1
|
Abreu C, Shirk EN, Queen SE, Beck SE, Mangus LM, Pate KAM, Mankowski JL, Gama L, Clements JE. Brain macrophages harbor latent, infectious simian immunodeficiency virus. AIDS 2019; 33 Suppl 2:S181-S188. [PMID: 31789817 PMCID: PMC7058191 DOI: 10.1097/qad.0000000000002269] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
: The current review examines the role of brain macrophages, that is perivascular macrophages and microglia, as a potential viral reservoir in antiretroviral therapy (ART) treated, simian immunodeficiency virus (SIV)-infected macaques. The role, if any, of latent viral reservoirs of HIV and SIV in the central nervous system during ART suppression is an unresolved issue. HIV and SIV infect both CD4 lymphocytes and myeloid cells in blood and tissues during acute and chronic infection. HIV spread to the brain occurs during acute infection by the infiltration of activated CD4 lymphocytes and monocytes from blood and is established in both embryonically derived resident microglia and monocyte-derived perivascular macrophages. ART controls viral replication in peripheral blood and cerebrospinal fluid in HIV-infected individuals but does not directly eliminate infected cells in blood, tissues or brain. Latently infected resting CD4 lymphocytes in blood and lymphoid tissues are a well recognized viral reservoir that can rebound once ART is withdrawn. In contrast, central nervous system resident microglia and perivascular macrophages in brain have not been examined as potential reservoirs for HIV during suppressive ART. Macrophages in tissues are long-lived cells that are HIV and SIV infected in tissues such as gut, lung, spleen, lymph node and brain and contribute to ongoing inflammation in tissues. However, their potential role in viral persistence and latency or their potential to rebound in the absence ART has not been examined. It has been shown that measurement of HIV latency by HIV DNA PCR in CD4 lymphocytes overestimates the size of the latent reservoirs of HIV that contribute to rebound that is cells containing the genomes of replicative viruses. Thus, the quantitative viral outgrowth assay has been used as a reliable measure of the number of latent cells that harbor infectious viral DNA and, may constitute a functional latent reservoir. Using quantitative viral outgrowth assays specifically designed to quantitate latently infected CD4 lymphocytes and myeloid cells in an SIV macaque model, we demonstrated that macrophages in brain harbor SIV genomes that reactivate and produce infectious virus in this assay, demonstrating that these cells have the potential to be a reservoir.
Collapse
Affiliation(s)
- Celina Abreu
- Department of Molecular and Comparative Pathobiology
| | - Erin N Shirk
- Department of Molecular and Comparative Pathobiology
| | | | - Sarah E Beck
- Department of Molecular and Comparative Pathobiology
| | - Lisa M Mangus
- Department of Molecular and Comparative Pathobiology
| | | | - Joseph L Mankowski
- Department of Molecular and Comparative Pathobiology
- Department of Neurology
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lucio Gama
- Department of Molecular and Comparative Pathobiology
| | - Janice E Clements
- Department of Molecular and Comparative Pathobiology
- Department of Neurology
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Han L, He H, Qu X, Liu Y, He S, Zheng X, He F, Bai H, Bo X. The relationships among host transcriptional responses reveal distinct signatures underlying viral infection-disease associations. MOLECULAR BIOSYSTEMS 2016; 12:653-65. [PMID: 26699092 DOI: 10.1039/c5mb00657k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Genome-scale DNA microarrays and computational biology facilitate new understanding of viral infections at the system level. Recent years have witnessed a major shift from microorganism-centric toward host-oriented characterization and categorization of viral infections and infection related diseases. We established host transcriptional response (HTR) relationships among 23 different types of human viral pathogens based on calculating HTR similarities using computational integration of 587 public available gene expression profiles. We further identified five virus clusters that show consensus internal HTRs and defined cluster signatures using common dysregulated genes. Individual cluster signature genes were distinguished from one another, and functional analysis revealed common and specific host cellular bioprocesses and signaling pathways involved in confronting viral infections. Through literature investigation and support from epidemiological studies, these were confirmed to be important gene factors associating viral infections with cluster-common and cluster-specific non-infectious human disease(s). Our analyses were the first to feature differential HTRs to viral infections as clusters, and they present a new perspective for understanding infection-disease associations and the underlying pathogeneses.
Collapse
Affiliation(s)
- Lu Han
- Department of Traditional Chinese Medicine and Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China and Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, China.
| | - Haochen He
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, China.
| | - Xinyan Qu
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, China.
| | - Yang Liu
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, China.
| | - Song He
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, China.
| | - Xiaofei Zheng
- Department of Biochemistry and Molecular Biology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Fuchu He
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, China.
| | - Hui Bai
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, China. and No. 451 Hospital of Chinese People's Liberation Army, Xi'an, China
| | - Xiaochen Bo
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, China.
| |
Collapse
|
3
|
Proliferation of Perivascular Macrophages Contributes to the Development of Encephalitic Lesions in HIV-Infected Humans and in SIV-Infected Macaques. Sci Rep 2016; 6:32900. [PMID: 27610547 PMCID: PMC5017189 DOI: 10.1038/srep32900] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/17/2016] [Indexed: 11/09/2022] Open
Abstract
The aim of the present study was to investigate if macrophage proliferation occurs in the brain during simian immunodeficiency virus (SIV) infection of adult macaques. We examined the expression of the Ki-67 proliferation marker in the brains of uninfected and SIV-infected macaques with or without encephalitis. Double-label immunohistochemistry using antibodies against the pan-macrophage marker CD68 and Ki-67 showed that there was a significant increase in CD68+Ki-67+ cells in macaques with SIV encephalitis (SIVE) compared to uninfected and SIV-infected animals without encephalitis, a trend that was also confirmed in brain samples from patients with HIV encephalitis. Multi-label immunofluorescence for CD163 and Ki-67 confirmed that the vast majority of Ki-67+ nuclei were localized to CD163+ macrophages in perivascular cuffs and lesions. The proliferative capacity of Ki-67+ perivascular macrophages (PVM) was confirmed by their nuclear incorporation of bromodeoxyuridine. Examining SIVE lesions, using double-label immunofluorescence with antibodies against SIV-Gag-p28 and Ki-67, showed that the population of Ki-67+ cells were productively infected and expanded proportionally with lesions. Altogether, this study shows that there are subpopulations of resident PVM that express Ki-67 and are SIV-infected, suggesting a mechanism of macrophage accumulation in the brain via PVM proliferation.
Collapse
|
4
|
Correia S, Cohen R, Gongvatana A, Ross S, Olchowski J, Devlin K, Tashima K, Navia B, Delamonte S. Relationship of plasma cytokines and clinical biomarkers to memory performance in HIV. J Neuroimmunol 2013; 265:117-23. [PMID: 24210837 DOI: 10.1016/j.jneuroim.2013.09.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 08/15/2013] [Accepted: 09/10/2013] [Indexed: 11/26/2022]
Abstract
Chronic systemic immune activation and inflammatory processes have been linked to brain dysfunction in medically stable HIV-infected people. We investigated the association between verbal memory performance and plasma concentrations of 13 cytokines measured using multiplexed bead array immunoassay in 74 HIV-seropositive individuals and 50 HIV-seronegative controls. Memory performance was positively related to levels of IL-8 and IFN-γ, and negatively related to IL-10 and IL-18 and to hepatitis C infection. Memory performance was not significantly related to HIV disease markers. The results indicate the importance of systemic immune and inflammatory markers to neurocognitive function in chronic and stable HIV disease.
Collapse
Affiliation(s)
- Stephen Correia
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Abstract
Human immunodeficiency virus type 1 is associated with the development of neurocognitive disorders in many infected individuals, including a broad spectrum of motor impairments and cognitive deficits. Despite extensive research, the pathogenesis of HIV-associated neurocognitive disorders (HAND) is still not clear. This review provides a comprehensive view of HAND, including HIV neuroinvasion, HAND diagnosis and different level of disturbances, influence of highly-active antiretroviral therapy to HIV-associated dementia (HAD), possible pathogenesis of HAD, etc. Together, this review will give a thorough and clear understanding of HAND, especially HAD, which will be vital for future research, diagnosis and treatment.
Collapse
Affiliation(s)
- Li Zhou
- Retroviral Genetics Division, Center for Virus Research, Westmead Millennium Institute, Westmead Hospital, The University of Sydney , Australia
| | - Nitin K Saksena
- Retroviral Genetics Division, Center for Virus Research, Westmead Millennium Institute, Westmead Hospital, The University of Sydney , Australia
| |
Collapse
|
6
|
Wilson NO, Solomon W, Anderson L, Patrickson J, Pitts S, Bond V, Liu M, Stiles JK. Pharmacologic inhibition of CXCL10 in combination with anti-malarial therapy eliminates mortality associated with murine model of cerebral malaria. PLoS One 2013; 8:e60898. [PMID: 23630573 PMCID: PMC3618178 DOI: 10.1371/journal.pone.0060898] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 03/04/2013] [Indexed: 02/05/2023] Open
Abstract
Despite appropriate anti-malarial treatment, cerebral malaria (CM)-associated mortalities remain as high as 30%. Thus, adjunctive therapies are urgently needed to prevent or reduce such mortalities. Overproduction of CXCL10 in a subset of CM patients has been shown to be tightly associated with fatal human CM. Mice with deleted CXCL10 gene are partially protected against experimental cerebral malaria (ECM) mortality indicating the importance of CXCL10 in the pathogenesis of CM. However, the direct effect of increased CXCL10 production on brain cells is unknown. We assessed apoptotic effects of CXCL10 on human brain microvascular endothelial cells (HBVECs) and neuroglia cells in vitro. We tested the hypothesis that reducing overexpression of CXCL10 with a synthetic drug during CM pathogenesis will increase survival and reduce mortality. We utilized atorvastatin, a widely used synthetic blood cholesterol-lowering drug that specifically targets and reduces plasma CXCL10 levels in humans, to determine the effects of atorvastatin and artemether combination therapy on murine ECM outcome. We assessed effects of atorvastatin treatment on immune determinants of severity, survival, and parasitemia in ECM mice receiving a combination therapy from onset of ECM (day 6 through 9 post-infection) and compared results with controls. The results indicate that CXCL10 induces apoptosis in HBVECs and neuroglia cells in a dose-dependent manner suggesting that increased levels of CXCL10 in CM patients may play a role in vasculopathy, neuropathogenesis, and brain injury during CM pathogenesis. Treatment of ECM in mice with atorvastatin significantly reduced systemic and brain inflammation by reducing the levels of the anti-angiogenic and apoptotic factor (CXCL10) and increasing angiogenic factor (VEGF) production. Treatment with a combination of atorvastatin and artemether improved survival (100%) when compared with artemether monotherapy (70%), p<0.05. Thus, adjunctively reducing CXCL10 levels and inflammation by atorvastatin treatment during anti-malarial therapy may represent a novel approach to treating CM patients.
Collapse
Affiliation(s)
- Nana O. Wilson
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Wesley Solomon
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Leonard Anderson
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - John Patrickson
- Department of Pathology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Sidney Pitts
- Department of Pathology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Vincent Bond
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Mingli Liu
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Jonathan K. Stiles
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
7
|
Li MD, Cao J, Wang S, Wang J, Sarkar S, Vigorito M, Ma JZ, Chang SL. Transcriptome sequencing of gene expression in the brain of the HIV-1 transgenic rat. PLoS One 2013; 8:e59582. [PMID: 23536882 PMCID: PMC3607591 DOI: 10.1371/journal.pone.0059582] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 02/15/2013] [Indexed: 11/19/2022] Open
Abstract
The noninfectious HIV-1 transgenic (HIV-1Tg) rat was developed as a model of AIDs-related pathology and immune dysfunction by manipulation of a noninfectious HIV-1gag-pol virus with a deleted 3-kb SphI-MscI fragment containing the 3′ -region of gag and the 5′ region of pol into F344 rats. Our previous studies revealed significant behavioral differences between HIV-1Tg and F344 control rats in their performance in the Morris water maze and responses to psychostimulants. However, the molecular mechanisms underlying these behavioral differences remain largely unknown. The primary goal of this study was to identify differentially expressed genes and enriched pathways affected by the gag-pol-deleted HIV-1 genome. Using RNA deep sequencing, we sequenced RNA transcripts in the prefrontal cortex, hippocampus, and striatum of HIV-1Tg and F344 rats. A total of 72 RNA samples were analyzed (i.e., 12 animals per group × 2 strains × 3 brain regions). Following deep-sequencing analysis of 50-bp paired-end reads of RNA-Seq, we used Bowtie/Tophat/Cufflinks suites to align these reads into transcripts based on the Rn4 rat reference genome and to measure the relative abundance of each transcript. Statistical analyses on each brain region in the two strains revealed that immune response- and neurotransmission-related pathways were altered in the HIV-1Tg rats, with brain region differences. Other neuronal survival-related pathways, including those encoding myelin proteins, growth factors, and translation regulators, were altered in the HIV-1Tg rats in a brain region-dependent manner. This study is the first deep-sequencing analysis of RNA transcripts associated the HIV-1Tg rat. Considering the functions of the pathways and brain regions examined in this study, our findings of abnormal gene expression patterns in HIV-1Tg rats suggest mechanisms underlying the deficits in learning and memory and vulnerability to drug addiction and other psychiatric disorders observed in HIV-positive patients.
Collapse
Affiliation(s)
- Ming D. Li
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail: (MDL); (SLC)
| | - Junran Cao
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| | - Shaolin Wang
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia, United States of America
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Ju Wang
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| | - Sraboni Sarkar
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, New Jersey, United States of America
- Department of Biological Sciences, Seton Hall University, South Orange, New Jersey, United States of America
| | - Michael Vigorito
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, New Jersey, United States of America
- Department of Psychology, Seton Hall University, South Orange, New Jersey, United States of America
| | - Jennie Z. Ma
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| | - Sulie L. Chang
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, New Jersey, United States of America
- Department of Biological Sciences, Seton Hall University, South Orange, New Jersey, United States of America
- * E-mail: (MDL); (SLC)
| |
Collapse
|
8
|
Yuan L, Qiao L, Wei F, Yin J, Liu L, Ji Y, Smith D, Li N, Chen D. Cytokines in CSF correlate with HIV-associated neurocognitive disorders in the post-HAART era in China. J Neurovirol 2013; 19:144-9. [PMID: 23389619 DOI: 10.1007/s13365-013-0150-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 01/07/2013] [Accepted: 01/21/2013] [Indexed: 11/26/2022]
Abstract
In the current era of highly active antiretroviral therapy (HAART), the incidence of HIV dementia has declined, but the prevalence of HIV-associated neurocognitive disorder (HAND) remains high. HIV-induced systemic and localized inflammation is considered to be one of the mechanisms of HAND. Changes in cytokine levels in the cerebrospinal fluid (CSF) during HIV infection might help to identify HAND. To investigate whether the cytokine profile of the CSF during HIV infection could be used as a biomarker of HAND, we compared cytokine levels in the CSF of HIV-infected cases with and without neurocognitive impairment. Cytokine concentrations in the CSF were measured by quantification bioassays (Luminex xMAP). HIV-infected cases with neurocognitive impairment demonstrated higher levels of interleukin (IL)-8, monocyte chemotactic protein (MCP)-1, induced protein (IP)-10, and granulocyte colony-stimulating factor (G-CSF) in the CSF than those without neurocognitive impairment (G-CSF (p = 0.0003), IL-8 (p = 0.0046), IP-10 (p < 0.0001), and MCP-1 (p < 0.0001)). There was no significant impact of HAART on cytokine levels in the CSF, except for IP-10, which was higher in HAART-treated patients with impaired cognition (p = 0.0182). Findings from this preliminary study suggest that elevated levels of the cytokines IL-8, MCP-1, G-CSF, and IP-10 in the CSF are associated with neurocognitive impairment in HIV infection, and these cytokines likely represent a biomarker profile for HAND.
Collapse
Affiliation(s)
- Lin Yuan
- Department of Medicine, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Zhou L, Pupo GM, Gupta P, Liu B, Tran SL, Rahme R, Wang B, Rua R, Rizos H, Carroll A, Cairns MJ, Saksena NK. A parallel genome-wide mRNA and microRNA profiling of the frontal cortex of HIV patients with and without HIV-associated dementia shows the role of axon guidance and downstream pathways in HIV-mediated neurodegeneration. BMC Genomics 2012; 13:677. [PMID: 23190615 PMCID: PMC3560210 DOI: 10.1186/1471-2164-13-677] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 11/20/2012] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND HIV-associated dementia (HAD) is the most common dementia type in young adults less than 40 years of age. Although the neurotoxins, oxidative/metabolic stress and impaired activity of neurotrophic factors are believed to be underlying reasons for the development of HAD, the genomic basis, which ultimately defines the virus-host interaction and leads to neurologic manifestation of HIV disease is lacking. Therefore, identifying HIV fingerprints on the host gene machinery and its regulation by microRNA holds a great promise and potential for improving our understanding of HAD pathogenesis, its diagnosis and therapy. RESULTS A parallel profiling of mRNA and miRNA of the frontal cortex autopsies from HIV positive patients with and without dementia was performed using Illumina Human-6 BeadChip and Affymetrix version 1.0 miRNA array, respectively. The gene ontology and pathway analysis of the two data sets showed high concordance between miRNA and mRNAs, revealing significant interference with the host axon guidance and its downstream signalling pathways in HAD brains. Moreover, the differentially expressed (DE) miRNAs identified in this study, in particular miR-137, 153 and 218, based on which most correlations were built cumulatively targeted neurodegeneration related pathways, implying their future potential in diagnosis, prognosis and possible therapies for HIV-mediated and possibly other neurodegenerative diseases. Furthermore, this relationship between DE miRNAs and DE mRNAs was also reflected in correlation analysis using Bayesian networks by splitting-averaging strategy (SA-BNs), which revealed 195 statistically significant correlated miRNA-mRNA pairs according to Pearson's correlation test (P<0.05). CONCLUSIONS Our study provides the first evidence on unambiguous support for intrinsic functional relationship between mRNA and miRNA in the context of HIV-mediated neurodegeneration, which shows that neurologic manifestation in HIV patients possibly occurs through the interference with the host axon guidance and its downstream signalling pathways. These data provide an excellent avenue for the development of new generation of diagnostic/prognostic biomarkers and therapeutic intervention strategies for HIV-associated neurodegeneration.
Collapse
Affiliation(s)
- Li Zhou
- Retroviral Genetics Division, Center for Virus Research, Westmead Millennium Institute, Westmead Hospital, The University of Sydney, Westmead, NSW 2145, Sydney, Australia
| | - Gulietta M Pupo
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead, NSW, 2145, Australia
| | - Priyanka Gupta
- Retroviral Genetics Division, Center for Virus Research, Westmead Millennium Institute, Westmead Hospital, The University of Sydney, Westmead, NSW 2145, Sydney, Australia
| | - Bing Liu
- School of Biomedical Sciences and Pharmacy, Faculty of Health and the Hunter Medical Research Institute, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Sieu L Tran
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead, NSW, 2145, Australia
| | - Raany Rahme
- Retroviral Genetics Division, Center for Virus Research, Westmead Millennium Institute, Westmead Hospital, The University of Sydney, Westmead, NSW 2145, Sydney, Australia
| | - Bin Wang
- Retroviral Genetics Division, Center for Virus Research, Westmead Millennium Institute, Westmead Hospital, The University of Sydney, Westmead, NSW 2145, Sydney, Australia
| | - Rejane Rua
- Retroviral Genetics Division, Center for Virus Research, Westmead Millennium Institute, Westmead Hospital, The University of Sydney, Westmead, NSW 2145, Sydney, Australia
| | - Helen Rizos
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead, NSW, 2145, Australia
| | - Adam Carroll
- School of Biomedical Sciences and Pharmacy, Faculty of Health and the Hunter Medical Research Institute, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy, Faculty of Health and the Hunter Medical Research Institute, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
- Schizophrenia Research Institute, Darlinghurst, Sydney NSW, Australia
| | - Nitin K Saksena
- Retroviral Genetics Division, Center for Virus Research, Westmead Millennium Institute, Westmead Hospital, The University of Sydney, Westmead, NSW 2145, Sydney, Australia
| |
Collapse
|
10
|
Cohen RA, de la Monte S, Gongvatana A, Ombao H, Gonzalez B, Devlin KN, Navia B, Tashima KT. Plasma cytokine concentrations associated with HIV/hepatitis C coinfection are related to attention, executive and psychomotor functioning. J Neuroimmunol 2011; 233:204-10. [PMID: 21146232 PMCID: PMC3074016 DOI: 10.1016/j.jneuroim.2010.11.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 11/06/2010] [Accepted: 11/16/2010] [Indexed: 01/26/2023]
Abstract
Cytokine disturbances have been linked to brain dysfunction among HIV-infected people. Past studies have not simultaneously examined a large set of cytokine measures and their relationships to HIV-associated neurocognitive deficits. We hypothesized that performance on measures of attention and executive and psychomotor functions would be associated with plasma cytokine concentrations in HIV-infected individuals. Plasma samples drawn from 30 HIV-infected and 37 HIV seronegative individuals were analyzed via xMAP multiplexed bead array immunoassay to determine concentrations of 13 cytokines. Performance on Trail Making A/B, Stroop Test, Letter-Number Sequencing, Digit Symbol Coding, Symbol Search, and Grooved Pegboard tests was assessed. Statistical analyses were performed to examine group differences in cytokine concentrations, and associations between cytokine and HIV clinical variables and neurocognitive performance. Significant HIV effects were found on 7 of the 13 cytokines, primarily with respect to interleukins. HIV clinical factors (CD4 and HIV RNA levels, duration of illness, antiretroviral treatment) and hepatitis C status were associated with specific plasma cytokine concentrations. Neurocognitive measures were associated with cytokine concentrations, most consistently among the interleukins and IP-10. Generally, cytokine concentrations were among the strongest predictors of neurocognitive function relative to other clinical factors, which reinforces their potential importance in examining the neuropathological processes of HIV. The findings also point to the potential value of simultaneously examining a panel of biomarkers. The current results suggest that a complex relationship likely exists among cytokines [how?] and that these relationships are mediated not only by HIV infection but also by antiretroviral treatment and other comorbid conditions.
Collapse
Affiliation(s)
- Ronald A Cohen
- Department of Psychiatry and Human Behavior, Brown University School of Medicine, Providence, RI, USA.
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Letendre SL, Zheng JC, Kaul M, Yiannoutsos CT, Ellis RJ, Taylor MJ, Marquie-Beck J, Navia B. Chemokines in cerebrospinal fluid correlate with cerebral metabolite patterns in HIV-infected individuals. J Neurovirol 2011; 17:63-9. [PMID: 21246320 PMCID: PMC3032187 DOI: 10.1007/s13365-010-0013-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 11/14/2010] [Accepted: 11/24/2010] [Indexed: 12/01/2022]
Abstract
Chemokines influence HIV neuropathogenesis by affecting the HIV life cycle, trafficking of macrophages into the nervous system, glial activation, and neuronal signaling and repair processes; however, knowledge of their relationship to in vivo measures of cerebral injury is limited. The primary objective of this study was to determine the relationship between a panel of chemokines in cerebrospinal fluid (CSF) and cerebral metabolites measured by proton magnetic resonance spectroscopy (MRS) in a cohort of HIV-infected individuals. One hundred seventy-one stored CSF specimens were assayed from HIV-infected individuals who were enrolled in two ACTG studies that evaluated the relationship between neuropsychological performance and cerebral metabolites. Concentrations of six chemokines (fractalkine, IL-8, IP-10, MCP-1, MIP-1β, and SDF-1) were measured and compared with cerebral metabolites individually and as composite neuronal, basal ganglia, and inflammatory patterns. IP-10 and MCP-1 were the chemokines most strongly associated with individual cerebral metabolites. Specifically, (1) higher IP-10 levels correlated with lower N-acetyl aspartate (NAA)/creatine (Cr) ratios in the frontal white matter and higher MI/Cr ratios in all three brain regions considered and (2) higher MCP-1 levels correlated with lower NAA/Cr ratios in frontal white matter and the parietal cortex. IP-10, MCP-1, and IL-8 had the strongest associations with patterns of cerebral metabolites. In particular, higher levels of IP-10 correlated with lower neuronal pattern scores and higher basal ganglia and inflammatory pattern scores, the same pattern which has been associated with HIV-associated neurocognitive disorders (HAND). Subgroup analysis indicated that the effects of IP-10 and IL-8 were influenced by effective antiretroviral therapy and that memantine treatment may mitigate the neuronal effects of IP-10. This study supports the role of chemokines in HAND and the validity of MRS as an assessment tool. In particular, the findings identify relationships between the immune response—particularly an interferon-inducible chemokine, IP-10—and cerebral metabolites and suggest that antiretroviral therapy and memantine modify the impact of the immune response on neurons.
Collapse
Affiliation(s)
- Scott L Letendre
- University of California, San Diego, 220 Dickinson Street, Suite A, San Diego, CA 92103, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Suh HS, Cosenza-Nashat M, Choi N, Zhao ML, Li JF, Pollard JW, Jirtle RL, Goldstein H, Lee SC. Insulin-like growth factor 2 receptor is an IFNgamma-inducible microglial protein that facilitates intracellular HIV replication: implications for HIV-induced neurocognitive disorders. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:2446-58. [PMID: 20889566 DOI: 10.2353/ajpath.2010.100399] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Insulin-like growth factor 2 receptor (IGF2R), also known as cation-independent mannose 6-phosphate (M6P) receptor, is a transmembrane glycoprotein localized in the trans-Golgi region and is involved in targeting both M6P-bearing enzymes and IGF2 to the lysosomal compartment. During development, IGF2R plays a crucial role in removing excess growth factors from both tissue and blood. Due to the perinatal lethality of the global Igf2r knockout, the function of IGF2R in adults, particularly in the CNS, is not known. We made a novel observation that IGF2R is highly expressed in microglial nodules in human brains with HIV encephalitis. In vitro, microglial IGF2R expression was uniquely enhanced by IFNγ among the several cytokines and TLR ligands examined. Furthermore, in several in vitro models of HIV infection, including human and murine microglia, macrophages, and nonmacrophage cells, IGF2R is repeatedly shown to be a positive regulator of HIV infection. IGF2R RNAi also down-regulated the production of the IP-10 chemokine in HIV-infected human microglia. Injection of VSVg env HIV into mouse brain induced HIV p24 expression in neurons, the only cell type normally expressing IGF2R in the adult brain. Our results demonstrate a novel role for IGF2R as an inducible microglial protein involved in regulation of HIV and chemokine expression. Mice with the Csf1r- driven Igf2r knockout should be useful for the investigation of macrophage-specific IGF2R function.
Collapse
Affiliation(s)
- Hyeon-Sook Suh
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Dhillon NK, Li F, Xue B, Tawfik O, Morgello S, Buch S, Ladner AO. Effect of cocaine on human immunodeficiency virus-mediated pulmonary endothelial and smooth muscle dysfunction. Am J Respir Cell Mol Biol 2010; 45:40-52. [PMID: 20802087 DOI: 10.1165/rcmb.2010-0097oc] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Human immunodeficiency virus (HIV)-associated pulmonary arterial hypertension (PAH) is a devastating, noninfectious complication of acquired immune deficiency syndrome, and the majority of HIV-PAH cases occur in individuals with a history of intravenous drug use (IVDU). However, although HIV-1 and IVDU have been associated with PAH independently or in combination, the pathogenesis of the disproportionate presence of HIV-PAH in association with IVDU has yet to be characterized. The objective of this study was to obtain a better understanding of the interactions between HIV-1 and cocaine to help uncover the mechanism(s) of the development of HIV-PAH. We observed that exposure of HIV-infected macrophages or HIV-Trans-Activator of Transcription (Tat)-treated pulmonary endothelial cells to cocaine enhanced the expression of platelet-derived growth factor (PDGF)-BB. Simultaneous treatment with Tat and cocaine, on the other hand, exacerbated both the disruption of tight junction proteins (TJPs), with enhanced permeability in pulmonary endothelial cells, and the proliferation of pulmonary smooth muscle cells (pSMCs) compared with either treatment alone. Histological examination of HIV plus IVDU human lung sections showed signs of early pulmonary arteriopathy, severe down-modulation of TJPs, and increased expression of PDGF-BB compared with the lung sections from individuals who are infected with HIV and without history of IVDU. Interestingly, blocking of PDGF receptor signaling with the receptor antagonist or small interfering RNA has been shown to inhibit the increase in proliferation of pSMCs on Tat and cocaine exposure. Our results, therefore, support an additive effect of cocaine to HIV infection in the development of pulmonary arteriopathy through enhancement of endothelial dysfunction and proliferation of pSMCs, while also suggesting PDGF-PDGF receptor axis as a potential target for use in clinical intervention.
Collapse
Affiliation(s)
- Navneet K Dhillon
- Department of Molecular and Integrative Physiology, 3008 Hixon, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA.
| | | | | | | | | | | | | |
Collapse
|
14
|
Zhou L, Diefenbach E, Crossett B, Tran SL, Ng T, Rizos H, Rua R, Wang B, Kapur A, Gandhi K, Brew BJ, Saksena NK. First evidence of overlaps between HIV-Associated Dementia (HAD) and non-viral neurodegenerative diseases: proteomic analysis of the frontal cortex from HIV+ patients with and without dementia. Mol Neurodegener 2010; 5:27. [PMID: 20573273 PMCID: PMC2904315 DOI: 10.1186/1750-1326-5-27] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2009] [Accepted: 06/24/2010] [Indexed: 12/12/2022] Open
Abstract
Background The pathogenesis of HIV-associated dementia (HAD) is poorly understood. To date, detailed proteomic fingerprinting directly from autopsied brain tissues of HAD and HIV non-dementia patients has not been performed. Result Here, we have analyzed total proteins from the frontal cortex of 9 HAD and 5 HIV non-dementia patients. Using 2-Dimensional differential in-gel electrophoresis (2-DIGE) to analyze the brain tissue proteome, 76 differentially expressed proteins (p < 0.05; fold change>1.25) were identified between HAD and HIV non-dementia patients, of which 36 protein spots (based on 3D appearance of spots on the images) were chosen for the mass spectrometry analysis. The large majority of identified proteins were represented in the energy metabolic (mitochondria) and signal transduction pathways. Furthermore, over 90% of the protein candidates are common to both HAD and other non-viral neurodegenerative disease, such as Alzheimer's disease. The data was further validated using specific antibodies to 4 proteins (CA2, GS, CKMT and CRMP2) by western blot (WB) in the same samples used for 2D-DIGE, with additional confirmation by immunohistochemitsry (IHC) using frontal lobe tissue from different HAD and HIV+ non-dementia patients. The validation for all 4 antibodies by WB and IHC was in concordance with the DIGE results, lending further credence to the current findings. Conclusion These results suggest not only convergent pathogenetic pathways for the two diseases but also the possibility of increased Alzheimer's disease (AD) susceptibility in HAD patients whose life expectancy has been significantly increased by highly active antiretroviral therapy.
Collapse
Affiliation(s)
- Li Zhou
- Center for Virus Research, Westmead Millennium Institute, Westmead Hospital, The University of Sydney, Westmead, NSW 2145, Sydney, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Greer KA, Daly P, Murphy KE, Callanan JJ. Analysis of gene expression in brain tissue from Greyhounds with meningoencephalitis. Am J Vet Res 2010; 71:547-54. [DOI: 10.2460/ajvr.71.5.547] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
16
|
Yao H, Bethel-Brown C, Li CZ, Buch SJ. HIV neuropathogenesis: a tight rope walk of innate immunity. J Neuroimmune Pharmacol 2010; 5:489-95. [PMID: 20354805 DOI: 10.1007/s11481-010-9211-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Accepted: 03/11/2010] [Indexed: 12/12/2022]
Abstract
During the course of HIV-1 disease, virus neuroinvasion occurs as an early event, within weeks following infection. Intriguingly, subsequent central nervous system (CNS) complications manifest only decades after the initial virus exposure. Although CNS is commonly regarded as an immune-privileged site, emerging evidence indicates that innate immunity elicited by the CNS glial cells is a critical determinant for the establishment of protective immunity. Sustained expression of these protective immune responses, however, can be a double-edged sword. As protective immune mediators, cytokines have the ability to function in networks and co-operate with other host/viral mediators to tip the balance from a protective to toxic state in the CNS. Herein, we present an overview of some of the essential elements of the cerebral innate immunity in HIV neuropathogenesis including the key immune cell types of the CNS with their respective soluble immune mediators: (1) cooperative interaction of IFN-γ with the host/virus factor (platelet-derived host factor (PDGF)/viral Tat) in the induction of neurotoxic chemokine CXCL10 by macrophages, (2) response of astrocytes to viral infection, and (3) protective role of PDGF and MCP-1 in neuronal survival against HIV Tat toxicity. These components of the cerebral innate immunity do not act separately from each other but form a functional immunity network. The ultimate outcome of HIV infection in the CNS will thus be dependent on the regulation of the net balance of cell-specific protective versus detrimental responses.
Collapse
Affiliation(s)
- Honghong Yao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | | | | | | |
Collapse
|
17
|
Borjabad A, Brooks AI, Volsky DJ. Gene expression profiles of HIV-1-infected glia and brain: toward better understanding of the role of astrocytes in HIV-1-associated neurocognitive disorders. J Neuroimmune Pharmacol 2010; 5:44-62. [PMID: 19697136 PMCID: PMC3107560 DOI: 10.1007/s11481-009-9167-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 07/27/2009] [Indexed: 12/17/2022]
Abstract
Astrocytes are the major cellular component of the central nervous system (CNS), and they play multiple roles in brain development, normal brain function, and CNS responses to pathogens and injury. The functional versatility of astrocytes is linked to their ability to respond to a wide array of biological stimuli through finely orchestrated changes in cellular gene expression. Dysregulation of gene expression programs, generally by chronic exposure to pathogenic stimuli, may lead to dysfunction of astrocytes and contribute to neuropathogenesis. Here, we review studies that employ functional genomics to characterize the effects of HIV-1 and viral pathogenic proteins on cellular gene expression in astrocytes in vitro. We also present the first microarray analysis of primary mouse astrocytes exposed to HIV-1 in culture. In spite of different experimental conditions and microarray platforms used, comparison of the astrocyte array data sets reveals several common gene-regulatory changes that may underlie responses of these cells to HIV-1 and its proteins. We also compared the transcriptional profiles of astrocytes with those obtained in analyses of brain tissues of patients with HIV-1 dementia and macaques infected with simian immunodeficiency virus (SIV). Notably, many of the gene characteristics of responses to HIV-1 in cultured astrocytes were also altered in HIV-1 or SIV-infected brains. Functional genomics, in conjunction with other approaches, may help clarify the role of astrocytes in HIV-1 neuropathogenesis.
Collapse
Affiliation(s)
- Alejandra Borjabad
- Molecular Virology Division, St. Luke's-Roosevelt Hospital Center, 432 West 58th Street, Antenucci Building, Room 709, New York, NY 10019, USA
| | | | | |
Collapse
|
18
|
Abstract
Viral encephalitis remains a significant cause of morbidity and mortality throughout the world. We performed microarray analysis to identify genes and pathways that are differentially regulated during reovirus encephalitis and that may provide novel therapeutic targets for virus-induced diseases of the central nervous system (CNS). An increase in the expression of 130 cellular genes was found in the brains of reovirus-infected mice at early times post infection, compared to mock-infected controls. The up-regulation of these genes was consistent with activation of innate immune responses, particularly interferon signaling. At later times post infection, when significant CNS injury is present and mice exhibit signs of severe neurologic disease, many more (1374) genes were up-regulated, indicating that increased gene expression correlates with disease pathology. Virus-induced gene expression at late times post infection was again consistent with the activation of innate immune responses. However, additional significant pathways included those associated with cytokine signaling and apoptosis, both of which can contribute to CNS injury. This is the first report comparing virus-induced cellular gene and pathway regulation at early and late times following virus infection of the brain. The shift of virus-induced gene expression from innate immune responses at early times post infection to cytokine signaling and apoptosis at later times suggests a potential therapeutic strategy that preserves early protective responses whilst inhibiting later responses that contribute to pathogenesis.
Collapse
Affiliation(s)
- Kenneth L Tyler
- Department of Neurology, University of Colorado–Denver, Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Medicine, University of Colorado–Denver, Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Microbiology, University of Colorado–Denver, Anschutz Medical Campus, Aurora, Colorado, USA
- Denver Veterans Administration, Denver, Colorado, USA
| | - J Smith Leser
- Department of Neurology, University of Colorado–Denver, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tzu L Phang
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado–Denver, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Penny Clarke
- Department of Neurology, University of Colorado–Denver, Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
19
|
Abstract
Chemokines are small chemoattractant cytokines involved in homeostatic and inflammatory immune cell migration. These small proteins have multiple functional properties that extend beyond their most recognized role in controlling cellular migration. The complex immunobiology of chemokines, coupled with the use of subsets of chemokine receptors as HIV-1 and SIV entry co-receptors, suggests that these immunomodulators could play important roles in the pathogenesis associated with infection by HIV-1 or SIV. This review provides an overview of the effects of pathogenic infection on chemokine expression in the SIV/macaque model system, and outlines potential mechanisms by which changes in these expression profiles could contribute to development of disease. Key challenges faced in studying chemokine function in vivo and new opportunities for further study and development of therapeutic interventions are discussed. Continued growth in our understanding of the effects of pathogenic SIV infection on chemokine expression and function and the continuing development of chemokine receptor targeted therapeutics will provide the tools and the systems necessary for future studies of the roles of chemokines in HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Todd A Reinhart
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, USA.
| | | | | |
Collapse
|
20
|
Noorbakhsh F, Overall CM, Power C. Deciphering complex mechanisms in neurodegenerative diseases: the advent of systems biology. Trends Neurosci 2009; 32:88-100. [DOI: 10.1016/j.tins.2008.10.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Revised: 10/08/2008] [Accepted: 10/08/2008] [Indexed: 11/28/2022]
|
21
|
Williams R, Bokhari S, Silverstein P, Pinson D, Kumar A, Buch S. Nonhuman primate models of NeuroAIDS. J Neurovirol 2009; 14:292-300. [PMID: 18780230 DOI: 10.1080/13550280802074539] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Human Immunodeficiency virus (HIV), the virus that causes acquired immunodeficiency syndrome (AIDS), also manifests neurological complications. HIV-associated dementia (HAD) is the most severe form of HIV-induced neurocognitive disorders. HIV encephalitis (HIVE), the pathological correlate of HAD, is characterized by the formation of multinucleated giant cells and microglial nodules, astrocytosis, and neuronal damage and loss. Pathological evaluation of HAD disease progression in humans is not possible, with the only data collected being from individuals who have succumbed to the disorder, a snap shot of end-stage disease at best. Therefore, pertinent animal models have been developed to alleviate this gap of knowledge in the field of neurovirology and neuroinflammation. In general, the most widely used animal models are the simian immunodeficiency virus (SIV) and the chimeric simian/human immunodeficiency virus (SHIV) macaque model systems. Although both SIV and SHIV model systems are able to potentiate neuroinvasion and the concomitant neuropathology similar to that seen in the human syndromes, the innate differences between the two in disease pathogenesis and progression make for two separate, yet effective, systems for the study of HIV-associated neuropathology.
Collapse
Affiliation(s)
- Rachel Williams
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66103, USA
| | | | | | | | | | | |
Collapse
|
22
|
Dhillon N, Zhu X, Peng F, Yao H, Williams R, Qiu J, Callen S, Ladner AO, Buch S. Molecular mechanism(s) involved in the synergistic induction of CXCL10 by human immunodeficiency virus type 1 Tat and interferon-gamma in macrophages. J Neurovirol 2008; 14:196-204. [PMID: 18569454 DOI: 10.1080/13550280801993648] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Synergistic interactions between viral proteins and soluble host factors released from infected mononuclear phagocytes play a critical role in the pathogenesis of human immunodeficiency virus (HIV)-associated dementia (HAD). The chemokine CXCL10 has been found to be closely associated with the progression of HIV-1-related central nervous system (CNS) disease and its related neuropsychiatric impairment. In this report the authors demonstrate that the HIV-1 protein Tat can interact with the proinflammatory cytokine interferon (IFN)-gamma to dramatically induce the expression of CXCL10 in macrophages. Synergistic induction of CXCL10 by both Tat and IFN-gamma was susceptible to inhibition by the MEK1/2 inhibitor U0126 and the p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580. In addition, JAK/STAT pathway plays a major role in Tat/gamma-mediated CXCL10 induction in macrophages because pretreatment of stimulated macrophages with JAK inhibitor completely abrogated the synergistic induction of the chemokine. Functionality of the synergistically induced CXCL10 was further demonstrated by its chemotactic activity for peripheral blood lymphocytes. Taken together, these findings demonstrate that the cooperative interaction of Tat and IFN-gamma results in enhanced chemokine expression, which in turn can amplify the inflammatory responses within the CNS of HAD patients by recruiting more lymphocytes in the brain.
Collapse
Affiliation(s)
- Navneet Dhillon
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Jeong AR, Nakamura S, Mitsunaga F. Gene expression profile of Th1 and Th2 cytokines and their receptors in human and nonhuman primates. J Med Primatol 2008; 37:290-6. [PMID: 18466281 DOI: 10.1111/j.1600-0684.2008.00289.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND To date comparative knowledge concerning gene expression profiles of T-helper 1(Th1)/Th2 cytokines and their receptors between human and non-human primates is scarce. METHODS We assessed the gene expression level of both Th1 [interleukin-4(IL-4)] and Th2 [IL-12, interferon-gamma(IFN-gamma)] cytokines and the receptors (IL-4Ralpha, IFN-gammaR1, IFN-gammaR2) in peripheral blood mononuclear cells (PBMC) from humans, chimpanzee, baboon, and macaque by a quantitative real-time reverse transcriptase-polymerase chain reaction(RT-PCR). RESULTS The expression level of the IFN-gamma gene was markedly lower in humans than that in non-human primates. The IL-4 gene expression was significantly higher, whereas that of IL-12 was distinctly lower, in human/chimpanzee than in baboon/macaque. The IFN-gammaR2 gene expression was especially higher in the macaque than in the other three primates. CONCLUSIONS These results indicate distinct gene expression of Th1/Th2 cytokines and their receptors in primates. These also suggest characteristic differences in Th1/Th2 immune responses affecting host defense and/or disease susceptibility among these primates.
Collapse
Affiliation(s)
- A Ram Jeong
- Department of Cellular and Molecular Biology, Primate Research Institute, Kyoto University, Inuyama, Aichi, Japan
| | | | | |
Collapse
|
24
|
Dhillon NK, Peng F, Ransohoff RM, Buch S. PDGF synergistically enhances IFN-gamma-induced expression of CXCL10 in blood-derived macrophages: implications for HIV dementia. THE JOURNAL OF IMMUNOLOGY 2007; 179:2722-30. [PMID: 17709485 DOI: 10.4049/jimmunol.179.5.2722] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
There is increasing cumulative evidence that activated mononuclear phagocytes (macrophages/microglia) releasing inflammatory mediators in the CNS are a better correlate of HIV-associated dementia (HAD) than the actual viral load in the brain. Earlier studies on simian HIV/rhesus macaque model of NeuroAIDS confirmed that pathological changes in brains of macaques with encephalitis were associated with up-regulation of platelet-derived growth factor (PDGF) and the chemokine, CXCL10. Because the complex interplay of inflammatory mediators released by macrophages often leads to the induction of neurotoxins in HAD, we hypothesized that PDGF could interact with IFN-gamma to modulate the expression of CXCL10 in these primary virus target cells. Although PDGF alone had no effect on the induction of CXCL10 in human macrophages, in conjunction with IFN-gamma, it significantly augmented the expression of CXCL10 RNA & protein through transcriptional and posttranscriptional mechanisms. Signaling molecules, such as JAK and STATs, PI3K, MAPK, and NF-kappaB were found to play a role in the synergistic induction of CXCL10. Furthermore, PDGF via its activation of p38 MAPK was able to increase the stability of IFN-gamma-induced CXCL10 mRNA. Understanding the mechanisms involved in the synergistic up-regulation of CXCL10 could aid in the development of therapeutic modalities for HAD.
Collapse
Affiliation(s)
- Navneet Kaur Dhillon
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | | | | | | |
Collapse
|
25
|
Armah HB, Wilson NO, Sarfo BY, Powell MD, Bond VC, Anderson W, Adjei AA, Gyasi RK, Tettey Y, Wiredu EK, Tongren JE, Udhayakumar V, Stiles JK. Cerebrospinal fluid and serum biomarkers of cerebral malaria mortality in Ghanaian children. Malar J 2007; 6:147. [PMID: 17997848 PMCID: PMC2186349 DOI: 10.1186/1475-2875-6-147] [Citation(s) in RCA: 163] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Accepted: 11/12/2007] [Indexed: 12/04/2022] Open
Abstract
Background Plasmodium falciparum can cause a diffuse encephalopathy known as cerebral malaria (CM), a major contributor to malaria associated mortality. Despite treatment, mortality due to CM can be as high as 30% while 10% of survivors of the disease may experience short- and long-term neurological complications. The pathogenesis of CM and other forms of severe malaria is multi-factorial and appear to involve cytokine and chemokine homeostasis, inflammation and vascular injury/repair. Identification of prognostic markers that can predict CM severity will enable development of better intervention. Methods Postmortem serum and cerebrospinal fluid (CSF) samples were obtained within 2–4 hours of death in Ghanaian children dying of CM, severe malarial anemia (SMA), and non-malarial (NM) causes. Serum and CSF levels of 36 different biomarkers (IL-1β, IL-1ra, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12 (p70), IL-13, IL-15, IL-17, Eotaxin, FGF basic protein, CRP, G-CSF, GM-CSF, IFN-γ, TNF-α, IP-10, MCP-1 (MCAF), MIP-1α, MIP-1β, RANTES, SDF-1α, CXCL11 (I-TAC), Fas-ligand [Fas-L], soluble Fas [sFas], sTNF-R1 (p55), sTNF-R2 (p75), MMP-9, TGF-β1, PDGF bb and VEGF) were measured and the results compared between the 3 groups. Results After Bonferroni adjustment for other biomarkers, IP-10 was the only serum biomarker independently associated with CM mortality when compared to SMA and NM deaths. Eight CSF biomarkers (IL-1ra, IL-8, IP-10, PDGFbb, MIP-1β, Fas-L, sTNF-R1, and sTNF-R2) were significantly elevated in CM mortality group when compared to SMA and NM deaths. Additionally, CSF IP-10/PDGFbb median ratio was statistically significantly higher in the CM group compared to SMA and NM groups. Conclusion The parasite-induced local cerebral dysregulation in the production of IP-10, 1L-8, MIP-1β, PDGFbb, IL-1ra, Fas-L, sTNF-R1, and sTNF-R2 may be involved in CM neuropathology, and their immunoassay may have potential utility in predicting mortality in CM.
Collapse
Affiliation(s)
- Henry B Armah
- Morehouse School of Medicine, Department of Microbiology, Biochemistry and Immunology, Atlanta, Georgia, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Peterson KE, Chesebro B. Influence of proinflammatory cytokines and chemokines on the neuropathogenesis of oncornavirus and immunosuppressive lentivirus infections. Curr Top Microbiol Immunol 2007; 303:67-95. [PMID: 16570857 DOI: 10.1007/978-3-540-33397-5_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Retroviral infection of the CNS can lead to severe debilitating neurological diseases in humans and other animals. Four general types of pathogenic effects with various retroviruses have been observed including: hemorrhage (TR1.3), spongiform encephalopathy (CasBrE, FrCasE, PVC211, NT40, Mol-ts1), demyelination with inflammatory lesions (HTLV-1, visna, CAEV), and encephalopathy with gliosis and proinflammatory chemokines and cytokines, usually with microglial giant cells and nodules [human immunodeficiencyvirus (HIV), feline immunodeficiencyvirus (FIV), simian immunodeficiency virus (SIV), Fr98]. This review focuses on this fourth group of retroviruses. In this latter group, proinflammatory cytokine and chemokine upregulation accompanies the disease process, and may influence pathogenesis by direct effects on resident CNS cells. The review first discusses the Fr98 murine polytropic virus system with particular reference to the roles of cytokines and chemokines in the pathogenic process. The Fr98 data are then compared and contrasted to the cytokine and chemokine data in the lentivirus systems, HIV, SIV, and FIV. Finally, various mechanisms are presented by which tumor necrosis factor (TNF) and several chemokines may alter the pathogenesis of retrovirus infection of the CNS.
Collapse
Affiliation(s)
- K E Peterson
- Dept. of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| | | |
Collapse
|
27
|
Huang D, Qiu L, Wang R, Lai X, Du G, Seghal P, Shen Y, Shao L, Halliday L, Fortman J, Shen L, Letvin NL, Chen ZW. Immune gene networks of mycobacterial vaccine-elicited cellular responses and immunity. J Infect Dis 2006; 195:55-69. [PMID: 17152009 PMCID: PMC2885892 DOI: 10.1086/509895] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2006] [Accepted: 08/22/2006] [Indexed: 11/03/2022] Open
Abstract
Gene networks of protective lymphocytes after immune activation with live attenuated vaccines remain poorly characterized. Because Mycobacterium bovis bacille Calmette-Guérin (BCG) vaccine can confer protection against fatal forms of tuberculosis in humans and monkeys, we made use of macaque models to optimally study immune gene networks after BCG vaccination/infection. We first established and validated a large-scale real-time quantitation system and then used it to measure expression levels of 138 immune genes after BCG vaccination/infection of rhesus macaques. Systemic BCG vaccination induced up to 600-fold increases in expression of 78 immune genes among the 138 genes tested at the time when BCG-elicited T cell responses and immunity were apparent. These up-regulated transcripts constituted multiple gene networks that were linked to various aspects of immune function. Surprisingly, the up-regulation of most of these immune genes in the gene networks occurred at 1 week and was sustained at > or = 6 weeks after BCG vaccination/infection. Although early activation of immune gene networks was an immune correlate of anti-BCG immunity, prolonged up-regulation of these networks coincided with the development of vaccine-elicited T cell responses after BCG vaccination/infection. These findings provide molecular evidence suggesting that the BCG-induced gene networks may represent global transcriptomes and proteomes underlying the development of T cell responses and, ultimately, immunity to mycobacteria.
Collapse
Affiliation(s)
- Dan Huang
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois at Chicago, Chicago
| | - Liyou Qiu
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois at Chicago, Chicago
| | - Richard Wang
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois at Chicago, Chicago
| | - Xioamin Lai
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois at Chicago, Chicago
| | - George Du
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois at Chicago, Chicago
| | | | - Yun Shen
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois at Chicago, Chicago
| | - Lingyun Shao
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois at Chicago, Chicago
| | - Lisa Halliday
- Biologic Resources Laboratory, University of Illinois at Chicago, Chicago
| | - Jeff Fortman
- Biologic Resources Laboratory, University of Illinois at Chicago, Chicago
| | - Ling Shen
- Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Norman L. Letvin
- Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Zheng W. Chen
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois at Chicago, Chicago
| |
Collapse
|
28
|
Stephens EB, Jackson M, Cui L, Pacyniak E, Choudhuri R, Liverman CS, Salomon DS, Berman NEJ. Early dysregulation of cripto-1 and immunomodulatory genes in the cerebral cortex in a macaque model of neuroAIDS. Neurosci Lett 2006; 410:94-9. [PMID: 17084529 DOI: 10.1016/j.neulet.2006.07.066] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2006] [Revised: 06/15/2006] [Accepted: 07/03/2006] [Indexed: 11/18/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) and related primate lentiviruses are known to enter the central nervous system (CNS) during the primary phase of infection. Neuroinvasion by simian immunodeficiency virus and simian human immunodeficiency virus (SHIV) is characterized by transient meningitis and astrocytosis. In this report, we used targeted cytokine cDNA arrays to analyze cortical brain tissue from four pig-tailed macaques inoculated for 2 weeks with pathogenic SHIV(50OLNV) and a normal age-matched pig-tailed macaque. Our results revealed that eight genes were significantly upregulated in all four macaques. These included: leukocyte interferon inducible peptide, corticotrophin releasing factor receptor 1, interleukin 6, CDW40 antigen, cysteine-rich fibroblast growth factor, neurotrophin 3, ciliary neurotrophin factor receptor and cripto-1. The upregulation of three of these genes was confirmed by reverse transcriptase PCR (RT-PCR). Since cripto-1 had not been previously identified within specific cell types within the primate central nervous system, we performed immunohistochemical studies, which revealed the presence of cripto-1 in neurons. RT-PCR studies demonstrated that cripto-1 mRNA was widely expressed in the CNS. These results indicate that immunomodulatory genes are upregulated during the primary phase of infection of the central nervous system. Cripto-1, which acts as a survival factor in tumor cells and may be neuroprotective, is expressed in neurons within the CNS and is upregulated during viral invasion.
Collapse
Affiliation(s)
- Edward B Stephens
- University of Kansas Medical Center, Anatomy & Cell Biology, 3901 Rainbow Blvd., Kansas City, KS 66160, United States
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Norgren RB. Expression arrays for macaque monkeys. Transplant Rev (Orlando) 2006. [DOI: 10.1016/j.trre.2006.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
30
|
Neuroimmunity and the blood-brain barrier: molecular regulation of leukocyte transmigration and viral entry into the nervous system with a focus on neuroAIDS. J Neuroimmune Pharmacol 2006; 1:160-81. [PMID: 18040782 DOI: 10.1007/s11481-006-9017-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2005] [Accepted: 02/27/2006] [Indexed: 01/07/2023]
Abstract
HIV infection of the central nervous system (CNS) can result in neurologic dysfunction with devastating consequences in a significant number of individuals with AIDS. Two main CNS complications in individuals with HIV are encephalitis and dementia, which are characterized by leukocyte infiltration into the CNS, microglia activation, aberrant chemokine expression, blood-brain barrier (BBB) disruption, and eventual damage and/or loss of neurons. One of the major mediators of NeuroAIDS is the transmigration of HIV-infected leukocytes across the BBB into the CNS. This review summarizes new key findings that support a critical role of the BBB in regulating leukocyte transmigration. In addition, we discuss studies on communication among cells of the immune system, BBB, and the CNS parenchyma, and suggest how these interactions contribute to the pathogenesis of NeuroAIDS. We also describe some of the animal models that have been used to study and characterize important mechanisms that have been proposed to be involved in HIV-induced CNS dysfunction. Finally, we review the pharmacologic interventions that address neuroinflammation, and the effect of substance abuse on HIV-1 related neuroimmunity.
Collapse
|
31
|
Jenner RG, Young RA. Insights into host responses against pathogens from transcriptional profiling. Nat Rev Microbiol 2005; 3:281-94. [PMID: 15806094 DOI: 10.1038/nrmicro1126] [Citation(s) in RCA: 432] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
DNA microarrays have allowed us to monitor the effects of pathogens on host-cell gene expression programmes in great depth and on a broad scale. The comparison of results that have been generated by these studies is complex, and such a comparison has not previously been attempted in a systematic manner. In this review, we have collated and compared published transcriptional-profiling data from 32 studies that involved 77 different host-pathogen interactions, and have defined a common host-transcriptional-response. We outline gene expression patterns in the context of Toll-like receptor and pathogen-mediated signalling pathways, and summarize the contributions that transcriptional-profiling studies have made to our understanding of the infectious disease process.
Collapse
Affiliation(s)
- Richard G Jenner
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, Massachusetts 02142, USA.
| | | |
Collapse
|
32
|
Motomura K, Toyoda N, Oishi K, Sato H, Nagai S, Hashimoto SI, Tugume SB, Enzama R, Mugewa R, Mutuluuza CK, Mugyeyi P, Nagatake T, Matsushima K. Identification of a host gene subset related to disease prognosis of HIV-1 infected individuals. Int Immunopharmacol 2005; 4:1829-36. [PMID: 15531298 DOI: 10.1016/j.intimp.2004.07.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2004] [Revised: 06/08/2004] [Accepted: 07/30/2004] [Indexed: 11/29/2022]
Abstract
Although plasma-virus-RNA level and CD4-positive-T-cell count are useful to monitor clinical status of the human immunodeficiency virus (HIV)-infected individuals, clinical course is often varied among patients and sometimes difficult to predict. To identify additional parameters associated with disease progression, we examined by cDNA microarray the expression profiles of 731 immune-response-related genes in the peripheral blood mononuclear cells (PBMCs) from 21 HIV-positive individuals in Uganda. The analysis enabled the patients to be classified into three distinct groups on the basis of the gene expression patterns. Notably, these groups, clusters I, II and III, were highly associated with clinical status of the patients defined by CDC classification, categories A, B, and C, respectively. Statistical analysis identified 40 genes whose expressions were significantly up- or down-regulated in the cluster III patients (p<0.05). Up- and down-regulated genes included ones involved in immature T lymphocytes differentiation, apoptosis signaling, and active HIV replication, suggesting that the levels of active destruction and regeneration of mature T lymphocytes associated with enhanced HIV-1 replication is related to the disease progression. Follow-up study showed that the cluster classification improved prediction of disease prognosis with the CDC classification. These findings provide new clues for studying perturbation of host immunity, pathogenesis, and disease prognosis of HIV-infected individuals.
Collapse
Affiliation(s)
- Kazushi Motomura
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Sui Y, Li S, Pinson D, Adany I, Li Z, Villinger F, Narayan O, Buch S. Simian human immunodeficiency virus-associated pneumonia correlates with increased expression of MCP-1, CXCL10, and viral RNA in the lungs of rhesus macaques. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:355-65. [PMID: 15681820 PMCID: PMC1602335 DOI: 10.1016/s0002-9440(10)62259-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Pulmonary disorders are the most frequent cause of death in HIV-1-infected individuals with AIDS and remain important even in the current era of potent antiretroviral therapy. Macaques infected with Simian/Human Immunodeficiency Virus (SHIV) develop pulmonary disease and concurrent opportunistic infections similar to those observed in HIV-infected individuals, thereby providing an excellent working model to elucidate the pathogenesis of the human lung disease. Since chemokines play a crucial role in the recruitment of inflammatory cells to tissues, we investigated the relationship between respiratory disease and the levels of chemokines, monocyte chemotactic protein-1 (MCP-1) and CXCL10, in the lungs of SHIV-infected rhesus macaques. We found that lung pathology in infected macaques was closely associated with overexpression of MCP-1 and CXCL10. In addition, these chemokines could, in part, be responsible for the recruitment of inflammatory cells infiltrating into the diseased lungs as demonstrated by chemotactic assays. Lung pathology and increased levels of MCP-1 and CXCL10 correlated with high viral loads in the lung parenchyma. Using confocal microscopy, we identified SHIV-infected macrophages as the major producers of MCP-1 and CXCL10 in the diseased lungs. These data suggest that chemokine overexpression plays an important role in the pathogenesis of SHIV-associated pulmonary disease in macaques.
Collapse
Affiliation(s)
- Yongjun Sui
- Department of Microbiology, Immunology, and Molecular Genetics, Marion Merrell Dow Laboratory of Viral Pathogenesis, 5000 Wahl Hall East, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Buch S, Sui Y, Dhillon N, Potula R, Zien C, Pinson D, Li S, Dhillon S, Nicolay B, Sidelnik A, Li C, Villinger T, Bisarriya K, Narayan O. Investigations on four host response factors whose expression is enhanced in X4 SHIV encephalitis. J Neuroimmunol 2005; 157:71-80. [PMID: 15579283 DOI: 10.1016/j.jneuroim.2004.08.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2004] [Indexed: 11/22/2022]
Abstract
HIV encephalopathy, one of the major complications of HIV infection, involves productive virus replication in macrophages in the brain in association with heightened expression of several host response factors. One or more of these factors are thought to be the cause of the degenerative changes in neurons in the brain. Macaques infected with SIV and SHIV viruses have provided excellent working models for studying mechanisms of the human disease. Although HIV encephalopathy is primarily associated with CCR5-utilizing viruses, our findings have shown that CXCR4-utilizing SHIVs were also capable of causing the syndrome in rhesus macaques. In SHIV-infected macaques, approximately 30% of the animals developed encephalitis. In order to understand the factors leading to end-stage encephalitis, we performed microarray analyses on brains of encephalitic and non-encephalitic-infected macaques, and found pronounced enhancement of expression of interleukin-4, platelet-derived growth factor-B chain, monocyte chemoattractant protein-1 and CXCL10 in the brains of the encephalitic animals. This review discusses the role of each of these factors in mediating SHIV encephalitis.
Collapse
Affiliation(s)
- Shilpa Buch
- Department of Microbiology, Immunology and Molecular Genetics, Marion Merrell Dow Laboratory of Viral Pathogenesis, 5000 Wahl Hall East, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kim SY, Li J, Bentsman G, Brooks AI, Volsky DJ. Microarray analysis of changes in cellular gene expression induced by productive infection of primary human astrocytes: implications for HAD. J Neuroimmunol 2005; 157:17-26. [PMID: 15579276 DOI: 10.1016/j.jneuroim.2004.08.032] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2004] [Indexed: 12/11/2022]
Abstract
The role of astrocytes in HIV-1 associated dementia (HAD) is not well understood. HIV-1 binds efficiently to astrocytes but infects only a small fraction of the cells in vitro and in vivo. To gain insight into the biology of HIV-1-expressing astrocytes, we productively infected human fetal astrocytes with pseudotyped HIV-1 and employed Affymetrix oligonucleotide microarrays to determine global changes in cellular gene expression at the peak of virus production. With a twofold change as a cutoff, HIV-1 increased transcription of 266 genes in astrocytes and suppressed expression of 468. The functions of highly expressed genes included interferon-mediated antiviral responses (OAS1, IFIT1), intercellular contacts (SH3, glia-derived nexin), cell homing/adhesion (matrix metalloproteinases), and cell-cell signaling (neuropilin 1 and 2). Surprisingly, genes involved in innate immune responses of astrocytes were largely unaffected. The single most significant effect of HIV-1, however, was down-modulation of at least 55 genes involved in control of cell cycle, DNA replication, and cell proliferation, which were overrepresented in these categories with probability scores of 10(-10)-10(-26). Our data suggest that HIV-1 expression in astrocytes profoundly alters host cell biology, with potential consequences for the physiological function of astrocytes during HIV-1 infection in the brain.
Collapse
Affiliation(s)
- Seon-Young Kim
- Molecular Virology Division, St. Luke's-Roosevelt Hospital Center, 432 West 58th Street, Antenucci Building, Room 709, New York, NY 10019, USA
| | | | | | | | | |
Collapse
|
36
|
van Marle G, Henry S, Todoruk T, Sullivan A, Silva C, Rourke SB, Holden J, McArthur JC, Gill MJ, Power C. Human immunodeficiency virus type 1 Nef protein mediates neural cell death: a neurotoxic role for IP-10. Virology 2005; 329:302-18. [PMID: 15518810 DOI: 10.1016/j.virol.2004.08.024] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2004] [Revised: 07/22/2004] [Accepted: 08/12/2004] [Indexed: 02/01/2023]
Abstract
HIV-1 Nef is expressed in astrocytes, but a contribution to neuropathogenesis and the development of HIV-associated dementia (HAD) remains uncertain. To determine the neuropathogenic actions of the HIV-1 Nef protein, the brain-derived (YU-2) and blood-derived (NL4-3) Nef proteins were expressed in neural cells using an alphavirus vector, which resulted in astrocyte death (P < 0.001). Supernatants from Nef-expressing astrocytes also caused neuronal death, suggesting the release of neurotoxic molecules by astrocytes. Analysis of pro-inflammatory gene induction in astrocytes expressing Nef revealed increased IP-10 mRNA expression (4000-fold) that was Nef sequence dependent. Recombinant IP-10 caused selective cell death in neurons (P < 0.001) but not astrocytes, and the cytotoxicity of supernatant from astrocytes expressing Nef YU-2 was blocked by an antibody directed against the chemokine receptor CXCR3 (P < 0.001). SCID/NOD mice implanted with a Nef YU-2-expressing vector displayed abnormal motor behavior (P < 0.05), neuroinflammation, and neuronal loss relative to controls. Analysis of mRNA levels in brains from patients with HAD also revealed increased expression of IP-10 (P < 0.05), which was confirmed by immunoreactivity detected principally in astrocytes. Phylogenetic and protein structure analyses of Nef sequences derived from HIV/AIDS patients with and without HAD suggested viral evolution toward a neurotropic Nef protein. These results indicate that HIV-1 Nef contributes to neuropathogenesis by directly causing astrocyte death together with indirect neuronal death through the cytotoxic actions of IP-10 on neurons. Furthermore, Nef molecular diversity was evident in brain tissue among patients with neurological disease and which may influence IP-10 production by astrocytes.
Collapse
MESH Headings
- AIDS Dementia Complex/metabolism
- AIDS Dementia Complex/physiopathology
- Animals
- Animals, Genetically Modified
- Astrocytes/metabolism
- Astrocytes/virology
- Cell Death
- Cells, Cultured
- Chemokine CCL2/biosynthesis
- Chemokine CCL2/genetics
- Chemokine CXCL10
- Chemokines, CXC/biosynthesis
- Chemokines, CXC/pharmacology
- Chemokines, CXC/physiology
- Gene Products, nef/biosynthesis
- Gene Products, nef/genetics
- Genetic Vectors
- HIV-1/genetics
- HIV-1/pathogenicity
- Humans
- Interleukin-1/biosynthesis
- Interleukin-1/genetics
- Interleukin-1beta
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Molecular Sequence Data
- Neurons/drug effects
- Neurons/metabolism
- Neurons/pathology
- Neurotoxins/pharmacology
- Peptide Fragments/biosynthesis
- Peptide Fragments/genetics
- RNA, Messenger/analysis
- Recombinant Proteins/pharmacology
- nef Gene Products, Human Immunodeficiency Virus
Collapse
Affiliation(s)
- Guido van Marle
- Department of Clinical Neurosciences, University of Calgary, Calgary AB, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Speth C, Dierich MP, Sopper S. HIV-infection of the central nervous system: the tightrope walk of innate immunity. Mol Immunol 2005; 42:213-28. [PMID: 15488609 DOI: 10.1016/j.molimm.2004.06.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Infection of the central nervous system (CNS) by HIV is a frequent and sometimes very early event in the course of HIV pathogenesis. Possible consequences are diverse symptoms of neurological dysfunction, but also the establishment of a lifelong latent viral reservoir in the brain. Whereas in the periphery innate and adaptive immunity are equal partners, the blood-brain barrier (BBB) with its restricted access of peripheral immune effectors shifts this balance in favour of the local innate immunity. Four main elements of cerebral innate immunity are discussed in the present article, including two cell types with immunological functions and two soluble immune systems: (1) the stimulation of microglial cells as the predominant brain-resident immune cell and the main local reservoir for the virus; (2) the reaction of astrocytes in response to viral infection; (3) the activation of the local complement system as important soluble immune cascade; and (4) the role of chemokines and cytokines which help to conduct and cross-link the interplay between the different immune elements. These components of the cerebral innate immunity do not act separately from each other but form a functional immunity network. A dual role of these components with both harmful and protective effects further enhances the complexity of the mutual interactions.
Collapse
Affiliation(s)
- Cornelia Speth
- Institute of Hygiene and Social Medicine, Medical University Innsbruck and Ludwig-Boltzmann-Institute for AIDS Research, Fritz-Pregl-Str. 3, A-6020 Innsbruck, Austria.
| | | | | |
Collapse
|
38
|
Minagar A, Shapshak P, Duran EM, Kablinger AS, Alexander JS, Kelley RE, Seth R, Kazic T. HIV-associated dementia, Alzheimer's disease, multiple sclerosis, and schizophrenia: gene expression review. J Neurol Sci 2004; 224:3-17. [PMID: 15450765 DOI: 10.1016/j.jns.2004.06.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2004] [Revised: 06/15/2004] [Accepted: 06/16/2004] [Indexed: 12/18/2022]
Abstract
RNA and protein gene expression technologies are revolutionizing our view and understanding of human diseases and enable us to analyze the concurrent expression patterns of large numbers of genes. These new technologies allow simultaneous study of thousands of genes and their changes in regulation and modulation patterns in relation to disease state, time, and tissue specificity. This review summarizes the application of this modern technology to four common neurological and psychiatric disorders: HIV-1-associated dementia, Alzheimer's disease, multiple sclerosis, and schizophrenia and is a first comparison of these diseases using this approach.
Collapse
Affiliation(s)
- Alireza Minagar
- Department of Neurology, Louisiana State University School of Medicine, Shreveport 71130, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Dhillon NK, Sui Y, Potula R, Dhillon S, Adany I, Li Z, Villinger F, Pinson D, Narayan O, Buch S. Inhibition of pathogenic SHIV replication in macaques treated with antisense DNA of interleukin-4. Blood 2004; 105:3094-9. [PMID: 15618469 DOI: 10.1182/blood-2004-09-3515] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interleukin-4 is implicated in the pathogenesis of HIV-induced AIDS and causes enhancement of replication of virus strains that use the CXCR4 (X4) coreceptor. In this study, we explored the effects of interleukin-4 (IL-4) antisense (AS) DNA on replication of X4, simian human immunodeficiency viruses, SHIV(KU-2) and SHIV89.6P. AS IL-4 oligomer caused inhibition of virus replication in cultures of CD4+ T cells and macrophages derived from macaques. Plasmid expressing AS IL-4 DNA was also effective in abrogating virus replication in macrophage cultures. Relevance of these cell culture studies was confirmed in vivo by treating SHIV89.6P-infected macaques with AS IL-4 DNA. Six macaques were inoculated with the virus, and 4 were treated with AS IL-4 DNA. This resulted in a significant decrease in viral RNA concentrations in the liver, lungs, and spleen tissues that are all sites of virus replication in macrophages. This is the first demonstration of effective inhibition of an HIV-like virus in tissues by AS DNA of a cytokine. In the present era of increasing resistance of HIV to antiviral compounds, exploration of adjunct therapies directed at host responses in combination with antiretroviral drugs may be of value for the treatment of AIDS.
Collapse
Affiliation(s)
- Navneet Kaur Dhillon
- Department of Microbiology, Immunology and Molecular Genetics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Peterson KE, Hughes S, Dimcheff DE, Wehrly K, Chesebro B. Separate sequences in a murine retroviral envelope protein mediate neuropathogenesis by complementary mechanisms with differing requirements for tumor necrosis factor alpha. J Virol 2004; 78:13104-12. [PMID: 15542662 PMCID: PMC525006 DOI: 10.1128/jvi.78.23.13104-13112.2004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The innate immune response, through the induction of proinflammatory cytokines and antiviral factors, plays an important role in protecting the host from pathogens. Several components of the innate response, including tumor necrosis factor alpha (TNF-alpha), monocyte chemoattractant protein 1, interferon-inducible protein 10, and RANTES, are upregulated in the brain following neurovirulent retrovirus infection in humans and in animal models. However, it remains unclear whether this immune response is protective, pathogenic, or both. In the present study, by using TNF-alpha(-/-) mice we analyzed the contribution of TNF-alpha to neurological disease induced by four neurovirulent murine retroviruses, with three of these viruses encoding portions of the same neurovirulent envelope protein. Surprisingly, only one retrovirus (EC) required TNF-alpha for disease induction, and this virus induced less TNF-alpha expression in the brain than did the other retroviruses. Analysis of glial fibrillary acidic protein and F4/80 in EC-infected TNF-alpha(-/-) mice showed normal activation of astrocytes but not of microglia. Thus, TNF-alpha-mediated microglial activation may be important in the pathogenic process initiated by EC infection. In contrast, TNF-alpha was not required for pathogenesis of the closely related BE virus and the BE virus induced disease in TNF-alpha(-/-) mice by a different mechanism that did not require microglial activation. These results provide new insights into the multifactorial mechanisms involved in retrovirus-induced neurodegeneration and may also have analogies to other types of neurodegeneration.
Collapse
Affiliation(s)
- Karin E Peterson
- Department of Pathobiological Sciences, School of Veterinary Medicine, Skip Bertman Dr., Louisiana State University, Baton Rouge, LA 70803, USA.
| | | | | | | | | |
Collapse
|
41
|
Albright AV, González-Scarano F. Microarray analysis of activated mixed glial (microglia) and monocyte-derived macrophage gene expression. J Neuroimmunol 2004; 157:27-38. [PMID: 15579277 DOI: 10.1016/j.jneuroim.2004.09.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Since macrophage activation can now be studied at a global level using modern microarray and proteomic analyses, discovery of novel macrophage activation genes is inevitable and important for understanding HIV-associated dementia (HAD). We isolated two different types of primary human macrophages: microglia and monocyte-derived macrophages (MDM) from brain tissue and whole blood, respectively. The microarray analysis of differentially regulated macrophage activation genes reported here supports our previous assertions that the mixed glia (MIX) cultured in starvation conditions (DMEM alone) are a non-activated, or "quiescent", tissue culture model for studying macrophage activation in the brain. Transcript levels from these quiescent cultures provided a background level of gene expression and allowed for the identification of upregulated macrophage activation genes in the MIX brain cultures upon treatment with an array of soluble activation factors: serum components, cytokines, and growth factors. We found that 914 genes in the MIX cultures and 734 genes in the MDM cultures had a greater than twofold increase in expression. We discovered 180 genes with expression that was increased more than twofold in both culture types. Microarray-specific statistical analyses were performed to complement fold change analysis: significance analysis of microarrays (SAM) and Partek Pro. In the MIX cultures, we detected over a 100-fold increase in IL-1beta and TIMP1 transcription; Caspase 9, S100A8 and 9, MMP12, IL-8, monocyte chemotactic protein 1 (MCP1), MRC-1, and IL-6 were also upregulated. Activation of starved MDM cultures resulted in fewer upregulated genes compared to MIX cultures. Genes upregulated in both MIX and MDM included CCL2 (MCP1), CCL7, CXCL5, TNFSF14, kinases, and phosphatases. These microarray data may provide leads for identifying previously unknown neurotoxins, disease biomarkers, and pathways responsible for the neuronal apoptosis observed in HAD and for the eventual identification of therapeutic targets and treatments.
Collapse
Affiliation(s)
- Andrew V Albright
- Department of Neurology, University of Pennsylvania, 255 Clinical Research Building, 415 Curie Boulevard, Philadelphia, PA 19104-6146, USA.
| | | |
Collapse
|
42
|
Potula R, Dhillion N, Sui Y, Zien CA, Funa K, Pinson D, Mayo MS, Singh DK, Narayan O, Buch S. Association of platelet-derived growth factor-B chain with simian human immunodeficiency virus encephalitis. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:815-24. [PMID: 15331406 PMCID: PMC1618609 DOI: 10.1016/s0002-9440(10)63344-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chemokines and cytokines play a critical role in HIV infection, serving both to modulate virus replication and to recruit target cells to the site of infection. Platelet-derived growth factor (PDGF), a mitogen and chemoattractant for a wide variety of cells, is secreted by macrophages. Since macrophages are the target cells for lentiviral infection in the brain and PDGF is a known inducer of macrophage chemoattractant protein-1 (MCP)-1, a potent chemokine closely associated with HIV encephalitis, we investigated the association of PDGF-B chain (PDGF-B) with encephalitis in macaques caused by simian human immunodeficiency virus (SHIV), a chimera of HIV and SIV. Northern blot analysis confirmed elevated expression of PDGF-B chain mRNA in the brains from encephalitic macaques. Validation of these in vivo studies was confirmed in rhesus macrophage cultures infected with SHIV(KU2) in which we demonstrated heightened expression of PDGF-B chain mRNA. Nuclear run-off analysis established transcriptional up-regulation of PDGF-B chain in virus-inoculated macrophage cultures. Reciprocally, addition of exogenous PDGF enhanced virus replication and MCP-1 expression in these cells. Inhibition of virus replication by tyrosine kinase inhibitor, STI-571, and by PDGF-B antisense oligonucleotides confirmed the specificity of the PDGF effect. Relevance of these findings was confirmed by analysis of archival brain tissue from SHIV encephalitic and non-encephalitic macaques for PDGF-B chain expression. PDGF-B chain protein expression was observed in the virus-infected cells in microglial nodules in the brains of SHIV-encephalitic macaques.
Collapse
Affiliation(s)
- Raghava Potula
- Department of Microbiology, Immunology, and Molecular Genetics, Marion Merrell Dow Laboratory of Viral Pathogenesis, Kansas City, Kansas, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Sui Y, Potula R, Dhillon N, Pinson D, Li S, Nath A, Anderson C, Turchan J, Kolson D, Narayan O, Buch S. Neuronal apoptosis is mediated by CXCL10 overexpression in simian human immunodeficiency virus encephalitis. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:1557-66. [PMID: 15111302 PMCID: PMC1615658 DOI: 10.1016/s0002-9440(10)63714-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inflammatory mediators play a crucial role in the pathophysiology of several neurodegenerative diseases including acquired immune deficiency syndrome dementia complex. In the present study we identified a link between CXCL10 overexpression in the brain and human immunodeficiency virus dementia and demonstrated the presence of the chemokine CXCL10 and its receptor, CXCR3, in the neurons in the brains of macaques with simian human immunodeficiency virus encephalitis. Using human fetal brain cultures, we showed that treatment of these cells with either SHIV89.6P or viral gp120 resulted in induction of CXCL10 in neurons. Cultured neurons treated with the chemokine developed increased membrane permeability followed by apoptosis via activation of caspase-3. We confirmed the relevance of these findings in sections of human and macaque brains with encephalopathy demonstrating that neurons expressing CXCL10 also expressed caspase-3.
Collapse
Affiliation(s)
- Yongjun Sui
- Department of Microbiology, Immunology, and Molecular Genetics, Marion Merrell Dow Laboratory of Viral Pathogenesis, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|