1
|
Sun Y, Cai M, Liang Y, Zhang Y. Disruption of blood-brain barrier: effects of HIV Tat on brain microvascular endothelial cells and tight junction proteins. J Neurovirol 2023; 29:658-668. [PMID: 37899420 DOI: 10.1007/s13365-023-01179-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/01/2023] [Accepted: 10/12/2023] [Indexed: 10/31/2023]
Abstract
Although the widespread use of antiretroviral therapy (ART) has prolonged the life span of people living with HIV (PLWH), the incidence of HIV-associated neurocognitive disorders (HAND) in PLWH is also gradually increasing, seriously affecting the quality of life for PLWH. However, the pathogenesis of HAND has not been elucidated, which leaves HAND without effective treatment. HIV protein transactivator of transcription (Tat), as an important regulatory protein, is crucial in the pathogenesis of HAND, and its mechanism of HAND has received widespread attention. The blood-brain barrier (BBB) and its cellular component brain microvascular endothelial cells (BMVECs) play a necessary role in protecting the central nervous system (CNS), and their damage associated with Tat is a potential therapeutic target of HAND. In this review, we will study the Tat-mediated damage mechanism of the BBB and present multiple lines of evidence related to BMVEC damage caused by Tat.
Collapse
Affiliation(s)
- Yuqing Sun
- Department of Respiratory and Critical Care Medicine, Beijing You An Hospital, Capital Medical University, Beijing, 100069, China
| | - Miaotian Cai
- Department of Respiratory and Critical Care Medicine, Beijing You An Hospital, Capital Medical University, Beijing, 100069, China
| | - Ying Liang
- Department of Respiratory and Critical Care Medicine, Beijing You An Hospital, Capital Medical University, Beijing, 100069, China
| | - Yulin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing You An Hospital, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
2
|
Sonti S, Tyagi K, Pande A, Daniel R, Sharma AL, Tyagi M. Crossroads of Drug Abuse and HIV Infection: Neurotoxicity and CNS Reservoir. Vaccines (Basel) 2022; 10:vaccines10020202. [PMID: 35214661 PMCID: PMC8875185 DOI: 10.3390/vaccines10020202] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 01/27/2023] Open
Abstract
Drug abuse is a common comorbidity in people infected with HIV. HIV-infected individuals who abuse drugs are a key population who frequently experience suboptimal outcomes along the HIV continuum of care. A modest proportion of HIV-infected individuals develop HIV-associated neurocognitive issues, the severity of which further increases with drug abuse. Moreover, the tendency of the virus to go into latency in certain cellular reservoirs again complicates the elimination of HIV and HIV-associated illnesses. Antiretroviral therapy (ART) successfully decreased the overall viral load in infected people, yet it does not effectively eliminate the virus from all latent reservoirs. Although ART increased the life expectancy of infected individuals, it showed inconsistent improvement in CNS functioning, thus decreasing the quality of life. Research efforts have been dedicated to identifying common mechanisms through which HIV and drug abuse lead to neurotoxicity and CNS dysfunction. Therefore, in order to develop an effective treatment regimen to treat neurocognitive and related symptoms in HIV-infected patients, it is crucial to understand the involved mechanisms of neurotoxicity. Eventually, those mechanisms could lead the way to design and develop novel therapeutic strategies addressing both CNS HIV reservoir and illicit drug use by HIV patients.
Collapse
Affiliation(s)
- Shilpa Sonti
- Center for Translational Medicine, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA; (S.S.); (A.L.S.)
| | - Kratika Tyagi
- Department of Biotechnology, Banasthali Vidyapith, Vanasthali, Jaipur 304022, Rajasthan, India;
| | - Amit Pande
- Cell Culture Laboratory, ICAR-Directorate of Coldwater Fisheries Research, Bhimtal, Nainital 263136, Uttarakhand, India;
| | - Rene Daniel
- Farber Hospitalist Service, Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Adhikarimayum Lakhikumar Sharma
- Center for Translational Medicine, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA; (S.S.); (A.L.S.)
| | - Mudit Tyagi
- Center for Translational Medicine, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA; (S.S.); (A.L.S.)
- Correspondence: ; Tel.: +1-215-503-5157 or +1-703-909-9420
| |
Collapse
|
3
|
Marino J, Maubert ME, Mele AR, Spector C, Wigdahl B, Nonnemacher MR. Functional impact of HIV-1 Tat on cells of the CNS and its role in HAND. Cell Mol Life Sci 2020; 77:5079-5099. [PMID: 32577796 PMCID: PMC7674201 DOI: 10.1007/s00018-020-03561-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/08/2020] [Accepted: 05/25/2020] [Indexed: 02/07/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) transactivator of transcription (Tat) is a potent mediator involved in the development of HIV-1-associated neurocognitive disorders (HAND). Tat is expressed even in the presence of antiretroviral therapy (ART) and is able to enter the central nervous system (CNS) through a variety of ways, where Tat can interact with microglia, astrocytes, brain microvascular endothelial cells, and neurons. The presence of low concentrations of extracellular Tat alone has been shown to lead to dysregulated gene expression, chronic cell activation, inflammation, neurotoxicity, and structural damage in the brain. The reported effects of Tat are dependent in part on the specific HIV-1 subtype and amino acid length of Tat used. HIV-1 subtype B Tat is the most common subtype in North American and therefore, most studies have been focused on subtype B Tat; however, studies have shown many genetic, biologic, and pathologic differences between HIV subtype B and subtype C Tat. This review will focus primarily on subtype B Tat where the full-length protein is 101 amino acids, but will also consider variants of Tat, such as Tat 72 and Tat 86, that have been reported to exhibit a number of distinctive activities with respect to mediating CNS damage and neurotoxicity.
Collapse
Affiliation(s)
- Jamie Marino
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Monique E Maubert
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Anthony R Mele
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Cassandra Spector
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Michael R Nonnemacher
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA.
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
4
|
MRS suggests multi-regional inflammation and white matter axonal damage at 11 years following perinatal HIV infection. NEUROIMAGE-CLINICAL 2020; 28:102505. [PMID: 33395994 PMCID: PMC7721646 DOI: 10.1016/j.nicl.2020.102505] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/30/2022]
Abstract
The neurological changes in children living with perinatal HIV (PHIV) on antiretroviral therapy (ART) can be studied at a metabolic level through proton magnetic resonance spectroscopy. While previous studies in children have largely focused on individual metabolite changes, investigating patterns within and across regions of interest can aid in identifying metabolic markers of HIV infection. In this study 76 children with PHIV from the Children with HIV Early AntiRetroviral (CHER) trial, 30 children who were HIV-exposed-uninfected (HEU) and 30 children who were HIV-unexposed (HU), were scanned at the age of 11.6 (sd = 0.3) years using a 3 T Skyra scanner. Metabolite concentrations were quantified within the basal ganglia (BG), midfrontal gray matter (MFGM) and peritrigonal white matter (PWM), comparing levels between HIV status groups using linear regression. Factor analysis and logistic regression were performed to identify metabolic patterns characteristic of HIV infection within and across the regions of interest. In the BG region we observed restored metabolic activity in children with PHIV and children who were HEU, despite differences being previously observed at younger ages, suggesting that treatment may effectively reduce the effects of HIV infection and exposure. Elevated MFGM choline levels in children with PHIV are indicative of inflammation. Further, we observed reduced N-acetyl-aspartate (NAA) in the PWM of children with PHIV and children who were HEU, indicating possible axonal damage. Lower levels of PWM creatine in children with PHIV suggest that this may not be a valid reference metabolite in HIV studies. Finally, factor scores for a cross-regional inflammatory factor and a PWM axonal factor, driven by PWM NAA and creatine levels, distinguished children with PHIV from children without HIV (HEU and HU) at 11 years. Therefore, the effects of perinatal HIV infection and exposure continue to be seen at 11 years despite early treatment.
Collapse
|
5
|
Ajasin D, Eugenin EA. HIV-1 Tat: Role in Bystander Toxicity. Front Cell Infect Microbiol 2020; 10:61. [PMID: 32158701 PMCID: PMC7052126 DOI: 10.3389/fcimb.2020.00061] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/06/2020] [Indexed: 12/21/2022] Open
Abstract
HIV Tat protein is a critical protein that plays multiple roles in HIV pathogenesis. While its role as the transactivator of HIV transcription is well-established, other non-viral replication-associated functions have been described in several HIV-comorbidities even in the current antiretroviral therapy (ART) era. HIV Tat protein is produced and released into the extracellular space from cells with active HIV replication or from latently HIV-infected cells into neighboring uninfected cells even in the absence of active HIV replication and viral production due to effective ART. Neighboring uninfected and HIV-infected cells can take up the released Tat resulting in the upregulation of inflammatory genes and activation of pathways that leads to cytotoxicity observed in several comorbidities such as HIV associated neurocognitive disorder (HAND), HIV associated cardiovascular impairment, and accelerated aging. Thus, understanding how Tat modulates host and viral response is important in designing novel therapeutic approaches to target the chronic inflammatory effects of soluble viral proteins in HIV infection.
Collapse
Affiliation(s)
- David Ajasin
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX, United States
| | - Eliseo A Eugenin
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
6
|
Welch JL, Kaufman TM, Stapleton JT, Okeoma CM. Semen exosomes inhibit HIV infection and HIV-induced proinflammatory cytokine production independent of the activation state of primary lymphocytes. FEBS Lett 2019; 594:695-709. [PMID: 31665815 DOI: 10.1002/1873-3468.13653] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 10/09/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022]
Abstract
Semen exosomes (SE) inhibit HIV infection. However, the effect of SE on cell activation and inflammation remains unknown. We characterized the response of peripheral blood mononuclear cells (PBMCs) from HIV-uninfected and antiretroviral therapy-suppressed HIV-infected (HIV+) subjects to SE. Quiescent PBMCs or T-cell receptor (TCR)-activated PBMCs from HIV- and HIV+ donors were stimulated with SE in the presence/absence of ex vivo HIV infection. In HIV-infected PBMCs, SE did not reactivate HIV, did not induce lymphoblast development, nor increase CD69+/CD25+ numbers. Furthermore, SE inhibited de novo HIV infection without altering cell activation. SE also asynchronously downregulated HIV-inducible IL1β, IL8, and TNFα and upregulated CXCL10. These data suggest that SE inhibits HIV infection and production of HIV-induced proinflammatory cytokines while preserving lymphocyte activation.
Collapse
Affiliation(s)
- Jennifer L Welch
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Medical Service, Iowa City Veterans Affairs Medical Center, IA, USA
| | - Thomas M Kaufman
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Medical Service, Iowa City Veterans Affairs Medical Center, IA, USA
| | - Jack T Stapleton
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Medical Service, Iowa City Veterans Affairs Medical Center, IA, USA
| | - Chioma M Okeoma
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, NY, USA
| |
Collapse
|
7
|
CCR5: Established paradigms and new frontiers for a 'celebrity' chemokine receptor. Cytokine 2019; 109:81-93. [PMID: 29903576 DOI: 10.1016/j.cyto.2018.02.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/14/2018] [Accepted: 02/16/2018] [Indexed: 01/04/2023]
Abstract
Because of the level of attention it received due to its role as the principal HIV coreceptor, CCR5 has been described as a 'celebrity' chemokine receptor. Here we describe the development of CCR5 inhibitory strategies that have been developed for HIV therapy and which are now additionally being considered for use in HIV prevention and cure. The wealth of CCR5-related tools that have been developed during the intensive investigation of CCR5 as an HIV drug target can now be turned towards the study of CCR5 as a model chemokine receptor. We also summarize what is currently known about the cell biology and pharmacology of CCR5, providing an update on new areas of investigation that have emerged in recent research. Finally, we discuss the potential of CCR5 as a drug target for diseases other than HIV, discussing the evidence linking CCR5 and its natural chemokine ligands with inflammatory diseases, particularly neuroinflammation, and certain cancers. These pathologies may provide new uses for the strategies for CCR5 blockade originally developed to combat HIV/AIDS.
Collapse
|
8
|
A central role for glial CCR5 in directing the neuropathological interactions of HIV-1 Tat and opiates. J Neuroinflammation 2018; 15:285. [PMID: 30305110 PMCID: PMC6180355 DOI: 10.1186/s12974-018-1320-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 09/19/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The collective cognitive and motor deficits known as HIV-associated neurocognitive disorders (HAND) remain high even among HIV+ individuals whose antiretroviral therapy is optimized. HAND is worsened in the context of opiate abuse. The mechanism of exacerbation remains unclear but likely involves chronic immune activation of glial cells resulting from persistent, low-level exposure to the virus and viral proteins. We tested whether signaling through C-C chemokine receptor type 5 (CCR5) contributes to neurotoxic interactions between HIV-1 transactivator of transcription (Tat) and opiates and explored potential mechanisms. METHODS Neuronal survival was tracked in neuronal and glial co-cultures over 72 h of treatment with HIV-1 Tat ± morphine using cells from CCR5-deficient and wild-type mice exposed to the CCR5 antagonist maraviroc or exogenously-added BDNF (analyzed by repeated measures ANOVA). Intracellular calcium changes in response to Tat ± morphine ± maraviroc were assessed by ratiometric Fura-2 imaging (analyzed by repeated measures ANOVA). Release of brain-derived neurotrophic factor (BDNF) and its precursor proBDNF from CCR5-deficient and wild-type glia was measured by ELISA (analyzed by two-way ANOVA). Levels of CCR5 and μ-opioid receptor (MOR) were measured by immunoblotting (analyzed by Student's t test). RESULTS HIV-1 Tat induces neurotoxicity, which is greatly exacerbated by morphine in wild-type cultures expressing CCR5. Loss of CCR5 from glia (but not neurons) eliminated neurotoxicity due to Tat and morphine interactions. Unexpectedly, when CCR5 was lost from glia, morphine appeared to entirely protect neurons from Tat-induced toxicity. Maraviroc pre-treatment similarly eliminated neurotoxicity and attenuated neuronal increases in [Ca2+]i caused by Tat ± morphine. proBDNF/BDNF ratios were increased in conditioned media from Tat ± morphine-treated wild-type glia compared to CCR5-deficient glia. Exogenous BDNF treatments mimicked the pro-survival effect of glial CCR5 deficiency against Tat ± morphine. CONCLUSIONS Our results suggest a critical role for glial CCR5 in mediating neurotoxic effects of HIV-1 Tat and morphine interactions on neurons. A shift in the proBDNF/BDNF ratio that favors neurotrophic support may occur when glial CCR5 signaling is blocked. Some neuroprotection occurred only in the presence of morphine, suggesting that loss of CCR5 may fundamentally change signaling through the MOR in glia.
Collapse
|
9
|
Mechanisms of neuropathogenesis in HIV and HCV: similarities, differences, and unknowns. J Neurovirol 2018; 24:670-678. [PMID: 30291565 DOI: 10.1007/s13365-018-0678-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/20/2018] [Accepted: 09/07/2018] [Indexed: 12/17/2022]
Abstract
HIV and hepatitis C virus (HCV) have both been associated with cognitive impairment. Combination antiretroviral therapy (cART) has dramatically changed the nature of cognitive impairment in HIV-infected persons, while the role of direct-acting antivirals (DAA) in neurocognition of HCV-infected individuals remains unclear. Also, whether HIV and HCV interact to promote neurocognitive decline or whether they each contribute an individual effect continues to be an open question. In this work, we review the virally mediated mechanisms of HIV- and HCV-mediated neuropathogenesis, with an emphasis on the role of dual infection, and discuss observed changes with HIV viral suppression and HCV functional cure on neurocognitive impairments.
Collapse
|
10
|
Abstract
Human immunodeficiency virus (HIV)-associated neurocognitive disorders (HAND) remain a common end-organ manifestation of viral infection. Subclinical and mild symptoms lead to neurocognitive and behavioral abnormalities. These are associated, in part, with viral penetrance and persistence in the central nervous system. Infections of peripheral blood monocytes, macrophages, and microglia are the primary drivers of neuroinflammation and neuronal impairments. While current antiretroviral therapy (ART) has reduced the incidence of HIV-associated dementia, milder forms of HAND continue. Depression, comorbid conditions such as infectious liver disease, drugs of abuse, antiretroviral drugs themselves, age-related neurodegenerative diseases, gastrointestinal maladies, and concurrent social and economic issues can make accurate diagnosis of HAND challenging. Increased life expectancy as a result of ART clearly creates this variety of comorbid conditions that often blur the link between the virus and disease. With the discovery of novel biomarkers, neuropsychologic testing, and imaging techniques to better diagnose HAND, the emergence of brain-penetrant ART, adjunctive therapies, longer life expectancy, and better understanding of disease pathogenesis, disease elimination is perhaps a realistic possibility. This review focuses on HIV-associated disease pathobiology with an eye towards changing trends in the face of widespread availability of ART.
Collapse
|
11
|
Santomasso BD, Park JH, Salloum D, Riviere I, Flynn J, Mead E, Halton E, Wang X, Senechal B, Purdon T, Cross JR, Liu H, Vachha B, Chen X, DeAngelis LM, Li D, Bernal Y, Gonen M, Wendel HG, Sadelain M, Brentjens RJ. Clinical and Biological Correlates of Neurotoxicity Associated with CAR T-cell Therapy in Patients with B-cell Acute Lymphoblastic Leukemia. Cancer Discov 2018; 8:958-971. [PMID: 29880584 DOI: 10.1158/2159-8290.cd-17-1319] [Citation(s) in RCA: 683] [Impact Index Per Article: 97.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 05/01/2018] [Accepted: 06/06/2018] [Indexed: 11/16/2022]
Abstract
CD19-specific chimeric antigen receptor (CAR) T-cell therapy is highly effective against relapsed or refractory acute lymphoblastic leukemia (ALL), but is hindered by neurotoxicity. In 53 adult patients with ALL, we found a significant association of severe neurotoxicity with high pretreatment disease burden, higher peak CAR T-cell expansion, and early and higher elevations of proinflammatory cytokines in blood. Patients with severe neurotoxicity had evidence of blood-cerebrospinal fluid (CSF) barrier disruption correlating with neurotoxicity grade without association with CSF white blood cell count or CAR T-cell quantity in CSF. Proinflammatory cytokines were enriched in CSF during severe neurotoxicity with disproportionately high levels of IL6, IL8, MCP1, and IP10, suggesting central nervous system-specific production. Seizures, seizure-like activity, myoclonus, and neuroimaging characteristics suggested excitatory neurotoxicity, and we found elevated levels of endogenous excitatory agonists in CSF during neurotoxicity.Significance: We detail the neurologic symptoms and blood, CSF, and neuroimaging correlates of neurotoxicity associated with CD19 CAR T cells and identify neurotoxicity risk factors. Our findings implicate cellular components other than T cells and suggest novel links between systemic inflammation and characteristic neurotoxicity symptoms. Cancer Discov; 8(8); 958-71. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 899.
Collapse
Affiliation(s)
- Bianca D Santomasso
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York.,Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Jae H Park
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. .,Department of Medicine, Joan and Sanford Weill Medical College of Cornell University, New York, New York.,Center for Cellular Therapy, Memorial Sloan Kettering Cancer Center, New York, New York.,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Darin Salloum
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Isabelle Riviere
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York.,Michael G. Harris Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jessica Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elena Mead
- Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elizabeth Halton
- Center for Cellular Therapy, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Nursing, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Xiuyan Wang
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York.,Michael G. Harris Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brigitte Senechal
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York.,Michael G. Harris Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Terence Purdon
- Center for Cellular Therapy, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Justin R Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hui Liu
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Behroze Vachha
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Xi Chen
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lisa M DeAngelis
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniel Li
- Juno Therapeutics, Seattle, Washington
| | - Yvette Bernal
- Center for Cellular Therapy, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mithat Gonen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hans-Guido Wendel
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michel Sadelain
- Center for Cellular Therapy, Memorial Sloan Kettering Cancer Center, New York, New York.,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Renier J Brentjens
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. .,Department of Medicine, Joan and Sanford Weill Medical College of Cornell University, New York, New York.,Center for Cellular Therapy, Memorial Sloan Kettering Cancer Center, New York, New York.,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
12
|
HIV-1 Tat inhibits EAAT-2 through AEG-1 upregulation in models of HIV-associated neurocognitive disorder. Oncotarget 2018; 8:39922-39934. [PMID: 28404980 PMCID: PMC5503662 DOI: 10.18632/oncotarget.16485] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 01/23/2017] [Indexed: 11/25/2022] Open
Abstract
During HIV-associated neurocognitive disorder (HAND), decreasing in excitatory amino acid transporter 2 (EAAT-2) in astrocyte plasma membranes leads to elevated levels of extracellular glutamate and, in turn, neuronal apoptosis. We used immunohistochemistry, western blot, qRT-PCR, and RNA interference to elucidate the molecular mechanisms underlying the decreased EAAT-2 expression during HAND at the tissue and cellular levels. We used simian immunodeficiency virus-human immunodeficiency virus chimeric virus (SHIV)-infected macaques as an in vivo model of HAND. Our results show that EAAT-2 expression was decreased in the cerebral cortex, while AEG-1 expression was increased, and the expression levels of these proteins were negatively correlated. In vitro analyses showed that HIV-1 Tat inhibited EAAT-2 expression by inducing overexpression of AEG-1. More specifically, HIV-1 Tat increased AEG-1 expression via the PI3-K signaling pathway, while increasing EAAT-2 inhibition by YinYan-1 (YY-1) via the NF-κB signaling pathway. These results warrant testing AEG-1 as a potential therapeutic target for treating HAND.
Collapse
|
13
|
Pittaluga A. CCL5-Glutamate Cross-Talk in Astrocyte-Neuron Communication in Multiple Sclerosis. Front Immunol 2017; 8:1079. [PMID: 28928746 PMCID: PMC5591427 DOI: 10.3389/fimmu.2017.01079] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/18/2017] [Indexed: 12/17/2022] Open
Abstract
The immune system (IS) and the central nervous system (CNS) are functionally coupled, and a large number of endogenous molecules (i.e., the chemokines for the IS and the classic neurotransmitters for the CNS) are shared in common between the two systems. These interactions are key elements for the elucidation of the pathogenesis of central inflammatory diseases. In recent years, evidence has been provided supporting the role of chemokines as modulators of central neurotransmission. It is the case of the chemokines CCL2 and CXCL12 that control pre- and/or post-synaptically the chemical transmission. This article aims to review the functional cross-talk linking another endogenous pro-inflammatory factor released by glial cells, i.e., the chemokine Regulated upon Activation Normal T-cell Expressed and Secreted (CCL5) and the principal neurotransmitter in CNS (i.e., glutamate) in physiological and pathological conditions. In particular, the review discusses preclinical data concerning the role of CCL5 as a modulator of central glutamatergic transmission in healthy and demyelinating disorders. The CCL5-mediated control of glutamate release at chemical synapses could be relevant either to the onset of psychiatric symptoms that often accompany the development of multiple sclerosis (MS), but also it might indirectly give a rationale for the progression of inflammation and demyelination. The impact of disease-modifying therapies for the cure of MS on the endogenous availability of CCL5 in CNS will be also summarized. We apologize in advance for omission in our coverage of the existing literature.
Collapse
Affiliation(s)
- Anna Pittaluga
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| |
Collapse
|
14
|
Hu XT. HIV-1 Tat-Mediated Calcium Dysregulation and Neuronal Dysfunction in Vulnerable Brain Regions. Curr Drug Targets 2016; 17:4-14. [PMID: 26028040 DOI: 10.2174/1389450116666150531162212] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 05/26/2015] [Indexed: 01/08/2023]
Abstract
Despite the success of combined antiretroviral therapy, more than half of HIV-1-infected patients in the USA show HIV-associated neurological and neuropsychiatric deficits. This is accompanied by anatomical and functional alterations in vulnerable brain regions of the mesocorticolimbic and nigrostriatal systems that regulate cognition, mood and motivation-driven behaviors, and could occur at early stages of infection. Neurons are not infected by HIV, but HIV-1 proteins (including but not limited to the HIV-1 trans-activator of transcription, Tat) induce Ca(2+) dysregulation, indicated by abnormal and excessive Ca(2+) influx and increased intracellular Ca(2+) release that consequentially elevate cytosolic free Ca(2+) levels ([Ca(2+)]in). Such alterations in intracellular Ca(2+) homeostasis significantly disturb normal functioning of neurons, and induce dysregulation, injury, and death of neurons or non-neuronal cells, and associated tissue loss in HIV-vulnerable brain regions. This review discusses certain unique mechanisms, particularly the over-activation and/or upregulation of the ligand-gated ionotropic glutamatergic NMDA receptor (NMDAR), the voltage-gated L-type Ca(2+) channel (L-channel) and the transient receptor potential canonical (TRPC) channel (a non-selective cation channel that is also permeable for Ca(2+)), which may underlie the deleterious effects of Tat on intracellular Ca(2+) homeostasis and neuronal hyper-excitation that could ultimately result in excitotoxicity. This review also seeks to provide summarized information for future studies focusing on comprehensive elucidation of molecular mechanisms underlying the pathophysiological effects of Tat (as well as some other HIV-1 proteins and immunoinflammatory molecules) on neuronal function, particularly in HIV-vulnerable brain regions.
Collapse
Affiliation(s)
- Xiu-Ti Hu
- Department of Pharmacology, Rush University Medical Center, Cohn Research Building, Rm. 414, 1735 W. Harrison Street, Chicago, IL 60612, USA.
| |
Collapse
|
15
|
Martin-Blondel G, Brassat D, Bauer J, Lassmann H, Liblau RS. CCR5 blockade for neuroinflammatory diseases — beyond control of HIV. Nat Rev Neurol 2016; 12:95-105. [DOI: 10.1038/nrneurol.2015.248] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
16
|
Fitting S, Zou S, El-Hage N, Suzuki M, Paris JJ, Schier CJ, Rodríguez JW, Rodriguez M, Knapp PE, Hauser KF. Opiate addiction therapies and HIV-1 Tat: interactive effects on glial [Ca²⁺]i, oxyradical and neuroinflammatory chemokine production and correlative neurotoxicity. Curr HIV Res 2015; 12:424-34. [PMID: 25760046 PMCID: PMC4475822 DOI: 10.2174/1570162x1206150311161147] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 12/13/2014] [Accepted: 02/12/2015] [Indexed: 01/05/2023]
Abstract
Few preclinical studies have compared the relative therapeutic efficacy of medications used to treat opiate addiction in relation to neuroAIDS. Here we compare the ability of methadone and buprenorphine, and the prototypic opiate morphine, to potentiate the neurotoxic and proinflammatory ([Ca2+]i, ROS, H2O2, chemokines) effects of HIV-1 Tat in neuronal and/or mixed-glial co-cultures. Repeated observations of neurons during 48 h exposure to combinations of Tat, equimolar concentrations (500 nM) of morphine, methadone, or buprenorphine exacerbated neurotoxicity significantly above levels seen with Tat alone. Buprenorphine alone displayed marked neurotoxicity at 500 nM, prompting additional studies of its neurotoxic effects at 5 nM and 50 nM concentrations ± Tat. In combination with Tat, buprenorphine displayed paradoxical, concentration-dependent, neurotoxic and neuroprotective actions. Buprenorphine neurotoxicity coincided with marked elevations in [Ca2+]i, but not increases in glial ROS or chemokine release. Tat by itself elevated the production of CCL5/RANTES, CCL4/MIP-1β, and CCL2/MCP-1. Methadone and buprenorphine alone had no effect, but methadone interacted with Tat to further increase production of CCL5/RANTES. In combination with Tat, all drugs significantly increased glial [Ca2+]i, but ROS was only significantly increased by co-exposure with morphine. Taken together, the increases in glial [Ca2+]i, ROS, and neuroinflammatory chemokines were not especially accurate predictors of neurotoxicity. Despite similarities, opiates displayed differences in their neurotoxic and neuroinflammatory interactions with Tat. Buprenorphine, in particular, was partially neuroprotective at a low concentration, which may result from its unique pharmacological profile at multiple opioid receptors. Overall, the results reveal differences among addiction medications that may impact neuroAIDS.
Collapse
|
17
|
Arenaccio C, Manfredi F, Anticoli S, Chiozzini C, Federico M. Uncovering the role of defective HIV-1 in spreading viral infection. Future Virol 2015. [DOI: 10.2217/fvl.15.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT Defective HIV-1 genomes populate blood cells of HIV-1 infected patients, especially during HAART treatment. They can express viral proteins which, if released, may induce bystander effects favoring viral spread. Here, we review recent literature regarding the effects of extracellular HIV-1 proteins which can act as effectors of transcriptionally active, defective HIV-1, including Gag p17, Env gp120, Vpr, Tat and Nef. It has been very recently described that, different to the other HIV products, the bystander effects of Nef can be mediated by exosomes, that is, nanovesicles constitutively released by all cell types. Exosomes from Nef-expressing cells induce cell activation and HIV-1 susceptibility in resting CD4+ T lymphocytes in a TNF-α-dependent way. This mechanism likely contributes to virus persistence in HAART-treated patients.
Collapse
Affiliation(s)
| | | | - Simona Anticoli
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | | | | |
Collapse
|
18
|
Duan M, Yao H, Cai Y, Liao K, Seth P, Buch S. HIV-1 Tat disrupts CX3CL1-CX3CR1 axis in microglia via the NF-κBYY1 pathway. Curr HIV Res 2015; 12:189-200. [PMID: 24862326 DOI: 10.2174/1570162x12666140526123119] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 03/20/2014] [Accepted: 03/22/2014] [Indexed: 11/22/2022]
Abstract
Microglia are critical for the pathogenesis of HIV-associated dementia not only by acting as conduits of viral entry but also as reservoirs for productive and latent virus infection, and as producers of neurotoxins. Interaction between CX3CL1 (fractalkine) and FKN receptor (CX3CR1) is highly functional in the brain, and is known to regulate a complex network of paracrine and autocrine interactions between neurons and microglia. The aim of the present study was to determine which extent of HIV-1 Tat protein causes the alteration of CX3CR1 expression and to investigate the regulatory mechanism for CX3CR1 expression. Here we showed that exposure of primary microglia and BV2 cells to exogenous Tat protein resulted in down-regulation of CX3CR1 mRNA and protein expression, with a concomitant induction of proinflammatory responses. Next, we further showed that NF-κB activation by Tat treatment negatively regulated CX3CR1 expression. Since a YY1 binding site ~10kb upstream of CX3CR1 promoter was predicted in rats, mice and humans, the classical NF-κB-YY1 regulatory pathway was considered. Our findings indicated that Tat repressed CX3CR1 expression via NF-κB-YY1 regulatory pathway. To gain insight into the effect of Tat on CX3CL1-CX3CR1 communication, calcium mobilization, MAPK activation and microglial migration, respectively, were tested in microglial cells after successive treatment with Tat and CX3CL1. The results suggested that Tat disrupted the responses of microglia to CX3CL1. Taken together, these results demonstrate that HIV-1 Tat protein suppresses CX3CR1 expression in microglia via NF-κB-YY1 pathway and attenuates CX3CL1-induced functional response of microglia.
Collapse
Affiliation(s)
| | | | | | | | | | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center (DRC 8011), University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| |
Collapse
|
19
|
Steiner JP, Bachani M, Wolfson-Stofko B, Lee MH, Wang T, Li G, Li W, Strayer D, Haughey NJ, Nath A. Interaction of paroxetine with mitochondrial proteins mediates neuroprotection. Neurotherapeutics 2015; 12:200-16. [PMID: 25404050 PMCID: PMC4322069 DOI: 10.1007/s13311-014-0315-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
There are severe neurological complications that arise from HIV infection, ranging from peripheral sensory neuropathy to cognitive decline and dementia for which no specific treatments are available. The HIV proteins secreted from infected macrophages, gp120 and Tat, are neurotoxic. The goal of this study was to screen, identify and develop neuroprotective compounds relevant to HIV-associated neurocognitive disorders (HAND). We screened more than 2000 compounds that included FDA approved drugs for protective efficacy against oxidative stress-mediated neurodegeneration and identified selective serotonin reuptake inhibitors (SSRIs) as potential neuroprotectants. Numerous SSRIs were then extensively evaluated as protectants against neurotoxicity as measured by changes in neuronal cell death, mitochondrial potential, and axodendritic degeneration elicited by HIV Tat and gp120 and other mitochondrial toxins. While many SSRIs demonstrated neuroprotective actions, paroxetine was potently neuroprotective (100 nM potency) against these toxins in vitro and in vivo following systemic administration in a gp120 neurotoxicity model. Interestingly, the inhibition of serotonin reuptake by paroxetine was not required for neuroprotection, since depletion of the serotonin transporter had no effect on its neuroprotective properties. We determined that paroxetine interacts selectively and preferentially with brain mitochondrial proteins and blocks calcium-dependent swelling but had less effect on liver mitochondria. Additionally, paroxetine induced proliferation of neural progenitor cells in vitro and in vivo in gp120 transgenic animals. Therefore, SSRIs such as paroxetine may provide a novel adjunctive neuroprotective and neuroregenerative therapy to treat HIV-infected individuals.
Collapse
Affiliation(s)
- Joseph P. Steiner
- />Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
- />Translational Neuroscience Center, National Institute of Neurological Diseases and Stroke, National Bldg 10, Room 7C-105, 10 Center Drive, Bethesda, MD 20892 USA
| | - Muznabanu Bachani
- />Translational Neuroscience Center, National Institute of Neurological Diseases and Stroke, National Bldg 10, Room 7C-105, 10 Center Drive, Bethesda, MD 20892 USA
| | - Brett Wolfson-Stofko
- />Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
| | - Myoung-Hwa Lee
- />Section of Infections of the Nervous System, National Institute of Neurological Diseases and Stroke, National Institute of Health, Bldg 10, Room 7C103, 10 Center Drive, Bethesda, MD 20892 USA
| | - Tonguang Wang
- />Translational Neuroscience Center, National Institute of Neurological Diseases and Stroke, National Bldg 10, Room 7C-105, 10 Center Drive, Bethesda, MD 20892 USA
| | - Guanhan Li
- />Section of Infections of the Nervous System, National Institute of Neurological Diseases and Stroke, National Institute of Health, Bldg 10, Room 7C103, 10 Center Drive, Bethesda, MD 20892 USA
| | - Wenxue Li
- />Section of Infections of the Nervous System, National Institute of Neurological Diseases and Stroke, National Institute of Health, Bldg 10, Room 7C103, 10 Center Drive, Bethesda, MD 20892 USA
| | - David Strayer
- />Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Norman J. Haughey
- />Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
| | - Avindra Nath
- />Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
- />Translational Neuroscience Center, National Institute of Neurological Diseases and Stroke, National Bldg 10, Room 7C-105, 10 Center Drive, Bethesda, MD 20892 USA
- />Section of Infections of the Nervous System, National Institute of Neurological Diseases and Stroke, National Institute of Health, Bldg 10, Room 7C103, 10 Center Drive, Bethesda, MD 20892 USA
| |
Collapse
|
20
|
Luzzi A, Morettini F, Gazaneo S, Mundo L, Onnis A, Mannucci S, Rogena EA, Bellan C, Leoncini L, De Falco G. HIV-1 Tat induces DNMT over-expression through microRNA dysregulation in HIV-related non Hodgkin lymphomas. Infect Agent Cancer 2014; 9:41. [PMID: 25705251 PMCID: PMC4334912 DOI: 10.1186/1750-9378-9-41] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/14/2014] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND A close association between HIV infection and the development of cancer exists. Although the advent of highly active antiretroviral therapy has changed the epidemiology of AIDS-associated malignancies, a better understanding on how HIV can induce malignant transformation will help the development of novel therapeutic agents. METHODS HIV has been reported to induce the expression of DNMT1 in vitro, but still no information is available about the mechanisms regulating DNMT expression in HIV-related B-cell lymphomas. In this paper, we investigated the expression of DNMT family members (DNMT1, DNMT3a/b) in primary cases of aggressive B-cell lymphomas of HIV-positive subjects. RESULTS Our results confirmed the activation of DNMT1 by HIV in vivo, and reported for the first time a marked up-regulation of DNMT3a and DNMT3b in HIV-positive aggressive B-cell lymphomas. DNMT up-regulation in HIV-positive tumors correlated with down-regulation of specific microRNAs, as the miR29 family, the miR148-152 cluster, known to regulate their expression. Literature reports the activation of DNMTs by the human polyomavirus BKV large T-antigen and adenovirus E1a, through the pRb/E2F pathway. We have previously demonstrated that the HIV Tat protein is able to bind to the pocket proteins and to inactivate their oncosuppressive properties, resulting in uncontrolled cell proliferation. Therefore, we focused on the role of Tat, due to its capability to be released from infected cells and to dysregulate uninfected ones, using an in vitro model in which Tat was ectopically expressed in B-cells. CONCLUSIONS Our findings demonstrated that the ectopic expression of Tat was per se sufficient to determine DNMT up-regulation, based on microRNA down-regulation, and that this results in aberrant hypermethylation of target genes and microRNAs. These results point at a direct role for Tat in participating in uninfected B-cell lymphomagenesis, through dysregulation of the epigenetical control of gene expression.
Collapse
Affiliation(s)
- Anna Luzzi
- />Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Federica Morettini
- />Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Sara Gazaneo
- />Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Lucia Mundo
- />Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Anna Onnis
- />Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Susanna Mannucci
- />Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Emily A Rogena
- />Department of Medical Biotechnologies, University of Siena, Siena, Italy
- />Department of Pathology, University of Nairobi, Nairobi, Kenya
| | - Cristiana Bellan
- />Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Lorenzo Leoncini
- />Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Giulia De Falco
- />Department of Medical Biotechnologies, University of Siena, Siena, Italy
- />School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| |
Collapse
|
21
|
Vartak-Sharma N, Gelman BB, Joshi C, Borgamann K, Ghorpade A. Astrocyte elevated gene-1 is a novel modulator of HIV-1-associated neuroinflammation via regulation of nuclear factor-κB signaling and excitatory amino acid transporter-2 repression. J Biol Chem 2014; 289:19599-612. [PMID: 24855648 DOI: 10.1074/jbc.m114.567644] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Astrocyte elevated gene-1 (AEG-1), a novel human immunodeficiency virus (HIV)-1 and tumor necrosis factor (TNF)-α-inducible oncogene, has generated significant interest in the field of cancer research as a therapeutic target for many metastatic aggressive tumors. However, little is known about its role in astrocyte responses during HIV-1 central nervous system (CNS) infection and whether it contributes toward the development of HIV-associated neurocognitive disorders (HAND). Therefore, in this study, we investigated changes in AEG-1 CNS expression in HIV-1-infected brain tissues and elucidated a potential mechanism of AEG-1-mediated regulation of HAND. Immunoblotting and immunohistochemical analyses of HIV-1 seropositive and HIV-1 encephalitic human brain tissues revealed significantly elevated levels of AEG-1 protein. Immunohistochemical analyses of HIV-1 Tat transgenic mouse brain tissues also showed a marked increase in AEG-1 staining. Similar to in vivo observations, cultured astrocytes expressing HIV-1 Tat also revealed AEG-1 and cytokine up-regulation. Astrocytes treated with HAND-relevant stimuli, TNF-α, interleukin (IL)-1β, and HIV-1, also significantly induced AEG-1 expression and nuclear translocation via activation of the nuclear factor (NF)-κB pathway. Co-immunoprecipitation studies demonstrated IL-1β- or TNF-α-induced AEG-1 interaction with NF-κB p65 subunit. AEG-1 knockdown decreased NF-κB activation, nuclear translocation, and transcriptional output in TNF-α-treated astrocytes. Moreover, IL-1β treatment of AEG-1-overexpressing astrocytes significantly lowered expression of excitatory amino acid transporter 2, increased expression of excitatory amino acid transporter 2 repressor ying yang 1, and reduced glutamate clearance, a major transducer of excitotoxic neuronal damage. Findings from this study identify a novel transcriptional co-factor function of AEG-1 and further implicate AEG-1 in HAND-associated neuroinflammation.
Collapse
Affiliation(s)
- Neha Vartak-Sharma
- From the Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas 76107 and
| | - Benjamin B Gelman
- the Departments of Pathology and Neuroscience & Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Chaitanya Joshi
- From the Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas 76107 and
| | - Kathleen Borgamann
- From the Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas 76107 and
| | - Anuja Ghorpade
- From the Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas 76107 and
| |
Collapse
|
22
|
Lipfert J, Ödemis V, Wagner DC, Boltze J, Engele J. CXCR4 and CXCR7 form a functional receptor unit for SDF-1/CXCL12 in primary rodent microglia. Neuropathol Appl Neurobiol 2014; 39:667-80. [PMID: 23289420 DOI: 10.1111/nan.12015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 11/30/2012] [Indexed: 12/25/2022]
Abstract
AIMS Microglial cells have been originally identified as a target for the CXC chemokine, SDF-1, by their expression of CXCR4. More recently, it has been recognized that SDF-1 additionally binds to CXCR7, which depending on the cell type acts as either a nonclassical, a classical or a scavenger chemokine receptor. Here, we asked whether primary microglial cells additionally express CXCR7 and if so how this chemokine receptor functions in this cell type. METHODS CXCR4 and CXCR7 expression was analysed in cultured rat microglia and in the brain of animals with permanent occlusion of the middle cerebral artery (MCAO) by either Western blotting, RT-PCR, flow cytometry and/or immunocytochemistry. The function of CXCR4 and CXCR7 was assessed in the presence of selective antagonists. RESULTS Cultured primary rat microglia expressed CXCR4 and CXCR7 to similar levels. Treatment with SDF-1 resulted in the activation of Erk1/2 and Akt signalling. Erk1/2 and Akt signalling were required for subsequent SDF-1-dependent promotion of microglial proliferation. In contrast, Erk1/2 signalling was sufficient for SDF-1-induced migration of microglial cells. Both SDF-1-dependent signalling and the resulting effects on microglial proliferation and migration were abrogated following pharmacological inactivation of either CXCR4 or CXCR7. Moreover, treatment of cultured microglia with lipopolysaccharide resulted in the co-ordinated up-regulation of CXCR4 and CXCR7 expression. Likewise, reactive microglia accumulating in the area adjacent to the lesion core in MCAO rats expressed both CXCR4 and CXCR7. CONCLUSIONS CXCR4 and CXCR7 form a functional receptor unit in microglial cells, which is up-regulated during activation of microglia both in vitro and in vivo.
Collapse
Affiliation(s)
- J Lipfert
- Institute of Anatomy, Medical Faculty, University of Leipzig, Leipzig, Germany
| | | | | | | | | |
Collapse
|
23
|
Nookala AR, Shah A, Noel RJ, Kumar A. HIV-1 Tat-mediated induction of CCL5 in astrocytes involves NF-κB, AP-1, C/EBPα and C/EBPγ transcription factors and JAK, PI3K/Akt and p38 MAPK signaling pathways. PLoS One 2013; 8:e78855. [PMID: 24244375 PMCID: PMC3823997 DOI: 10.1371/journal.pone.0078855] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 09/23/2013] [Indexed: 12/23/2022] Open
Abstract
The incidence of HIV-associated neurological disorders (HAND) has increased during recent years even though the highly active antiretroviral therapy (HAART) has significantly curtailed the virus replication and increased the life expectancy among HIV-1 infected individuals. These neurological deficits have been attributed to HIV proteins including HIV-1 Tat. HIV-1 Tat is known to up-regulate CCL5 expression in mouse astrocytes, but the mechanism of up-regulation is not known. The present study was undertaken with the objective of determining the mechanism(s) underlying HIV-1 Tat-mediated expression of CCL5 in astrocytes. SVGA astrocytes were transiently transfected with a plasmid encoding Tat, and expression of CCL5 was studied at the mRNA and protein levels using real time RT-PCR and multiplex cytokine bead array, respectively. HIV-1 Tat showed a time-dependent increase in the CCL5 expression with peak mRNA and protein levels, observed at 1 h and 48 h post-transfection, respectively. In order to explore the mechanism(s), pharmacological inhibitors and siRNA against different pathway(s) were used. Pre-treatment with SC514 (NF-κB inhibitor), LY294002 (PI3K inhibitor), AG490 (JAK2 inhibitor) and Janex-1 (JAK3 inhibitor) showed partial reduction of the Tat-mediated induction of CCL5 suggesting involvement of JAK, PI3K/Akt and NF-κB in CCL5 expression. These results were further confirmed by knockdown of the respective genes using siRNA. Furthermore, p38 MAPK was found to be involved since the knockdown of p38δ but not other isoforms showed partial reduction in CCL5 induction. This was further confirmed at transcriptional level that AP-1, C/EBPα and C/EBPγ were involved in CCL5 up-regulation.
Collapse
Affiliation(s)
- Anantha R. Nookala
- Division of Pharmacology and Toxicology, UMKC-School of Pharmacy, Kansas City, Missouri, United States of America
| | - Ankit Shah
- Division of Pharmacology and Toxicology, UMKC-School of Pharmacy, Kansas City, Missouri, United States of America
| | - Richard J. Noel
- Department of Biochemistry, Ponce School of Medicine and Health Sciences, Ponce, Puerto Rico, United States of America
| | - Anil Kumar
- Division of Pharmacology and Toxicology, UMKC-School of Pharmacy, Kansas City, Missouri, United States of America
- * E-mail:
| |
Collapse
|
24
|
Marker DF, Tremblay MÈ, Puccini JM, Barbieri J, Gantz Marker MA, Loweth CJ, Muly EC, Lu SM, Goodfellow VS, Dewhurst S, Gelbard HA. The new small-molecule mixed-lineage kinase 3 inhibitor URMC-099 is neuroprotective and anti-inflammatory in models of human immunodeficiency virus-associated neurocognitive disorders. J Neurosci 2013; 33:9998-10010. [PMID: 23761895 PMCID: PMC3682381 DOI: 10.1523/jneurosci.0598-13.2013] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 04/09/2013] [Accepted: 05/06/2013] [Indexed: 01/11/2023] Open
Abstract
Human immunodeficiency virus (HIV)-associated neurocognitive disorders (HAND) is a significant source of disability in the HIV-infected population. Even with stringent adherence to anti-retroviral therapy, >50% of patients living with HIV-1 will develop HAND (Heaton et al., 2010). Because suppression of viral replication alone is not enough to stop HAND progression, there is a need for an adjunctive neuroprotective therapy in this population. To this end, we have developed a small-molecule brain-penetrant inhibitor with activity against mixed-lineage kinase 3 (MLK3), named URMC-099. MLK3 activation is associated with many of the pathologic hallmarks of HAND (Bodner et al., 2002, 2004; Sui et al., 2006) and therefore represents a prime target for adjunctive therapy based on small-molecule kinase inhibition. Here we demonstrate the anti-inflammatory and neuroprotective effects of URMC-099 in multiple murine and rodent models of HAND. In vitro, URMC-099 treatment reduced inflammatory cytokine production by HIV-1 Tat-exposed microglia and prevented destruction and phagocytosis of cultured neuronal axons by these cells. In vivo, URMC-099 treatment reduced inflammatory cytokine production, protected neuronal architecture, and altered the morphologic and ultrastructural response of microglia to HIV-1 Tat exposure. In conclusion, these data provide compelling in vitro and in vivo evidence to investigate the utility of URMC-099 in other models of HAND with the goal of advancement to an adjunctive therapeutic agent.
Collapse
MESH Headings
- Animals
- Bone Marrow Transplantation
- CX3C Chemokine Receptor 1
- Cell Line, Transformed/drug effects
- Cell Line, Transformed/virology
- Cells, Cultured
- Cytokines
- Disease Models, Animal
- Embryo, Mammalian
- Gene Products, tat/immunology
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- HIV Infections/complications
- HIV Infections/drug therapy
- HIV Infections/virology
- HIV-1/physiology
- Hippocampus/pathology
- Humans
- Inflammation/genetics
- Inflammation/pathology
- Inflammation/prevention & control
- Inflammation/virology
- MAP Kinase Kinase Kinases/antagonists & inhibitors
- Mice
- Mice, Transgenic
- Microscopy, Immunoelectron
- Neuroprotective Agents/therapeutic use
- Phagocytosis/drug effects
- Phagocytosis/genetics
- Phosphorylation/drug effects
- Pyridines/pharmacology
- Pyridines/therapeutic use
- Pyrroles/pharmacology
- Pyrroles/therapeutic use
- Rats
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Statistics, Nonparametric
- Time Factors
- Transfection
- tat Gene Products, Human Immunodeficiency Virus
- Mitogen-Activated Protein Kinase Kinase Kinase 11
Collapse
Affiliation(s)
- Daniel F Marker
- Centers for Neural Development and Disease, University of Rochester, Rochester, New York 14642, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
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
|
26
|
Gaskill PJ, Calderon TM, Coley JS, Berman JW. Drug induced increases in CNS dopamine alter monocyte, macrophage and T cell functions: implications for HAND. J Neuroimmune Pharmacol 2013; 8:621-42. [PMID: 23456305 PMCID: PMC4303241 DOI: 10.1007/s11481-013-9443-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 02/13/2013] [Indexed: 02/08/2023]
Abstract
Central nervous system (CNS) complications resulting from HIV infection remain a major public health problem as individuals live longer due to the success of combined antiretroviral therapy (cART). As many as 70 % of HIV infected people have HIV associated neurocognitive disorders (HAND). Many HIV infected individuals abuse drugs, such as cocaine, heroin or methamphetamine, that may be important cofactors in the development of HIV CNS disease. Despite different mechanisms of action, all drugs of abuse increase extracellular dopamine in the CNS. The effects of dopamine on HIV neuropathogenesis are not well understood, and drug induced increases in CNS dopamine may be a common mechanism by which different types of drugs of abuse impact the development of HAND. Monocytes and macrophages are central to HIV infection of the CNS and to HAND. While T cells have not been shown to be a major factor in HIV-associated neuropathogenesis, studies indicate that T cells may play a larger role in the development of HAND in HIV infected drug abusers. Drug induced increases in CNS dopamine may dysregulate functions of, or increase HIV infection in, monocytes, macrophages and T cells in the brain. Thus, characterizing the effects of dopamine on these cells is important for understanding the mechanisms that mediate the development of HAND in drug abusers.
Collapse
Affiliation(s)
- Peter J Gaskill
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | | | |
Collapse
|
27
|
Peng F, Yao H, Akturk HK, Buch S. Platelet-derived growth factor CC-mediated neuroprotection against HIV Tat involves TRPC-mediated inactivation of GSK 3beta. PLoS One 2012; 7:e47572. [PMID: 23077641 PMCID: PMC3471979 DOI: 10.1371/journal.pone.0047572] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 09/17/2012] [Indexed: 12/14/2022] Open
Abstract
Platelet-derived growth factor-CC (PDGF-CC) is the third member of the PDGF family, and has been implicated both in embryogenesis and development of the CNS. The biological function of this isoform however, remains largely unexplored in the context of HIV-associated dementia (HAD). In the present study, we demonstrate that exposure of human neuroblastoma cells SH-SY5Y to HIV transactivator protein Tat resulted in decreased intrinsic expression of PDGF-CC as evidenced by RT-PCR and western blot assays. Reciprocally, pretreatment of SH-SY5Y cells with PDGF-CC abrogated Tat-mediated neurotoxicity by mitigating apoptosis and neurite & MAP-2 loss. Using pharmacological and loss of function approaches we identified the role of phosphatidylinositol 3-kinase (PI3K)/Akt signaling in PDGF-CC-mediated neuroprotection. We report herein a novel role about the involvement of transient receptor potential canonical (TRPC) channel 1 in modulation of calcium transients in PDGF-CC-mediated neuroprotection. Furthermore we also demonstrated PDGF-CC-mediated inactivation of the downstream mediator - glycogen synthase kinase 3β (GSK3β) evidenced by its phosphorylation at Ser-9. This was further validated by gain and loss of function studies using cells transfected with either the wild type or mutant GSK3β constructs. Intriguingly, pretreatment of cells with either the PI3K inhibitor or TRPC blocker resulted in failure of PDGF-CC to inactivate GSK3β, thereby suggesting the intersection of PI3K and TRPC signaling at GSK3β. Taken together our findings lead to the suggestion that PDGF-CC could be developed as a therapeutic target to reverse Tat-mediated neurotoxicity with implications for HAD.
Collapse
Affiliation(s)
- Fuwang Peng
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Honghong Yao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Halis Kaan Akturk
- Department of Medicine, Creighton Medical Center, Omaha, Nebraska, United States of America
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
28
|
Zhu XB, Wang YB, Chen O, Zhang DQ, Zhang ZH, Cao AH, Huang SY, Sun RP. Characterization of the expression of macrophage inflammatory protein-1α (MIP-1α) and C-C chemokine receptor 5 (CCR5) after kainic acid-induced status epilepticus (SE) in juvenile rats. Neuropathol Appl Neurobiol 2012; 38:602-16. [DOI: 10.1111/j.1365-2990.2012.01251.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
29
|
Interactive Effects of Morphine on HIV Infection: Role in HIV-Associated Neurocognitive Disorder. AIDS Res Treat 2012; 2012:953678. [PMID: 22666564 PMCID: PMC3362817 DOI: 10.1155/2012/953678] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 02/13/2012] [Accepted: 03/02/2012] [Indexed: 01/06/2023] Open
Abstract
HIV epidemic continues to be a severe public health problem and concern within USA and across the globe with about 33 million people infected with HIV. The frequency of drug abuse among HIV infected patients is rapidly increasing and is another major issue since injection drug users are at a greater risk of developing HIV associated neurocognitive dysfunctions compared to non-drug users infected with HIV. Brain is a major target for many of the recreational drugs and HIV. Evidences suggest that opiate drug abuse is a risk factor in HIV infection, neural dysfunction and progression to AIDS. The information available on the role of morphine as a cofactor in the neuropathogenesis of HIV is scanty. This review summarizes the results that help in understanding the role of morphine use in HIV infection and neural dysfunction. Studies show that morphine enhances HIV-1 infection by suppressing IL-8, downregulating chemokines with reciprocal upregulation of HIV coreceptors. Morphine also activates MAPK signaling and downregulates cAMP response element-binding protein (CREB). Better understanding on the role of morphine in HIV infection and mechanisms through which morphine mediates its effects may help in devising novel therapeutic strategies against HIV-1 infection in opiate using HIV-infected population.
Collapse
|
30
|
Fiume G, Vecchio E, De Laurentiis A, Trimboli F, Palmieri C, Pisano A, Falcone C, Pontoriero M, Rossi A, Scialdone A, Fasanella Masci F, Scala G, Quinto I. Human immunodeficiency virus-1 Tat activates NF-κB via physical interaction with IκB-α and p65. Nucleic Acids Res 2011; 40:3548-62. [PMID: 22187158 PMCID: PMC3333881 DOI: 10.1093/nar/gkr1224] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nuclear factor (NF)-κB is a master regulator of pro-inflammatory genes and is upregulated in human immunodeficiency virus 1 (HIV-1) infection. Mechanisms underlying the NF-κB deregulation by HIV-1 are relevant for immune dysfunction in AIDS. We report that in single round HIV-1 infection, or single-pulse PMA stimulation, the HIV-1 Tat transactivator activated NF-κB by hijacking the inhibitor IκB-α and by preventing the repressor binding to the NF-κB complex. Moreover, Tat associated with the p65 subunit of NF-κB and increased the p65 DNA-binding affinity and transcriptional activity. The arginine- and cysteine-rich domains of Tat were required for IκB-α and p65 association, respectively, and for sustaining the NF-κB activity. Among an array of NF-κB-responsive genes, Tat mostly activated the MIP-1α expression in a p65-dependent manner, and bound to the MIP-1α NF-κB enhancer thus promoting the recruitment of p65 with displacement of IκB-α; similar findings were obtained for the NF-κB-responsive genes CSF3, LTA, NFKBIA and TLR2. Our results support a novel mechanism of NF-κB activation via physical interaction of Tat with IκB-α and p65, and may contribute to further insights into the deregulation of the inflammatory response by HIV-1.
Collapse
Affiliation(s)
- Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University of Catanzaro Magna Graecia, Viale Europa-Germaneto, 88100 Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
In the past 25 years revelations on the genesis of human cancer have come at an increasing pace. Research on oncogenic infectious agents, especially viruses, has helped us to understand the process of malignant transformation of cells because the cellular events in viral-driven transformation mirror, often brilliantly, basic cellular processes that culminate in cancer, even those not associated with viruses. Infectious agents, especially viruses, account for several of the most common malignancies-up to 20% of all cancers. Some of these cancers are endemic, with a high incidence in certain geographic locations, but sporadic/lower incidence in other parts of the world. Lymphomas arise frequently in association with infectious agents such as Epstein-Barr virus, human immunodeficiency virus, human herpes virus 8, Helicobacter pylori, and hepatitis C virus. In this review, we will focus on the association between infectious agents and lymphomas, with a look at the molecular mechanisms they use to disturb cell regulation and eventually result in cancer.
Collapse
Affiliation(s)
- Giulia De Falco
- Department of Human Pathology and Oncology, University of Siena, Siena, Italy
| | | | | |
Collapse
|
32
|
Borjabad A, Morgello S, Chao W, Kim SY, Brooks AI, Murray J, Potash MJ, Volsky DJ. Significant effects of antiretroviral therapy on global gene expression in brain tissues of patients with HIV-1-associated neurocognitive disorders. PLoS Pathog 2011; 7:e1002213. [PMID: 21909266 PMCID: PMC3164642 DOI: 10.1371/journal.ppat.1002213] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 07/01/2011] [Indexed: 11/18/2022] Open
Abstract
Antiretroviral therapy (ART) has reduced morbidity and mortality in HIV-1 infection; however HIV-1-associated neurocognitive disorders (HAND) persist despite treatment. The reasons for the limited efficacy of ART in the brain are unknown. Here we used functional genomics to determine ART effectiveness in the brain and to identify molecular signatures of HAND under ART. We performed genome-wide microarray analysis using Affymetrix U133 Plus 2.0 Arrays, real-time PCR, and immunohistochemistry in brain tissues from seven treated and eight untreated HAND patients and six uninfected controls. We also determined brain virus burdens by real-time PCR. Treated and untreated HAND brains had distinct gene expression profiles with ART transcriptomes clustering with HIV-1-negative controls. The molecular disease profile of untreated HAND showed dysregulated expression of 1470 genes at p<0.05, with activation of antiviral and immune responses and suppression of synaptic transmission and neurogenesis. The overall brain transcriptome changes in these patients were independent of histological manifestation of HIV-1 encephalitis and brain virus burdens. Depending on treatment compliance, brain transcriptomes from patients on ART had 83% to 93% fewer dysregulated genes and significantly lower dysregulation of biological pathways compared to untreated patients, with particular improvement indicated for nervous system functions. However a core of about 100 genes remained similarly dysregulated in both treated and untreated patient brain tissues. These genes participate in adaptive immune responses, and in interferon, cell cycle, and myelin pathways. Fluctuations of cellular gene expression in the brain correlated in Pearson's formula analysis with plasma but not brain virus burden. Our results define for the first time an aberrant genome-wide brain transcriptome of untreated HAND and they suggest that antiretroviral treatment can be broadly effective in reducing pathophysiological changes in the brain associated with HAND. Aberrantly expressed transcripts common to untreated and treated HAND may contribute to neurocognitive changes defying ART.
Collapse
Affiliation(s)
- Alejandra Borjabad
- Molecular Virology Division, St. Luke's-Roosevelt Hospital Center and Columbia University, New York, New York, United States of America
| | - Susan Morgello
- Department of Pathology and Neuroscience, The Mount Sinai Medical Center, New York, New York, United States of America
| | - Wei Chao
- Molecular Virology Division, St. Luke's-Roosevelt Hospital Center and Columbia University, New York, New York, United States of America
| | - Seon-Young Kim
- Human Genomics Laboratory, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Andrew I. Brooks
- Department of Genetics, Environmental and Occupational Health Science Institute (EOHSI), Rutgers University, Piscataway, New Jersey, United States of America
| | - Jacinta Murray
- Department of Pathology and Neuroscience, The Mount Sinai Medical Center, New York, New York, United States of America
| | - Mary Jane Potash
- Molecular Virology Division, St. Luke's-Roosevelt Hospital Center and Columbia University, New York, New York, United States of America
| | - David J. Volsky
- Molecular Virology Division, St. Luke's-Roosevelt Hospital Center and Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
33
|
Renner NA, Ivey NS, Redmann RK, Lackner AA, MacLean AG. MCP-3/CCL7 production by astrocytes: implications for SIV neuroinvasion and AIDS encephalitis. J Neurovirol 2011; 17:146-52. [PMID: 21279498 PMCID: PMC3086688 DOI: 10.1007/s13365-010-0017-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 12/03/2010] [Accepted: 12/10/2010] [Indexed: 10/18/2022]
Abstract
Monocyte/macrophages and activated lymphocytes traffic through normal brain, and this trafficking is increased in inflammatory conditions such as HIV encephalitis (HIVE). HIVE is characterized in part by perivascular accumulations of macrophages. The earliest events in this process are poorly understood and difficult or impossible to address in humans. The SIV-infected macaque model of neuroAIDS has demonstrated migration of monocytes into the brain early in disease, coincident with peak SIV viremia. The chemotactic signals that initiate the increased emigration of mononuclear cells into the CNS have not been described. Here, we describe astrocytes as a primary source of chemokines to facilitate basal levels of monocyte trafficking to CNS and that increased chemokine (C-C motif) ligand 7 (CCL7) production may be responsible for initiating the increased trafficking in neuroAIDS. We have previously published complementary in vivo work demonstrating the presence of monocyte chemoattractant protein 3 (MCP-3)/CCL7 within the brain of SIV-infected macaques. Here, we demonstrate that MCP-3/CCL7 is a significant chemokine produced by astrocytes, that basal monocyte migration may be facilitated by astrocyte-derived CCL7, that production of CCL7 is rapidly increased by TNF-α and thus likely plays a critical role in initiating neuroinvasion by SIV/HIV.
Collapse
Affiliation(s)
- Nicole A. Renner
- Tulane National Primate Research Center, Covington, LA 70433
- Graduate Program in Neuroscience, Tulane University, New Orleans, LA 70118
| | - Nathan S. Ivey
- Tulane National Primate Research Center, Covington, LA 70433
- Graduate Program in Neuroscience, Tulane University, New Orleans, LA 70118
| | | | - Andrew A. Lackner
- Tulane National Primate Research Center, Covington, LA 70433
- Graduate Program in Neuroscience, Tulane University, New Orleans, LA 70118
| | - Andrew G. MacLean
- Tulane National Primate Research Center, Covington, LA 70433
- Graduate Program in Neuroscience, Tulane University, New Orleans, LA 70118
| |
Collapse
|
34
|
Gelbard HA, Dewhurst S, Maggirwar SB, Kiebala M, Polesskaya O, Gendelman HE. Rebuilding synaptic architecture in HIV-1 associated neurocognitive disease: a therapeutic strategy based on modulation of mixed lineage kinase. Neurotherapeutics 2010; 7:392-8. [PMID: 20880503 PMCID: PMC2948545 DOI: 10.1016/j.nurt.2010.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Revised: 07/28/2010] [Accepted: 08/04/2010] [Indexed: 12/13/2022] Open
Abstract
Work from our laboratories has validated mixed lineage kinase type 3 (MLK3) as an enzyme pathologically activated in the CNS by human immunodeficiency virus 1 (HIV-1) neurotoxins. In this review, we discuss MLK3 activation in the context of the neuropathogenesis of HIV-1 associated neurocognitive deficits (HAND). We use findings from the literature to substantiate the neuropathologic relevance of MLK3 to neurodegenerative disease, with an emphasis on Parkinson's disease that shares a number of important phenotypic and neuropathologic characteristics with HAND. We discuss signal transduction pathways downstream from MLK3 activation, with an emphasis on their involvement in microglia and neurons in preclinical models of HAND. Finally, we make a case for pharmacologic intervention targeted at inhibition of MLK3 as a strategy to reverse HAND, in light of the fact that combination antiretroviral therapy, despite successfully managing systemic infection of HIV-1, has been largely unsuccessful in eradicating HAND.
Collapse
Affiliation(s)
- Harris A Gelbard
- Center for Neural Development and Disease, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Hahn YK, Vo P, Fitting S, Block ML, Hauser KF, Knapp PE. beta-Chemokine production by neural and glial progenitor cells is enhanced by HIV-1 Tat: effects on microglial migration. J Neurochem 2010; 114:97-109. [PMID: 20403075 PMCID: PMC2992981 DOI: 10.1111/j.1471-4159.2010.06744.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Human immunodeficiency virus (HIV)-1 neuropathology results from collective effects of viral proteins and inflammatory mediators on several cell types. Significant damage is mediated indirectly through inflammatory conditions promulgated by glial cells, including microglia that are productively infected by HIV-1, and astroglia. Neural and glial progenitors exist in both developing and adult brains. To determine whether progenitors are targets of HIV-1, a multi-plex assay was performed to assess chemokine/cytokine expression after treatment with viral proteins transactivator of transcription (Tat) or glycoprotein 120 (gp120). In the initial screen, ten analytes were basally released by murine striatal progenitors. The beta-chemokines CCL5/regulated upon activation, normal T cell expressed and secreted, CCL3/macrophage inflammatory protein-1alpha, and CCL4/macrophage inflammatory protein-1beta were increased by 12-h exposure to HIV-1 Tat. Secreted factors from Tat-treated progenitors were chemoattractive towards microglia, an effect blocked by 2D7 anti-CCR5 antibody pre-treatment. Tat and opiates have interactive effects on astroglial chemokine secretion, but this interaction did not occur in progenitors. gp120 did not affect chemokine/cytokine release, although both CCR5 and CXCR4, which serve as gp120 co-receptors, were detected in progenitors. We postulate that chemokine production by progenitors may be a normal, adaptive process that encourages immune inspection of newly generated cells. Pathogens such as HIV might usurp this function to create a maladaptive state, especially during development or regeneration, when progenitors are numerous.
Collapse
Affiliation(s)
- Yun Kyung Hahn
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Phu Vo
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Sylvia Fitting
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Michelle L. Block
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Kurt F. Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298 USA
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Pamela E. Knapp
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298 USA
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298 USA
| |
Collapse
|
36
|
Hazleton JE, Berman JW, Eugenin EA. Novel mechanisms of central nervous system damage in HIV infection. HIV AIDS-RESEARCH AND PALLIATIVE CARE 2010; 2:39-49. [PMID: 22096383 PMCID: PMC3218694 DOI: 10.2147/hiv.s9186] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Human immunodeficiency virus-1 infection of the central nervous system is an early event after primary infection, resulting in motor and cognitive defects in a significant number of individuals despite successful antiretroviral therapy. The pathology of the infected brain is characterized by enhanced leukocyte infiltration, microglial activation and nodules, aberrant expression of inflammatory factors, neuronal dysregulation and loss, and blood–brain barrier disruption. Months to years following the primary infection, these central nervous system insults result in a spectrum of motor and cognitive dysfunction, ranging from mild impairment to frank dementia. The mechanisms that mediate impairment are still not fully defined. In this review we discuss the cellular and molecular mechanisms that facilitate impairment and new data that implicate intercellular communication systems, gap junctions and tunneling nanotubes, as mediators of human immunodeficiency virus-1 toxicity and infection within the central nervous system. These data suggest potential targets for novel therapeutics.
Collapse
Affiliation(s)
- Joy E Hazleton
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | |
Collapse
|
37
|
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: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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
|
38
|
Ragin AB, Wu Y, Ochs R, Scheidegger R, Cohen BA, Edelman RR, Epstein LG, McArthur J. Biomarkers of neurological status in HIV infection: a 3-year study. Proteomics Clin Appl 2010; 4:295-303. [PMID: 21137050 PMCID: PMC3243487 DOI: 10.1002/prca.200900083] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 07/24/2009] [Accepted: 10/08/2009] [Indexed: 01/12/2023]
Abstract
PURPOSE To evaluate circulating cytokines and chemokines as correlates of the degree of brain injury in individuals with advanced human immunodeficiency virus (HIV) infection. EXPERIMENTAL DESIGN Study participants included ten well-characterized subjects in advanced stage HIV infection. High-throughput multiplexed analysis was used to quantify markers of interest at baseline and 3 years later in the clinical course. Objective measurements of the brain were derived in vivo with quantitative magnetic resonance segmentation algorithms and with diffusion tensor imaging. RESULTS Of the markers examined, monocyte chemoattractant protein-1 (MCP-1 or CCL-2) was the most prominent correlate of brain injury. Elevated MCP-1 levels correlated with brain white matter alterations at the initial assessment. The relationship to injury was more extensive 3 years later; elevated MCP-1 was significantly correlated with measures of brain microstructural alterations and of abject atrophy. CONCLUSIONS AND CLINICAL RELEVANCE The findings build on our prior observations that elevated MCP-1 levels may be a useful predictive marker for HIV-associated neurocognitive disorder. As a potent chemoattractant, MCP-1 may mediate injury through participation in self-reinforcing cycles of chronic immune activation and cytokine/chemokine-mediated neurotoxicity.
Collapse
Affiliation(s)
- Ann B Ragin
- Department of Radiology, Northwestern University, Chicago, IL 60611, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Eugenin EA, King JE, Hazleton JE, Major EO, Bennett MVL, Zukin RS, Berman JW. Differences in NMDA receptor expression during human development determine the response of neurons to HIV-tat-mediated neurotoxicity. Neurotox Res 2010; 19:138-48. [PMID: 20094923 DOI: 10.1007/s12640-010-9150-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Revised: 12/30/2009] [Accepted: 01/06/2010] [Indexed: 12/12/2022]
Abstract
HIV infection of the CNS can result in neurologic dysfunction in a significant number of infected individuals. NeuroAIDS is characterized by neuronal injury and loss, yet there is no evidence of HIV infection in neurons. Thus, neuronal damage and dropout are likely due to indirect effects of HIV infection of other CNS cells, through elaboration of inflammatory factors and neurotoxic viral proteins, including the viral transactivating protein tat. We and others demonstrated that tat induces apoptosis in differentiated mature human neurons. We now demonstrate that the high level of tat toxicity observed in human neurons involves specific developmental stages that correlate with N-methyl-D-aspartate receptor (NMDAR) expression, and that tat toxicity is also dependent upon the species being analyzed. Our results indicate that tat treatment of primary cultures of differentiated human neurons with significant amounts of NMDAR expression induces extensive apoptosis. In contrast, tat treatment induces only low levels of apoptosis in primary cultures of immature human neurons with low or minimal expression of NMDAR. In addition, tat treatment has minimal effect on rat hippocampal neurons in culture, despite their high expression of NMDAR. We propose that this difference may be due to low expression of the NR2A subunit. These findings are important for an understanding of the many differences among tissue culture systems and species used to study HIV-tat-mediated toxicity.
Collapse
Affiliation(s)
- E A Eugenin
- Department of Pathology, Albert Einstein College of Medicine, Forchheimer 727, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Meucci O. HIV Coreceptors and Their Roles in Leukocyte Trafficking During Neuroinflammatory Diseases. CHEMOKINE RECEPTORS AND NEUROAIDS 2010. [PMCID: PMC7120588 DOI: 10.1007/978-1-4419-0793-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Due to the increasing resistance of HIV-1 to antiretroviral therapies, there has been much emphasis on the discovery and development of alternative therapeutics for HIV-1-infected individuals. The chemokine receptors CXCR4 (Bleul et al. 1996a; Feng et al. 1996; Nagasawa et al. 1996; Oberlin et al. 1996) and CCR5 (Alkhatib et al. 1996; Deng et al. 1996; Dragic et al. 1996) were identified as target molecules from the time their role as coreceptors for HIV-1 entry into leukocytes was first discovered 10 years ago. Initial studies focused on the use of the chemokine ligands, or altered derivatives, of CXCR4 and CCR5 to prevent the entrance of HIV-1 into immune cells (Schols 2006). While these studies showed some initial promise, there was evidence of significant caveats to their use, including selection of alternative coreceptor utilizing strains (Marechal et al. 1999; Mosier et al. 1999) and the potential to cause inflammatory side effects. These data prompted the development and study of small molecule inhibitors of CXCR4 and CCR5, which have also been used to examine the roles of these molecules in a variety of inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Olimpia Meucci
- Dept. Pharmacology & Physiology, & Institute of Molecular Medicine, Drexel University College of Medicine, North 15th St. 245, Philadelphia, 19102-1101 USA
| |
Collapse
|
41
|
Coordinated regulation of SIV replication and immune responses in the CNS. PLoS One 2009; 4:e8129. [PMID: 20019816 PMCID: PMC2790080 DOI: 10.1371/journal.pone.0008129] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Accepted: 09/11/2009] [Indexed: 11/19/2022] Open
Abstract
Central nervous system (CNS) invasion during acute-stage HIV-infection has been demonstrated in a small number of individuals, but there is no evidence of neurological impairment at this stage and virus infection in brain appears to be controlled until late-stage disease. Using our reproducible SIV macaque model to examine the earliest stages of infection in the CNS, we identified immune responses that differentially regulate inflammation and virus replication in the brain compared to the peripheral blood and lymphoid tissues. SIV replication in brain macrophages and in brain of SIV-infected macaques was detected at 4 days post-inoculation (p.i.). This was accompanied by upregulation of innate immune responses, including IFNβ, IFNβ-induced gene MxA mRNA, and TNFα. Additionally, IL-10, the chemokine CCL2, and activation markers in macrophages, endothelial cells, and astrocytes were all increased in the brain at four days p.i. We observed synchronous control of virus replication, cytokine mRNA levels and inflammatory markers (MHC Class II, CD68 and GFAP) by 14 days p.i.; however, control failure was followed by development of CNS lesions in the brain. SIV infection was accompanied by induction of the dominant-negative isoform of C/EBPβ, which regulates SIV, CCL2, and IL6 transcription, as well as inflammatory responses in macrophages and astrocytes. This synchronous response in the CNS is in part due to the effect of the C/EBPβ on virus replication and cytokine expression in macrophage-lineage cells in contrast to CD4+ lymphocytes in peripheral blood and lymphoid tissues. Thus, we have identified a crucial period in the brain when virus replication and inflammation are controlled. As in HIV-infected individuals, though, this control is not sustained in the brain. Our results suggest that intervention with antiretroviral drugs or anti-inflammatory therapeutics with CNS penetration would sustain early control. These studies further suggest that interventions should target HIV-infected individuals with increased CCL2 levels or HIV RNA in the CNS.
Collapse
|
42
|
Xu JH, Long L, Tang YC, Zhang JT, Hut HT, Tang FR. CCR3, CCR2A and macrophage inflammatory protein (MIP)-1a, monocyte chemotactic protein-1 (MCP-1) in the mouse hippocampus during and after pilocarpine-induced status epilepticus (PISE) . Neuropathol Appl Neurobiol 2009; 35:496-514. [PMID: 19490431 DOI: 10.1111/j.1365-2990.2009.01022.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AIMS To investigate protein and gene expressions of chemokine subtypes CCR3, CCR2A and their respective ligands macrophage inflammatory protein 1-alpha (MIP-1alpha), monocyte chemotactic protein-1 (MCP-1) in the normal mouse central nervous system (CNS) and in the hippocampus at different time points during and after pilocarpine-induced status epilepticus (PISE). METHODS CCR3 and MIP-1alpha protein expressions were mapped in the mouse CNS. The protein and gene expressions of CCR3 and CCR2A and their respective ligands MIP-1alpha, MCP-1 in the hippocampus were studies by immunocytochemical and quantitative real-time RT-PCR during and after PISE. RESULTS CCR3 and MIP-1alpha gene expression and immunopositive neurones were broadly distributed in the CNS. CCR3 and CCA2A gene and their protein expression were downregulated in the hippocampus at 1 h during PISE. The protein expression of MIP-1alpha, MCP-1 decreased but gene expression increased at 2 h during PISE. In the hilus of the dentate gyrus, significant reduction of the numbers of CCR3, CCR2A, MCP-1 immunopositive neurones occurred from 1 h during to 2 months after PISE, but the number of MIP-1alpha neurones reduced from 2 h during to 2 months after PISE. Induced expression of CCR3 at 1 week, CCR2A, MCP-1 or MIP-1alpha at 1 week and 2 months after PISE was found in reactive astrocytes. MCP-1 was also demonstrated in the blood vessels of the hippocampus at 2 months after PISE. CONCLUSIONS CCR3 and MIP-1alpha may play important functional roles in the mouse brain. The downregulation of CCR3, CCR2A, MIP-1alpha and MCP-1 in the hippocampal neurones at the acute stage during and after PISE may weaken the neuroprotective mechanisms. However, induced expression of MCP-1 in hippocampal blood vessel may be related to changes in permeability of the blood-brain barrier during epileptogenesis.
Collapse
Affiliation(s)
- J H Xu
- Epilepsy Research Lab, National Neuroscience Institute, Singapore
| | | | | | | | | | | |
Collapse
|
43
|
Zhu X, Yao H, Peng F, Callen S, Buch S. PDGF-mediated protection of SH-SY5Y cells against Tat toxin involves regulation of extracellular glutamate and intracellular calcium. Toxicol Appl Pharmacol 2009; 240:286-91. [PMID: 19576918 PMCID: PMC2753679 DOI: 10.1016/j.taap.2009.06.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 06/22/2009] [Accepted: 06/24/2009] [Indexed: 11/20/2022]
Abstract
The human immunodeficiency virus (HIV-1) protein Tat has been implicated in mediating neuronal apoptosis, one of the hallmark features of HIV-associated dementia (HAD). Mitigation of the toxic effects of Tat could thus be a potential mechanism for reducing HIV toxicity in the brain. In this study we demonstrated that Tat-induced neurotoxicity was abolished by NMDA antagonist-MK801, suggesting the role of glutamate in this process. Furthermore, we also found that pretreatment of SH-SY5Y cells with PDGF exerted protection against Tat toxicity by decreasing extracellular glutamate levels. We also demonstrated that extracellular calcium chelator EGTA was able to abolish PDGF-mediated neuroprotection, thereby underscoring the role of calcium signaling in PDGF-mediated neuroprotection. We also showed that Erk signaling pathway was critical for PDGF-mediated protection of cells. Additionally, blocking calcium entry with EGTA resulted in suppression of PDGF-induced Erk activation. These findings thus underscore the role of PDGF-mediated calcium signaling and Erk phosphorylation in the protection of cells against HIV Tat toxicity.
Collapse
Affiliation(s)
- Xuhui Zhu
- Departments of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160
- Department of Laboratory Medicine, Tongji Hospital and Tongji Medical College of Huazhong University of Science & Technology, Wuhan,China
| | - Honghong Yao
- Departments of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Fuwang Peng
- Departments of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Shannon Callen
- Departments of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Shilpa Buch
- Departments of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160
| |
Collapse
|
44
|
Role of Tat protein in HIV neuropathogenesis. Neurotox Res 2009; 16:205-20. [PMID: 19526283 DOI: 10.1007/s12640-009-9047-8] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Revised: 03/09/2009] [Accepted: 03/09/2009] [Indexed: 12/13/2022]
Abstract
The Tat protein of the human immunodeficiency virus (HIV) has been implicated in the pathophysiology of the neurocognitive deficits associated with HIV infection. This is the earliest protein to be produced by the proviral DNA in the infected cell. The protein not only drives the regulatory regions of the virus but may also be actively released from the cell and then interact with the cell surface receptors of other uninfected cells in the brain leading to cellular dysfunction. It may also be taken up by these cells and can then activate a number of host genes. The Tat protein is highly potent and has the unique ability to travel along neuronal pathways. Importantly, its production is not impacted by the use of antiretroviral drugs once the proviral DNA has been formed. This article reviews the pleomorphic actions of Tat protein and the evidence supporting its central role in the neuropathogenesis of the HIV infection.
Collapse
|
45
|
Gaskill PJ, Calderon TM, Luers AJ, Eugenin EA, Javitch JA, Berman JW. Human immunodeficiency virus (HIV) infection of human macrophages is increased by dopamine: a bridge between HIV-associated neurologic disorders and drug abuse. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:1148-59. [PMID: 19661443 DOI: 10.2353/ajpath.2009.081067] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The prevalence of human immunodeficiency virus (HIV)-associated neurocognitive disorders (HAND) that result from HIV infection of the central nervous system is increasing. Macrophages, the primary target for HIV within the central nervous system, play a central role in HIV-induced neuropathogenesis. Drug abuse exacerbates HAND, but the mechanism(s) by which this increased neuropathology results in more severe forms of HAND in HIV-infected drug abusers is unclear. The addictive and reinforcing effects of many drugs of abuse, such as cocaine and methamphetamine, are mediated by increased extracellular dopamine in the brain. We propose a novel mechanism by which drugs of abuse intensify HIV neuropathogenesis through direct effects of the neurotransmitter dopamine on HIV infection of macrophages. We found that macrophages express dopamine receptors 1 and 2, and dopamine activates macrophages by increasing ERK 1 phosphorylation. Our results demonstrate for the first time that dopamine increases HIV replication in human macrophages and that the mechanism by which dopamine mediates this change is by increasing the total number of HIV-infected macrophages. This increase in HIV replication is mediated by activation of dopamine receptor 2. These findings suggest a common mechanism by which drugs of abuse enhance HIV replication in macrophages and indicate that the drug abuse-heightened levels of central nervous system dopamine could increase viral replication, thereby accelerating the development of HAND.
Collapse
Affiliation(s)
- Peter J Gaskill
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
The effects of the recombinant chemokine human RANTES (hRANTES) on the release of glutamate from human neocortex glutamatergic nerve endings were investigated. hRANTES facilitated the spontaneous release of d [(3)H]D-aspartate ([(3)H]DASP-) by binding Pertussis toxin-sensitive G-protein-coupled receptors (GPCRs), whose activation caused Ca(2+) mobilization from inositol trisphosphate-sensitive stores and cytosolic tyrosine kinase-mediated phosphorylations. Facilitation of release switched to inhibition when the effects of hRANTES on the 12 mM K(+)-evoked [(3)H]D-ASP exocytosis were studied. Inhibition of exocytosis relied on activation of Pertussis toxin-sensitive GPCRs negatively coupled to adenylyl cyclase. Both hRANTES effects were prevented by met-RANTES, an antagonist at the chemokine receptors (CCRs) of the CCR1, CCR3, and CCR5 subtypes. Interestingly, human neocortex glutamatergic nerve endings seem to possess all three receptor subtypes. Blockade of CCR1 and CCR5 by antibodies against the extracellular domain of CCRs prevented both the hRANTES effect on [(3)H]D-ASP release, whereas blockade of CCR3 prevented inhibition, but not facilitation, of release. The effects of RANTES on the spontaneous and the evoked release of [(3)H]D-ASP were also observed in experiments with mouse cortical synaptosomes, which may therefore represent an appropriate animal model to study RANTES-induced effects on neurotransmission. It is concluded that glutamate transmission can be modulated in opposite directions by RANTES acting at distinct CCR receptor subtypes coupled to different transduction pathways, consistent with the multiple and sometimes contrasting effects of the chemokine.
Collapse
|
47
|
El-Hage N, Bruce-Keller AJ, Knapp PE, Hauser KF. CCL5/RANTES gene deletion attenuates opioid-induced increases in glial CCL2/MCP-1 immunoreactivity and activation in HIV-1 Tat-exposed mice. J Neuroimmune Pharmacol 2008; 3:275-85. [PMID: 18815890 PMCID: PMC2722754 DOI: 10.1007/s11481-008-9127-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2008] [Accepted: 09/02/2008] [Indexed: 12/18/2022]
Abstract
To assess the role of CC-chemokine ligand 5 (CCL5)/RANTES in opiate drug abuse and human immunodeficiency virus type 1 (HIV-1) comorbidity, the effects of systemic morphine and intrastriatal HIV-1 Tat on macrophage/microglial and astroglial activation were assessed in wild-type and CCL5 knockout mice. Mice were injected intrastriatally with vehicle or Tat and assessed after 7 days. Morphine was administered to some Tat-injected mice via time-release implant (5 mg/day, s.c. for 5 days) starting at 2 days post injection. Glial activation was significantly reduced in CCL5(-/-) compared to wild-type mice at 7 days following combined Tat and morphine exposure. Moreover, the percentage of 3-nitrotyrosine immunopositive macrophages/microglia was markedly reduced in CCL5(-/-) mice injected with Tat +/- morphine compared to wild-type counterparts, suggesting that CCL5 contributes to nitrosative stress in HIV-1 encephalitis. In CCL5(-/-) mice, the reductions in Tat +/- morphine-induced gliosis coincided with significant declines in the proportion of CCL2/MCP-1-immunoreactive astrocytes and macrophages/microglia compared to wild-type counterparts. In knockout mice, neither Tat alone nor in combination with morphine increased the proportion of CCL2-immunoreactive astrocytes above percentages seen in vehicle-injected controls. Macrophages/microglia differed showing modest, albeit significant, increases in the proportion of CCL2-positive cells with combined Tat and morphine exposure, suggesting that CCL5 preferentially affects CCL2 expression by astroglia. Thus, CCL5 mediates glial activation caused by Tat and morphine, thereby aggravating HIV-1 neuropathogenesis in opiate abusers and non-abusers. CCL5 is implicated as mediating the cytokine-driven amplification of CCL2 production by astrocytes and resultant macrophage/microglial recruitment and activation.
Collapse
Affiliation(s)
- Nazira El-Hage
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA 23298
| | - Annadora J. Bruce-Keller
- Neurodegeneration and Inflammation, Pennington Biomedical Research Center, Louisiana State University Baton Rouge, LA 70808
| | - Pamela E. Knapp
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298
- Institute for Drug and Addiction Research, Virginia Commonwealth University, Richmond, VA 23298
| | - Kurt F. Hauser
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA 23298
- Institute for Drug and Addiction Research, Virginia Commonwealth University, Richmond, VA 23298
| |
Collapse
|
48
|
Tambuyzer BR, Ponsaerts P, Nouwen EJ. Microglia: gatekeepers of central nervous system immunology. J Leukoc Biol 2008; 85:352-70. [DOI: 10.1189/jlb.0608385] [Citation(s) in RCA: 238] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
49
|
Butchi NB, Pourciau S, Du M, Morgan TW, Peterson KE. Analysis of the neuroinflammatory response to TLR7 stimulation in the brain: comparison of multiple TLR7 and/or TLR8 agonists. THE JOURNAL OF IMMUNOLOGY 2008; 180:7604-12. [PMID: 18490763 DOI: 10.4049/jimmunol.180.11.7604] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Activation of astrocytes and microglia and the production of proinflammatory cytokines and chemokines are often associated with virus infection in the CNS as well as a number of neurological diseases of unknown etiology. These inflammatory responses may be initiated by recognition of pathogen-associated molecular patterns (PAMPs) that stimulate TLRs. TLR7 and TLR8 were identified as eliciting antiviral effects when stimulated by viral ssRNA. In the present study, we examined the potential of TLR7 and/or TLR8 agonists to induce glial activation and neuroinflammation in the CNS by intracerebroventricular inoculation of TLR7 and/or TLR8 agonists in newborn mice. The TLR7 agonist imiquimod induced astrocyte activation and up-regulation of proinflammatory cytokines and chemokines, including IFN-beta, TNF, CCL2, and CXCL10. However, these responses were only of short duration when compared with responses induced by the TLR4 agonist LPS. Interestingly, some of the TLR7 and/or TLR8 agonists differed in their ability to activate glial cells as evidenced by their ability to induce cytokine and chemokine expression both in vivo and in vitro. Thus, TLR7 stimulation can induce neuroinflammatory responses in the brain, but individual TLR7 agonists may differ in their ability to stimulate cells of the CNS.
Collapse
Affiliation(s)
- Niranjan B Butchi
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | | | | | | | | |
Collapse
|
50
|
Benamar K, Geller EB, Adler MW. First in vivo evidence for a functional interaction between chemokine and cannabinoid systems in the brain. J Pharmacol Exp Ther 2008; 325:641-5. [PMID: 18281594 DOI: 10.1124/jpet.107.135053] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
Growing evidence supports the idea that in addition to their well established role in the immune system, chemokines might play a role in both normal and pathological brain function, and the chemokine network could interact with other neuromodulators. The chemokine stromal cell-derived growth factor (SDF)-1alpha/CXCL12, a member of the CXC chemokine family, was tested for its possible effect on the analgesic responses of the cannabinoid receptor agonist aminoalkylindole 4,5-dihydro-2-methyl-4-(4-morpholinylmethyl)-1-(1-naphthalenyl-carbonyl)-6H-pyrrolo-[3,2,1ij]quinolin-6-one [(+)-WIN 55,212-2, hereafter WIN 55,212-2] at the level of the periaqueductal gray (PAG), a brain region critical to the processing of pain signals, and a primary site of action of many analgesic compounds. The administration of WIN 55,212-2 (0.1-0.4 microg/microl) into the PAG resulted in antinociception in a dose-dependent manner. The selective cannabinoid (CB)1 antagonist N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide hydrochloride (SR 141716A; 1-10 microg) given into the PAG blocked the WIN 55,212-2-induced antinociception. In contrast, the selective CB2 antagonist N-[(1S)-endo-1,3,3-trimethyl bicyclo heptan-2-yl]-5-(4-chloro-3-methylphenyl)-1-(4-methylbenzyl)-pyrazole-3-carboxamide (SR144528; 10 microg) did not alter the WIN 55,212-2-induced antinociception. Pretreatment with SDF-1alpha/CXCL12 (100 ng) caused a reduction in antinociceptive responses of WIN 55,212-2. The inhibitory effect of SDF-1alpha/CXCL12 on WIN 55,212-2-induced antinociception was reversed by octahydrochloride [corrected] hydrate (AMD 3100) (10-50 ng), an antagonist of the SDF-1alpha/CXCL12, acting at its receptor, CXCR4. This study reports the first in vivo evidence of a functional interaction between chemokine and cannabinoid systems in the brain, showing that the activation of SDF-1alpha/CXCL12 receptors (CXCR4) in the PAG interferes with the analgesic effects of WIN 55212-2.
Collapse
MESH Headings
- Analgesics/therapeutic use
- Animals
- Benzoxazines/therapeutic use
- Camphanes/pharmacology
- Chemokine CXCL12/therapeutic use
- Male
- Morpholines/therapeutic use
- Naphthalenes/therapeutic use
- Pain/drug therapy
- Pain/metabolism
- Periaqueductal Gray/drug effects
- Periaqueductal Gray/metabolism
- Piperidines/pharmacology
- Pyrazoles/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB2/antagonists & inhibitors
- Receptor, Cannabinoid, CB2/metabolism
- Receptors, CXCR4/metabolism
- Rimonabant
Collapse
Affiliation(s)
- Khalid Benamar
- Center of Substance Abuse Research, Temple University School of Medicine, 3400 N. Broad St., Philadelphia, PA 19140, USA.
| | | | | |
Collapse
|