
| | | | | |
Collapse
|
149
|
Mishra R, Singh SK. HIV-1 Tat C phosphorylates VE-cadherin complex and increases human brain microvascular endothelial cell permeability. BMC Neurosci 2014; 15:80. [PMID: 24965120 PMCID: PMC4230799 DOI: 10.1186/1471-2202-15-80] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 06/18/2014] [Indexed: 11/10/2022] Open
Abstract
Background Human brain microvascular endothelial cells (hBMVECs) are integral part of the blood brain barrier. Post-translational modifications of adherens junction proteins regulate the permeability of human brain microvascular endothelial cells. Pro-inflammatory signals can induce tyrosine phosphorylation of adherens junction proteins. The primary objective of this work is to provide a molecular model; how the HIV-1 Tat protein can compromise the BBB integrity and eventually lead to neurological consequences. We exposed hBMVECs to recombinant HIV-1 clade C Tat protein to study the effect of HIV-1 Tat C on permeability of hBMVECs. Trans-endothelial electrical resistance and fluorescent dye migration assay have been used to check the permeability of hBMVECs. DCFDA staining has been used for intracellular reactive oxygen species (ROS) detection. Western blotting has been used to study the expression levels and co-immunoprecipitation has been used to study the interactions among adherens junction proteins. Results HIV-1 Tat C protein induced NOX2 and NOX4 expression level and increased intracellular ROS level. Redox-sensitive kinase; PYK2 activation led to increased tyrosine phosphorylation of VE-cadherin and β-catenin, leading to disruption of junctional assembly. The dissociation of tyrosine phosphatases VE-PTP and SHP2 from cadherin complex resulted into increased tyrosine phosphorylation of VE-cadherin and β-catenin in HIV-1 Tat C treated hBMVECs. Conclusion Unrestricted phosphorylation of junctional proteins in hBMVECs, in response to HIV-1 Tat C protein; leads to the disruption of junctional complexes and increased endothelial permeability.
Collapse
Affiliation(s)
| | - Sunit Kumar Singh
- Laboratory of Neurovirology and Inflammation Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500007, India.
| |
Collapse
|
150
|
Endothelin-1 and its role in the pathogenesis of infectious diseases. Life Sci 2014; 118:110-9. [PMID: 24780317 DOI: 10.1016/j.lfs.2014.04.021] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 04/08/2014] [Accepted: 04/15/2014] [Indexed: 12/12/2022]
Abstract
Endothelins are potent regulators of vascular tone, which also have mitogenic, apoptotic, and immunomodulatory properties (Rubanyi and Polokoff, 1994; Kedzierski and Yanagisawa, 2001; Bagnato et al., 2011). Three isoforms of endothelin have been identified to date, with endothelin-1 (ET-1) being the best studied. ET-1 is classically considered a potent vasoconstrictor. However, in addition to the effects of ET-1 on vascular smooth muscle cells, the peptide is increasingly recognized as a pro-inflammatory cytokine (Teder and Noble, 2000; Sessa et al., 1991). ET-1 causes platelet aggregation and plays a role in the increased expression of leukocyte adhesion molecules, the synthesis of inflammatory mediators contributing to vascular dysfunction. High levels of ET-1 are found in alveolar macrophages, leukocytes (Sessa et al., 1991) and fibroblasts (Gu et al., 1991). Clinical and experimental data indicate that ET-1 is involved in the pathogenesis of sepsis (Tschaikowsky et al., 2000; Goto et al., 2012), viral and bacterial pneumonia (Schuetz et al., 2008; Samransamruajkit et al., 2002), Rickettsia conorii infections (Davi et al., 1995), Chagas disease (Petkova et al., 2000, 2001), and severe malaria (Dai et al., 2012; Machado et al., 2006; Wenisch et al., 1996a; Dietmann et al., 2008). In this minireview, we will discuss the role of endothelin in the pathogenesis of infectious processes.
Collapse
|