1
|
Coventry A, Sikorskii A, Zalwango SK, Familiar-Lopez I, Cardino VN, Giordani B, Ezeamama AE. In utero/peripartum antiretroviral therapy exposure and mental health outcomes at 8-18 years old: A longitudinal comparative study of children with perinatally acquired HIV, children perinatally HIV exposed but uninfected, and children unexposed uninfected from Uganda. Res Nurs Health 2024; 47:195-207. [PMID: 38031814 DOI: 10.1002/nur.22359] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/28/2023] [Accepted: 11/19/2023] [Indexed: 12/01/2023]
Abstract
In utero/peripartum antiretroviral therapy (IPA) exposure type was examined in relationship to mental health symptoms among 577 children with perinatally acquired HIV (CPHIV), children perinatally HIV exposed but uninfected (CHEU), and children HIV unexposed uninfected (CHUU). IPA exposure was categorized for CPHIV and CHEU as none, single-dose nevirapine with or without zidovudine (sdNVP±AZT), sdNVP+AZT+lamivudine (3TC), or combination antiretroviral therapy (cART). Anxiety and depressive symptoms were reported at baseline, 6-, and 12-month follow-up per behavioral assessment system for children. Multivariable linear mixed models were used to estimate differences (b) with 95% confidence intervals (95% CI) for IPA exposure types versus CHEU without IPA exposure. Depressive and anxiety symptoms were lower in CHUU relative to CHEU and CPHIV but did not differ between CPHIV and CHEU. CHEU with sdNVP±AZT exposure had greater anxiety (b = 0.51, 95% CI: [0.06, 0.96]) and depressive symptoms (b = 0.48, 95% CI: [0.07, 0.89]) than CHEU without IPA exposure. CHEU with sdNVP+AZT+3TC exposure had higher anxiety (b = 0.0.45, 95% CI: [0.03, 0.86]) and depressive symptoms (b = 0.72, 95% CI: [0.27, 1.17]) versus CHEU without IPA exposure. Depressive and anxiety symptoms were not different for CHEU and CPHIV exposed to cART (b = 0.12-0.60, 95% CI: [-0.41, 1.30]) and CHEU and CHUU (b = -0.04 to 0.08, 95% CI: [-0.24, 0.29]) without IPA exposure. Among CHEU, peripartum sdNVP±AZT and sdNVP+AZT+3TC but not cART compared to no IPA exposure was associated with clinically important elevations in anxiety and depressive symptoms. Monitoring of mental health trajectory of HIV-affected children considering IPA is needed to inform mental health interventions. Patient Contribution: Caregivers and their dependents provided consent for participation and collaborated with study team to identify mutually convenient times for protocol implementation.
Collapse
Affiliation(s)
- Audrey Coventry
- Department of Psychiatry, University of Washington, Seattle, Washington, USA
| | - Alla Sikorskii
- Department of Psychiatry, Michigan State University, East Lansing, Michigan, USA
| | - Sarah K Zalwango
- Public Health and Environment, Kampala Capital City Authority, Kampala, Uganda
| | | | - Vanessa N Cardino
- Department of Psychiatry, Michigan State University, East Lansing, Michigan, USA
| | - Bruno Giordani
- Departments of Psychiatry, Neurology, Psychology, and School of Nursing, University of Michigan, Ann Arbor, Michigan, USA
| | - Amara E Ezeamama
- Department of Psychiatry, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
2
|
Rosario-Rodríguez LJ, Cantres-Rosario YM, Carrasquillo-Carrión K, Rodríguez-De Jesús AE, Cartagena-Isern LJ, García-Requena LA, Roche-Lima A, Meléndez LM. Quantitative Proteomics Reveal That CB2R Agonist JWH-133 Downregulates NF-κB Activation, Oxidative Stress, and Lysosomal Exocytosis from HIV-Infected Macrophages. Int J Mol Sci 2024; 25:3246. [PMID: 38542221 PMCID: PMC10970132 DOI: 10.3390/ijms25063246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 04/13/2024] Open
Abstract
HIV-associated neurocognitive disorders (HAND) affect 15-55% of HIV-positive patients and effective therapies are unavailable. HIV-infected monocyte-derived macrophages (MDM) invade the brain of these individuals, promoting neurotoxicity. We demonstrated an increased expression of cathepsin B (CATB), a lysosomal protease, in monocytes and post-mortem brain tissues of women with HAND. Increased CATB release from HIV-infected MDM leads to neurotoxicity, and their secretion is associated with NF-κB activation, oxidative stress, and lysosomal exocytosis. Cannabinoid receptor 2 (CB2R) agonist, JWH-133, decreases HIV-1 replication, CATB secretion, and neurotoxicity from HIV-infected MDM, but the mechanisms are not entirely understood. We hypothesized that HIV-1 infection upregulates the expression of proteins associated with oxidative stress and that a CB2R agonist could reverse these effects. MDM were isolated from healthy women donors (n = 3), infected with HIV-1ADA, and treated with JWH-133. After 13 days post-infection, cell lysates were labeled by Tandem Mass Tag (TMT) and analyzed by LC/MS/MS quantitative proteomics bioinformatics. While HIV-1 infection upregulated CATB, NF-κB signaling, Nrf2-mediated oxidative stress response, and lysosomal exocytosis, JWH-133 treatment downregulated the expression of the proteins involved in these pathways. Our results suggest that JWH-133 is a potential alternative therapy against HIV-induced neurotoxicity and warrant in vivo studies to test its potential against HAND.
Collapse
Affiliation(s)
- Lester J. Rosario-Rodríguez
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan 00935, Puerto Rico;
| | - Yadira M. Cantres-Rosario
- Translational Proteomics Center, Center for Collaborative Research in Health Disparities, University of Puerto Rico-Medical Sciences Campus, San Juan 00935, Puerto Rico; (Y.M.C.-R.); (A.E.R.-D.J.)
| | - Kelvin Carrasquillo-Carrión
- Integrated Informatics Core, Center for Collaborative Research in Health Disparities, University of Puerto Rico-Medical Sciences Campus, San Juan 00935, Puerto Rico; (K.C.-C.); (A.R.-L.)
| | - Ana E. Rodríguez-De Jesús
- Translational Proteomics Center, Center for Collaborative Research in Health Disparities, University of Puerto Rico-Medical Sciences Campus, San Juan 00935, Puerto Rico; (Y.M.C.-R.); (A.E.R.-D.J.)
| | - Luz J. Cartagena-Isern
- Department of Biology, University of Puerto Rico-Río Piedras Campus, San Juan 00925, Puerto Rico; (L.J.C.-I.); (L.A.G.-R.)
| | - Luis A. García-Requena
- Department of Biology, University of Puerto Rico-Río Piedras Campus, San Juan 00925, Puerto Rico; (L.J.C.-I.); (L.A.G.-R.)
| | - Abiel Roche-Lima
- Integrated Informatics Core, Center for Collaborative Research in Health Disparities, University of Puerto Rico-Medical Sciences Campus, San Juan 00935, Puerto Rico; (K.C.-C.); (A.R.-L.)
| | - Loyda M. Meléndez
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan 00935, Puerto Rico;
- Translational Proteomics Center, Center for Collaborative Research in Health Disparities, University of Puerto Rico-Medical Sciences Campus, San Juan 00935, Puerto Rico; (Y.M.C.-R.); (A.E.R.-D.J.)
| |
Collapse
|
3
|
Levine AJ, Thadani C, Soontornniyomkij V, Lopez-Aranda MF, Mesa YG, Kitchen S, Rezek V, Silva A, Kolson DL. Behavioral and histological assessment of a novel treatment of neuroHIV in humanized mice. RESEARCH SQUARE 2023:rs.3.rs-3678629. [PMID: 38168407 PMCID: PMC10760308 DOI: 10.21203/rs.3.rs-3678629/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Neurocognitive deficits are prevalent among people living with HIV, likely due to chronic inflammation and oxidative stress in the brain. To date, no pharmaceutical treatments beyond antiretroviral therapy (ARV) has been shown to reduce risk for, or severity of, HIV-associated neurocognitive disorder. Here we investigate a novel compound, CDDO-Me, with documented neuroprotective effects via activation of the nrf2 and inhibition of the NFkB pathways. Methods We conducted three studies to assess the efficacy of CDDO-Me alone or in combination with antiretroviral therapy in humanized mice infected with HIV; behavioral, histopathological, and immunohistochemical. Results CDDO-Me in combination with ARV rescued social interaction deficits; however, only ARV was associated with preserved functioning in other behaviors, and CDDO-Me may have attenuated those benefits. A modest neuroprotective effect was found for CDDO-Me when administered with ARV, via preservation of PSD-95 expression; however, ARV alone had a more consistent protective effect. No significant changes in antioxidant enzyme expression levels were observed in CDDO-Me-treated animals. Only ARV use seemed to affect some antioxidant levels, indicating that it is ARV rather than CDDO-Me that is the major factor providing neuroprotection in this animal model. Finally, immunohistochemical analysis found that several cellular markers in various brain regions varied due to ARV rather than CDDO-Me. Conclusion Limited benefit of CDDO-Me on behavior and neuroprotection were observed. Instead, ARV was shown to be the more beneficial treatment. These experiments support the future use of this chimeric mouse for behavioral experiments in neuroHIV research.
Collapse
Affiliation(s)
| | | | | | | | | | - Scott Kitchen
- UCLA Humanized Mouse Core Laboratory, University of California
| | - Valerie Rezek
- UCLA Humanized Mouse Core Laboratory, University of California
| | | | | |
Collapse
|
4
|
Siddiqui A, He C, Lee G, Figueroa A, Slaughter A, Robinson-Papp J. Neuropathogenesis of HIV and emerging therapeutic targets. Expert Opin Ther Targets 2022; 26:603-615. [PMID: 35815686 PMCID: PMC9887458 DOI: 10.1080/14728222.2022.2100253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/07/2022] [Indexed: 02/02/2023]
Abstract
INTRODUCTION HIV infection causes a wide range of neurological complications, many of which are among the most common complications of chronic HIV infection in the era of combined antiretroviral therapy. These neurological conditions arise due to complex interactions between HIV viral proteins and neuronal and glial cells that lead to the activation of various inflammatory and neurotoxic pathways across the nervous system. AREAS COVERED This review summarizes the current literature on the pathogenesis and clinical manifestations of neurological injuries associated with HIV in the brain, spinal cord, and peripheral nervous system. Molecular pathways relevant for possible therapeutic targets or advancements are emphasized. Gaps in knowledge and current challenges in therapeutic design are also discussed. EXPERT OPINION Several challenges exist in the development of therapeutic targets for HIV-associated cognitive impairments. However, recent developments in drug delivery systems and treatment strategies are encouraging. Treatments for HIV-associated pain and peripheral sensory neuropathies currently consist of symptomatic management, but a greater understanding of their pathogenesis can lead to the development of targeted molecular therapies and disease-modifying therapies. HIV-associated autonomic dysfunction may affect the course of systemic disease via disrupted neuro-immune interactions; however, more research is needed to facilitate our understanding of how these processes present clinically.
Collapse
Affiliation(s)
- Alina Siddiqui
- Icahn School of Medicine at Mount Sinai, 5 East 98th Street, New York City, NY, 10029 USA
| | - Celestine He
- Icahn School of Medicine at Mount Sinai, 5 East 98th Street, New York City, NY, 10029 USA
| | - Gina Lee
- Icahn School of Medicine at Mount Sinai, 5 East 98th Street, New York City, NY, 10029 USA
| | - Alex Figueroa
- University of Texas at Southwestern Medical School, Dallas, TX, 75390 USA
| | - Alexander Slaughter
- Icahn School of Medicine at Mount Sinai, 5 East 98th Street, New York City, NY, 10029 USA
| | - Jessica Robinson-Papp
- Icahn School of Medicine at Mount Sinai, 5 East 98th Street, New York City, NY, 10029 USA
| |
Collapse
|
5
|
Induction of HOXA3 by PRRSV inhibits IFN-I response through negatively regulation of HO-1 transcription. J Virol 2021; 96:e0186321. [PMID: 34851144 DOI: 10.1128/jvi.01863-21] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Type I interferons (IFN-I) play a key role in the host defense against virus infection, but porcine reproductive and respiratory syndrome virus (PRRSV) infection does not effectively activate IFN-I response, and the underlying molecular mechanisms are poorly characterized. In this study, a novel transcription factor of the heme oxygenase-1 (HO-1) gene, homeobox A3 (HOXA3), was screened and identified. Here, we found that HOXA3 was significantly increased during PRRSV infection. We demonstrated that HOXA3 promotes PRRSV replication by negatively regulating the HO-1 gene transcription, which is achieved by regulating type I interferons (IFN-I) production. A detailed analysis showed that PRRSV exploits HOXA3 to suppress beta interferon (IFN-β) and IFN-stimulated gene (ISG) expression in host cells. We also provide direct evidence that the activation of IFN-I by HO-1 depends on its interaction with IRF3. Then we further proved that deficiency of HOXA3 promoted the HO-1-IRF3 interaction, and subsequently enhanced IRF3 phosphorylation and nuclear translocation in PRRSV-infected cells. These data suggest that PRRSV uses HOXA3 to negatively regulate the transcription of the HO-1 gene to suppress the IFN-I response for immune evasion. IMPORTANCE Porcine reproductive and respiratory syndrome (PRRS), caused by PRRSV, leads the pork industry worldwide to significant economic losses. HOXA3 is generally considered to be an important molecule in the process of body development and cell differentiation. Here, we found a novel transcription factor of the HO-1 gene, HOXA3, can negatively regulate the transcription of the HO-1 gene and play an important role in the suppression of IFN-I response by PRRSV. PRRSV induces the upregulation of HOXA3, which can negatively regulate HO-1 gene transcription, thereby weakening the interaction between HO-1 and IRF3 for inhibiting the type I IFN response. This study extends the function of HOXA3 to the virus field for the first time and provides new insights into PRRSV immune evasion mechanism.
Collapse
|
6
|
HIV-Associated Neurotoxicity: The Interplay of Host and Viral Proteins. Mediators Inflamm 2021; 2021:1267041. [PMID: 34483726 PMCID: PMC8410439 DOI: 10.1155/2021/1267041] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/12/2021] [Accepted: 08/09/2021] [Indexed: 11/18/2022] Open
Abstract
HIV-1 can incite activation of chemokine receptors, inflammatory mediators, and glutamate receptor-mediated excitotoxicity. The mechanisms associated with such immune activation can disrupt neuronal and glial functions. HIV-associated neurocognitive disorder (HAND) is being observed since the beginning of the AIDS epidemic due to a change in the functional integrity of cells from the central nervous system (CNS). Even with the presence of antiretroviral therapy, there is a decline in the functioning of the brain especially movement skills, noticeable swings in mood, and routine performance activities. Under the umbrella of HAND, various symptomatic and asymptomatic conditions are categorized and are on a rise despite the use of newer antiretroviral agents. Due to the use of long-lasting antiretroviral agents, this deadly disease is becoming a manageable chronic condition with the occurrence of asymptomatic neurocognitive impairment (ANI), symptomatic mild neurocognitive disorder, or HIV-associated dementia. In-depth research in the pathogenesis of HIV has focused on various mechanisms involved in neuronal dysfunction and associated toxicities ultimately showcasing the involvement of various pathways. Increasing evidence-based studies have emphasized a need to focus and explore the specific pathways in inflammation-associated neurodegenerative disorders. In the current review, we have highlighted the association of various HIV proteins and neuronal cells with their involvement in various pathways responsible for the development of neurotoxicity.
Collapse
|
7
|
Confound, Cause, or Cure: The Effect of Cannabinoids on HIV-Associated Neurological Sequelae. Viruses 2021; 13:v13071242. [PMID: 34206839 PMCID: PMC8310358 DOI: 10.3390/v13071242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/12/2021] [Accepted: 06/15/2021] [Indexed: 12/15/2022] Open
Abstract
The persistence of human immunodeficiency virus-1 (HIV)-associated neurocognitive disorders (HAND) in the era of effective antiretroviral therapy suggests that modern HIV neuropathogenesis is driven, at least in part, by mechanisms distinct from the viral life cycle. Identifying more subtle mechanisms is complicated by frequent comorbidities in HIV+ populations. One of the common confounds is substance abuse, with cannabis being the most frequently used psychoactive substance among people living with HIV. The psychoactive effects of cannabis use can themselves mimic, and perhaps magnify, the cognitive deficits observed in HAND; however, the neuromodulatory and anti-inflammatory properties of cannabinoids may counter HIV-induced excitotoxicity and neuroinflammation. Here, we review our understanding of the cross talk between HIV and cannabinoids in the central nervous system by exploring both clinical observations and evidence from preclinical in vivo and in vitro models. Additionally, we comment on recent advances in human, multi-cell in vitro systems that allow for more translatable, mechanistic studies of the relationship between cannabinoid pharmacology and this uniquely human virus.
Collapse
|
8
|
Ezeamama AE, Zalwango SK, Sikorskii A, Tuke R, Musoke PM, Giordani B, Boivin MJ. In utero and peripartum antiretroviral exposure as predictor of cognition in 6- to 10-year-old HIV-exposed Ugandan children - a prospective cohort study. HIV Med 2021; 22:592-604. [PMID: 33860626 DOI: 10.1111/hiv.13094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/28/2021] [Accepted: 02/08/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To quantify association between in utero/peripartum antiretroviral (IPA) exposure and cognition, i.e. executive function (EF) and socioemotional adjustment (SEA), in school-aged Ugandan children who were perinatally HIV-infected (CPHIV, n = 100) and children who were HIV-exposed but uninfected (CHEU, n = 101). METHODS Children were enrolled at age 6-10 years and followed for 12 months from March 2017 to December 2018. Caregiver-reported child EF and SEA competencies were assessed using validated questionnaires at baseline, 6 and 12 months. IPA type - combination antiretroviral therapy (cART), intrapartum single-dose nevirapine ± zidovudine (sdNVP ± ZDV), nevirapine + zidovudine + lamivudine (sdNVP + ZDV + 3TC) - or no IPA (reference) was verified via medical records. IPA-related standardized mean differences (SMDs) with corresponding 95% confidence intervals (CIs) in cognitive competencies were estimated from regression models with adjustment for caregiver sociodemographic and contextual factors. Models were fitted separately for CPHIV and CHEU. RESULTS Among CPHIV children, cART (SMD = -0.82, 95% CI: -1.37 to -0.28) and sdNVP ± ZDV (SMD = -0.41, 95% CI: -0.81 to -0.00) vs. no IPA predicted lower executive dysfunction over 12 months. Intrapartum sdNVP + ZDV + 3TC vs. no IPA predicted executive dysfunction (SMD = 0.80, 95% CI: 0.30-1.31), SEA problems (SMD = 0.63-0.76, 95% CI: 0.00-1.24) and lower adaptive skills (SMD = -0.36, 95% CI: -0.75-0.02) over 12 months among CHEU. Further adjustment for contextual factors attenuated associations, although most remained of moderate clinical importance (|SMD| > 0.33). CONCLUSIONS Among CPHIV children, cART and sdNVP ± ZDV IPA exposure predicted, on average, lower executive dysfunction 6-10 years later. However, peripartum sdNVP + ZDV + 3TC predicted executive and SEA dysfunction among CHEU 6-10 years later. These data underscore the need for more research into long-term effects of in utero ART to inform development of appropriate interventions so as to mitigate cognitive sequelae.
Collapse
Affiliation(s)
- A E Ezeamama
- Department of Psychiatry, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - S K Zalwango
- Directorate of Public Health and Environment, Kampala Capital City Authority, Kampala, Uganda
| | - A Sikorskii
- Department of Psychiatry, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - R Tuke
- Department of Psychiatry, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - P M Musoke
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda.,Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda
| | - B Giordani
- Departments of Psychiatry, Neurology, and Psychology, University of Michigan, Ann Arbor, MI, USA
| | - M J Boivin
- Departments of Psychiatry and Neurology & Ophthamology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA.,Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
9
|
Dimethyl Fumarate, an Approved Multiple Sclerosis Treatment, Reduces Brain Oxidative Stress in SIV-Infected Rhesus Macaques: Potential Therapeutic Repurposing for HIV Neuroprotection. Antioxidants (Basel) 2021; 10:antiox10030416. [PMID: 33803289 PMCID: PMC7998206 DOI: 10.3390/antiox10030416] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 12/16/2022] Open
Abstract
Dimethyl fumarate (DMF), an antioxidant/anti-inflammatory drug approved for the treatment of multiple sclerosis, induces antioxidant enzymes, in part through transcriptional upregulation. We hypothesized that DMF administration to simian immunodeficiency virus (SIV)-infected rhesus macaques would induce antioxidant enzyme expression and reduce oxidative injury and inflammation throughout the brain. Nine SIV-infected, CD8+-T-lymphocyte-depleted rhesus macaques were studied. Five received oral DMF prior to the SIV infection and through to the necropsy day. Protein expression was analyzed in 11 brain regions, as well as the thymus, liver, and spleen, using Western blot and immunohistochemistry for antioxidant, inflammatory, and neuronal proteins. Additionally, oxidative stress was determined in brain sections using immunohistochemistry (8-OHdG, 3NT) and optical redox imaging of oxidized flavoproteins containing flavin adenine dinucleotide (Fp) and reduced nicotinamide adenine dinucleotide (NADH). The DMF treatment was associated with no changes in virus replication; higher expressions of the antioxidant enzymes NQO1, GPX1, and HO-1 in the brain and PRDX1 and HO-2 in the spleen; lower levels of 8-OHdG and 3NT; a lower optical redox ratio. The DMF treatment was also associated with increased expressions of cell-adhesion molecules (VCAM-1, ICAM-1) and no changes in HLA-DR, CD68, GFAP, NFL, or synaptic proteins. The concordantly increased brain antioxidant enzyme expressions and reduced oxidative stress in DMF-treated SIV-infected macaques suggest that DMF could limit oxidative stress throughout the brain through effective induction of the endogenous antioxidant response. We propose that DMF could potentially induce neuroprotective brain responses in persons living with HIV.
Collapse
|
10
|
Yuan NY, Kaul M. Beneficial and Adverse Effects of cART Affect Neurocognitive Function in HIV-1 Infection: Balancing Viral Suppression against Neuronal Stress and Injury. J Neuroimmune Pharmacol 2021; 16:90-112. [PMID: 31385157 PMCID: PMC7233291 DOI: 10.1007/s11481-019-09868-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023]
Abstract
HIV-associated neurocognitive disorders (HAND) persist despite the successful introduction of combination antiretroviral therapy (cART). While insufficient concentration of certain antiretrovirals (ARV) may lead to incomplete viral suppression in the brain, many ARVs are found to cause neuropsychiatric adverse effects, indicating their penetration into the central nervous system (CNS). Several lines of evidence suggest shared critical roles of oxidative and endoplasmic reticulum stress, compromised neuronal energy homeostasis, and autophagy in the promotion of neuronal dysfunction associated with both HIV-1 infection and long-term cART or ARV use. As the lifespans of HIV patients are increased, unique challenges have surfaced. Longer lives convey prolonged exposure of the CNS to viral toxins, neurotoxic ARVs, polypharmacy with prescribed or illicit drug use, and age-related diseases. All of these factors can contribute to increased risks for the development of neuropsychiatric conditions and cognitive impairment, which can significantly impact patient well-being, cART adherence, and overall health outcome. Strategies to increase the penetration of cART into the brain to lower viral toxicity may detrimentally increase ARV neurotoxicity and neuropsychiatric adverse effects. As clinicians attempt to control peripheral viremia in an aging population of HIV-infected patients, they must navigate an increasingly complex myriad of comorbidities, pharmacogenetics, drug-drug interactions, and psychiatric and cognitive dysfunction. Here we review in comparison to the neuropathological effects of HIV-1 the available information on neuropsychiatric adverse effects and neurotoxicity of clinically used ARV and cART. It appears altogether that future cART aiming at controlling HIV-1 in the CNS and preventing HAND will require an intricate balancing act of suppressing viral replication while minimizing neurotoxicity, impairment of neurocognition, and neuropsychiatric adverse effects. Graphical abstract Schematic summary of the effects exerted on the brain and neurocognitive function by HIV-1 infection, comorbidities, psychostimulatory, illicit drugs, therapeutic drugs, such as antiretrovirals, the resulting polypharmacy and aging, as well as the potential interactions of all these factors.
Collapse
Affiliation(s)
- Nina Y Yuan
- School of Medicine, Division of Biomedical Sciences, University of California Riverside, 900 University Ave, Riverside, CA, 92521, USA
| | - Marcus Kaul
- School of Medicine, Division of Biomedical Sciences, University of California Riverside, 900 University Ave, Riverside, CA, 92521, USA.
- Sanford Burnham Prebys Medical Discovery Institute, Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|
11
|
Regional Brain Recovery from Acute Synaptic Injury in Simian Immunodeficiency Virus-Infected Rhesus Macaques Associates with Heme Oxygenase Isoform Expression. J Virol 2020; 94:JVI.01102-20. [PMID: 32669339 PMCID: PMC7495379 DOI: 10.1128/jvi.01102-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/10/2020] [Indexed: 12/21/2022] Open
Abstract
Brain injury induced by acute simian (or human) immunodeficiency virus infection may persist or spontaneously resolve in different brain regions. Identifying the host factor(s) that promotes spontaneous recovery from such injury may reveal targets for therapeutic drug strategies for promoting recovery from acute neuronal injury. The gradual recovery from such injury observed in many, but not all, brain regions in the rhesus macaque model is consistent with the possible existence of a therapeutic window of opportunity for intervening to promote recovery, even in those regions not showing spontaneous recovery. In persons living with human immunodeficiency virus infection, such neuroprotective treatments could ultimately be considered as adjuncts to the initiation of antiretroviral drug therapy. Brain injury occurs within days in simian immunodeficiency virus (SIV) or human immunodeficiency virus (HIV) infection, and some recovery may occur within weeks. Inflammation and oxidative stress associate with such injury, but what drives recovery is unknown. Chronic HIV infection associates with reduced brain frontal cortex expression of the antioxidant/anti-inflammatory enzyme heme oxygenase-1 (HO-1) and increased neuroinflammation in individuals with cognitive impairment. We hypothesized that acute regional brain injury and recovery associate with differences in regional brain HO-1 expression. Using SIV-infected rhesus macaques, we analyzed multiple brain regions through acute and chronic infection (90 days postinfection [dpi]) and quantified viral (SIV gag RNA), synaptic (PSD-95; synaptophysin), axonal (neurofilament/neurofilament light chain [NFL]), inflammatory, and antioxidant (enzymes, including heme oxygenase isoforms [HO-1, HO-2]) markers. PSD-95 was reduced in the brainstem, basal ganglia, neocortex, and cerebellum within 13 dpi, indicating acute synaptic injury throughout the brain. All areas except the brainstem recovered. Unchanged NFL was consistent with no acute axonal injury. SIV RNA expression was highest in the brainstem throughout infection, and it associated with neuroinflammation. Surprisingly, during the synaptic injury and recovery phases, HO-2, and not HO-1, progressively decreased in the brainstem. Thus, acute SIV synaptic injury occurs throughout the brain, with spontaneous recovery in regions other than the brainstem. Within the brainstem, the high SIV load and inflammation, along with reduction of HO-2, may impair recovery. In other brain regions, stable HO-2 expression, with or without increasing HO-1, may promote recovery. Our data support roles for heme oxygenase isoforms in modulating recovery from synaptic injury in SIV infection and suggest their therapeutic targeting for promoting neuronal recovery. IMPORTANCE Brain injury induced by acute simian (or human) immunodeficiency virus infection may persist or spontaneously resolve in different brain regions. Identifying the host factor(s) that promotes spontaneous recovery from such injury may reveal targets for therapeutic drug strategies for promoting recovery from acute neuronal injury. The gradual recovery from such injury observed in many, but not all, brain regions in the rhesus macaque model is consistent with the possible existence of a therapeutic window of opportunity for intervening to promote recovery, even in those regions not showing spontaneous recovery. In persons living with human immunodeficiency virus infection, such neuroprotective treatments could ultimately be considered as adjuncts to the initiation of antiretroviral drug therapy.
Collapse
|
12
|
Gruenewald AL, Garcia-Mesa Y, Gill AJ, Garza R, Gelman BB, Kolson DL. Neuroinflammation associates with antioxidant heme oxygenase-1 response throughout the brain in persons living with HIV. J Neurovirol 2020; 26:846-862. [PMID: 32910432 PMCID: PMC7716923 DOI: 10.1007/s13365-020-00902-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/17/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022]
Abstract
Previous studies showed that persons living with HIV (PLWH) demonstrate higher brain prefrontal cortex neuroinflammation and immunoproteasome expression compared to HIV-negative individuals; these associate positively with HIV levels. Lower expression of the antioxidant enzyme heme oxygenase 1 (HO-1) was observed in PLWH with HIV-associated neurocognitive impairment (HIV-NCI) compared to neurocognitively normal PLWH. We hypothesized that similar expression patterns occur throughout cortical, subcortical, and brainstem regions in PLWH, and that neuroinflammation and immunoproteasome expression associate with lower expression of neuronal markers. We analyzed autopsied brains (15 regions) from 9 PLWH without HIV-NCI and 7 matched HIV-negative individuals. Using Western blot and RT-qPCR, we quantified synaptic, inflammatory, immunoproteasome, endothelial, and antioxidant biomarkers, including HO-1 and its isoform heme oxygenase 2 (HO-2). In these PLWH without HIV-NCI, we observed higher expression of neuroinflammatory, endothelial, and immunoproteasome markers in multiple cortical and subcortical regions compared to HIV-negative individuals, suggesting a global brain inflammatory response to HIV. Several regions, including posterior cingulate cortex, globus pallidus, and cerebellum, showed a distinct pattern of higher type I interferon (IFN)-stimulated gene and immunoproteasome expression. PLWH without HIV-NCI also had (i) stable or higher HO-1 expression and positive associations between (ii) HO-1 and HIV levels (CSF, plasma) and (iii) HO-1 expression and neuroinflammation, in multiple cortical, subcortical, and brainstem regions. We observed no differences in synaptic marker expression, suggesting little, if any, associated neuronal injury. We speculate that this may reflect a neuroprotective effect of a concurrent HO-1 antioxidant response despite global neuroinflammation, which will require further investigation.
Collapse
Affiliation(s)
- Analise L Gruenewald
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 280 Clinical Research Building, 415 Curie Blvd., Philadelphia, PA, 19104, USA
| | - Yoelvis Garcia-Mesa
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 280 Clinical Research Building, 415 Curie Blvd., Philadelphia, PA, 19104, USA
| | - Alexander J Gill
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 280 Clinical Research Building, 415 Curie Blvd., Philadelphia, PA, 19104, USA
| | - Rolando Garza
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 280 Clinical Research Building, 415 Curie Blvd., Philadelphia, PA, 19104, USA
| | - Benjamin B Gelman
- Department of Pathology, University of Texas Medical Branch, 301 University Blvd., Keiller 3.118A, Route 0609, Galveston, TX, 77555, USA
| | - Dennis L Kolson
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 280 Clinical Research Building, 415 Curie Blvd., Philadelphia, PA, 19104, USA.
| |
Collapse
|
13
|
León-Rivera R, Morsey B, Niu M, Fox HS, Berman JW. Interactions of Monocytes, HIV, and ART Identified by an Innovative scRNAseq Pipeline: Pathways to Reservoirs and HIV-Associated Comorbidities. mBio 2020; 11:e01037-20. [PMID: 32723919 PMCID: PMC7387797 DOI: 10.1128/mbio.01037-20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/22/2020] [Indexed: 12/31/2022] Open
Abstract
HIV reservoirs persist despite successful antiretroviral therapy (ART) and are a major obstacle to the eradication and cure of HIV. The mature monocyte subset, CD14+CD16+, contributes to viral reservoirs and HIV-associated comorbidities. Only a subset of monocytes harbors HIV (HIV+), while the rest remain uninfected, exposed cells (HIVexp). We developed an innovative single cell RNA sequencing (scRNAseq) pipeline that detects HIV and host transcripts simultaneously, enabling us to examine differences between HIV+ and HIVexp mature monocytes. Using this, we characterized uninfected, HIV+, and HIVexp primary human mature monocytes with and without ART. We showed that HIV+ mature monocytes do not form their own cluster separately from HIVexp but can be distinguished by significant differential gene expression. We found that ART decreased levels of unspliced HIV transcripts potentially by modulating host transcriptional regulators shown to decrease viral infection and replication. We also identified and characterized mature monocyte subpopulations differentially impacted by HIV and ART. We identified genes dysregulated by ART in HIVexp monocytes compared to their uninfected counterpart and, of interest, the junctional protein ALCAM, suggesting that ART impacts monocyte functions. Our data provide a novel method for simultaneous detection of HIV and host transcripts. We identify potential targets, such as those genes whose expression is increased in HIV+ mature monocytes compared to HIVexp, to block their entry into tissues, preventing establishment/replenishment of HIV reservoirs even with ART, thereby reducing and/or eliminating viral burden and HIV-associated comorbidities. Our data also highlight the heterogeneity of mature monocyte subsets and their potential contributions to HIV pathogenesis in the ART era.IMPORTANCE HIV enters tissues early after infection, leading to establishment and persistence of HIV reservoirs despite antiretroviral therapy (ART). Viral reservoirs are a major obstacle to the eradication and cure of HIV. CD14+CD16+ (mature) monocytes may contribute to establishment and reseeding of reservoirs. A subset of monocytes, consisting mainly of CD14+CD16+ cells, harbors HIV (HIV+), while the rest remain uninfected, exposed cells (HIVexp). It is important to identify cells harboring virus to eliminate reservoirs. Using an innovative single-cell RNA sequencing (scRNAseq) pipeline to detect HIV and host transcripts simultaneously, we characterized HIV+ and HIVexp primary human mature monocytes with and without ART. HIV+ mature monocytes are not a unique subpopulation but rather can be distinguished from HIVexp by differential gene expression. We characterized mature monocyte subpopulations differently impacted by HIV and ART, highlighting their potential contributions to HIV-associated comorbidities. Our data propose therapeutic targets to block HIV+ monocyte entry into tissues, preventing establishment and replenishment of reservoirs even with ART.
Collapse
Affiliation(s)
- Rosiris León-Rivera
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Brenda Morsey
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Meng Niu
- Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Howard S Fox
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Joan W Berman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
14
|
Garza R, Gill AJ, Bastien BL, Garcia-Mesa Y, Gruenewald AL, Gelman BB, Tsima B, Gross R, Letendre SL, Kolson DL. Heme oxygenase-1 promoter (GT) n polymorphism associates with HIV neurocognitive impairment. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2020; 7:e710. [PMID: 32277015 PMCID: PMC7176253 DOI: 10.1212/nxi.0000000000000710] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/21/2020] [Indexed: 01/21/2023]
Abstract
OBJECTIVE To determine whether regulatory variations in the heme oxygenase-1 (HO-1) promoter (GT) n dinucleotide repeat length could identify unique population genetic risks for neurocognitive impairment (NCI) in persons living with HIV (PLWH), we genotyped 528 neurocognitively assessed PLWH of European American and African American descent and linked genotypes to cognitive status. METHODS In this cross-sectional study of PLWH (the CNS HIV Antiretroviral Therapy Effect Research cohort), we determined HO-1 (GT) n repeat lengths in 276 African Americans and 252 European Americans. Using validated criteria for HIV-associated NCI (HIV NCI), we found associations between allele length genotypes and HIV NCI and between genotypes and plasma markers of monocyte activation and inflammation. For comparison of HO-1 (GT) n allele frequencies with another population of African ancestry, we determined HO-1 (GT) n allele lengths in African PLWH from Botswana (n = 428). RESULTS PLWH with short HO-1 (GT) n alleles had a lower risk for HIV NCI (OR = 0.63, 95% CI: 0.42-0.94). People of African ancestry had a lower prevalence of short alleles and higher prevalence of long alleles compared with European Americans, and in subgroup analyses, the protective effect of the short allele was observed in African Americans and not in European Americans. CONCLUSIONS Our study identified the short HO-1 (GT) n allele as partially protective against developing HIV NCI. It further suggests that this clinical protective effect is particularly relevant in persons of African ancestry, where the lower prevalence of short HO-1 (GT) n alleles may limit induction of HO-1 expression in response to inflammation and oxidative stress. Therapeutic strategies that enhance HO-1 expression may decrease HIV-associated neuroinflammation and limit HIV NCI.
Collapse
Affiliation(s)
- Rolando Garza
- From the Department of Neurology (R. Garza, A.J.G., B.L.B., Y.G.-M., A.L.G., D.L.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Department of Pathology (B.B.G.), University of Texas Medical Branch, Galveston; Department of Family Medicine & Public Health (B.T.), University of Botswana, Gaborone; Departments of Medicine and Biostatistics (R. Gross), Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Department of Medicine (S.L.L.), University of California, San Diego
| | - Alexander J Gill
- From the Department of Neurology (R. Garza, A.J.G., B.L.B., Y.G.-M., A.L.G., D.L.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Department of Pathology (B.B.G.), University of Texas Medical Branch, Galveston; Department of Family Medicine & Public Health (B.T.), University of Botswana, Gaborone; Departments of Medicine and Biostatistics (R. Gross), Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Department of Medicine (S.L.L.), University of California, San Diego
| | - Brandon L Bastien
- From the Department of Neurology (R. Garza, A.J.G., B.L.B., Y.G.-M., A.L.G., D.L.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Department of Pathology (B.B.G.), University of Texas Medical Branch, Galveston; Department of Family Medicine & Public Health (B.T.), University of Botswana, Gaborone; Departments of Medicine and Biostatistics (R. Gross), Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Department of Medicine (S.L.L.), University of California, San Diego
| | - Yoelvis Garcia-Mesa
- From the Department of Neurology (R. Garza, A.J.G., B.L.B., Y.G.-M., A.L.G., D.L.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Department of Pathology (B.B.G.), University of Texas Medical Branch, Galveston; Department of Family Medicine & Public Health (B.T.), University of Botswana, Gaborone; Departments of Medicine and Biostatistics (R. Gross), Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Department of Medicine (S.L.L.), University of California, San Diego
| | - Analise L Gruenewald
- From the Department of Neurology (R. Garza, A.J.G., B.L.B., Y.G.-M., A.L.G., D.L.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Department of Pathology (B.B.G.), University of Texas Medical Branch, Galveston; Department of Family Medicine & Public Health (B.T.), University of Botswana, Gaborone; Departments of Medicine and Biostatistics (R. Gross), Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Department of Medicine (S.L.L.), University of California, San Diego
| | - Benjamin B Gelman
- From the Department of Neurology (R. Garza, A.J.G., B.L.B., Y.G.-M., A.L.G., D.L.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Department of Pathology (B.B.G.), University of Texas Medical Branch, Galveston; Department of Family Medicine & Public Health (B.T.), University of Botswana, Gaborone; Departments of Medicine and Biostatistics (R. Gross), Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Department of Medicine (S.L.L.), University of California, San Diego
| | - Billy Tsima
- From the Department of Neurology (R. Garza, A.J.G., B.L.B., Y.G.-M., A.L.G., D.L.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Department of Pathology (B.B.G.), University of Texas Medical Branch, Galveston; Department of Family Medicine & Public Health (B.T.), University of Botswana, Gaborone; Departments of Medicine and Biostatistics (R. Gross), Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Department of Medicine (S.L.L.), University of California, San Diego
| | - Robert Gross
- From the Department of Neurology (R. Garza, A.J.G., B.L.B., Y.G.-M., A.L.G., D.L.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Department of Pathology (B.B.G.), University of Texas Medical Branch, Galveston; Department of Family Medicine & Public Health (B.T.), University of Botswana, Gaborone; Departments of Medicine and Biostatistics (R. Gross), Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Department of Medicine (S.L.L.), University of California, San Diego
| | - Scott L Letendre
- From the Department of Neurology (R. Garza, A.J.G., B.L.B., Y.G.-M., A.L.G., D.L.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Department of Pathology (B.B.G.), University of Texas Medical Branch, Galveston; Department of Family Medicine & Public Health (B.T.), University of Botswana, Gaborone; Departments of Medicine and Biostatistics (R. Gross), Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Department of Medicine (S.L.L.), University of California, San Diego
| | - Dennis L Kolson
- From the Department of Neurology (R. Garza, A.J.G., B.L.B., Y.G.-M., A.L.G., D.L.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Department of Pathology (B.B.G.), University of Texas Medical Branch, Galveston; Department of Family Medicine & Public Health (B.T.), University of Botswana, Gaborone; Departments of Medicine and Biostatistics (R. Gross), Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Department of Medicine (S.L.L.), University of California, San Diego.
| |
Collapse
|
15
|
Murphy A, Barbaro J, Martínez-Aguado P, Chilunda V, Jaureguiberry-Bravo M, Berman JW. The Effects of Opioids on HIV Neuropathogenesis. Front Immunol 2019; 10:2445. [PMID: 31681322 PMCID: PMC6813247 DOI: 10.3389/fimmu.2019.02445] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/01/2019] [Indexed: 12/13/2022] Open
Abstract
HIV associated neurocognitive disorders (HAND) are a group of neurological deficits that affect approximately half of people living with HIV (PLWH) despite effective antiretroviral therapy (ART). There are currently no reliable molecular biomarkers or treatments for HAND. Given the national opioid epidemic, as well as illegal and prescription use of opioid drugs among PLWH, it is critical to characterize the molecular interactions between HIV and opioids in cells of the CNS. It is also important to study the role of opioid substitution therapies in the context of HIV and CNS damage in vitro and in vivo. A major mechanism contributing to HIV neuropathogenesis is chronic, low-level inflammation in the CNS. HIV enters the brain within 4–8 days after peripheral infection and establishes CNS reservoirs, even in the context of ART, that are difficult to identify and eliminate. Infected cells, including monocytes, macrophages, and microglia, produce chemokines, cytokines, neurotoxic mediators, and viral proteins that contribute to chronic inflammation and ongoing neuronal damage. Opioids have been shown to impact these immune cells through a variety of molecular mechanisms, including opioid receptor binding and cross desensitization with chemokine receptors. The effects of opioid use on cognitive outcomes in individuals with HAND in clinical studies is variable, and thus multiple biological mechanisms are likely to contribute to the complex relationship between opioids and HIV in the CNS. In this review, we will examine what is known about both HIV and opioid mediated neuropathogenesis, and discuss key molecular processes that may be impacted by HIV and opioids in the context of neuroinflammation and CNS damage. We will also assess what is known about the effects of ART on these processes, and highlight areas of study that should be addressed in the context of ART.
Collapse
Affiliation(s)
- Aniella Murphy
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - John Barbaro
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Pablo Martínez-Aguado
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Vanessa Chilunda
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Matias Jaureguiberry-Bravo
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Joan W Berman
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States.,Laboratory of Dr. Joan W. Berman, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
16
|
Rojas-Celis V, Valiente-Echeverría F, Soto-Rifo R, Toro-Ascuy D. New Challenges of HIV-1 Infection: How HIV-1 Attacks and Resides in the Central Nervous System. Cells 2019; 8:cells8101245. [PMID: 31614895 PMCID: PMC6829584 DOI: 10.3390/cells8101245] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/05/2019] [Accepted: 10/11/2019] [Indexed: 12/15/2022] Open
Abstract
Acquired immunodeficiency syndrome (AIDS) has become one of the most devastating pandemics in recorded history. The main causal agent of AIDS is the human immunodeficiency virus (HIV), which infects various cell types of the immune system that express the CD4 receptor on their surfaces. Today, combined antiretroviral therapy (cART) is the standard treatment for all people with HIV; although it has improved the quality of life of people living with HIV (PLWH), it cannot eliminate the latent reservoir of the virus. Therefore HIV/AIDS has turned from a fatal disease to a chronic disease requiring lifelong treatment. Despite significant viral load suppression, it has been observed that at least half of patients under cART present HIV-associated neurocognitive disorders (HAND), which have been related to HIV-1 infection and replication in the central nervous system (CNS). Several studies have focused on elucidating the mechanism by which HIV-1 can invade the CNS and how it can generate the effects seen in HAND. This review summarizes the research on HIV-1 and its interaction with the CNS with an emphasis on the generation of HAND, how the virus enters the CNS, the relationship between HIV-1 and cells of the CNS, and the effect of cART on these cells.
Collapse
Affiliation(s)
- Victoria Rojas-Celis
- Instituto de Ciencias Biomedicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910060, Chile.
| | - Fernando Valiente-Echeverría
- Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad of Chile, Santiago 8389100, Chile.
| | - Ricardo Soto-Rifo
- Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad of Chile, Santiago 8389100, Chile.
| | - Daniela Toro-Ascuy
- Instituto de Ciencias Biomedicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910060, Chile.
| |
Collapse
|
17
|
Ramezani A, Nahad MP, Faghihloo E. The role of Nrf2 transcription factor in viral infection. J Cell Biochem 2018; 119:6366-6382. [DOI: 10.1002/jcb.26897] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/28/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Ali Ramezani
- Virology DepartmentSchool of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
- Hepatitis Research CenterBirjand University of Medical SciencesBirjandIran
| | - Mehdi Parsa Nahad
- Virology DepartmentSchool of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Ebrahim Faghihloo
- Department of MicrobiologySchool of MedicineShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
18
|
HIV and the Macrophage: From Cell Reservoirs to Drug Delivery to Viral Eradication. J Neuroimmune Pharmacol 2018; 14:52-67. [PMID: 29572681 DOI: 10.1007/s11481-018-9785-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/16/2018] [Indexed: 12/25/2022]
Abstract
Macrophages serve as host cells, inflammatory disease drivers and drug runners for human immunodeficiency virus infection and treatments. Low-level viral persistence continues in these cells in the absence of macrophage death. However, the cellular microenvironment changes as a consequence of viral infection with aberrant production of pro-inflammatory factors and promotion of oxidative stress. These herald viral spread from macrophages to neighboring CD4+ T cells and end organ damage. Virus replicates in tissue reservoir sites that include the nervous, pulmonary, cardiovascular, gut, and renal organs. However, each of these events are held in check by antiretroviral therapy. A hidden and often overlooked resource of the macrophage rests in its high cytoplasmic nuclear ratios that allow the cell to sense its environment and rid it of the cellular waste products and microbial pathogens it encounters. These phagocytic and intracellular killing sensing mechanisms can also be used in service as macrophages serve as cellular carriage depots for antiretroviral nanoparticles and are able to deliver medicines to infectious disease sites with improved therapeutic outcomes. These undiscovered cellular functions can lead to reductions in persistent infection and may potentially facilitate the eradication of residual virus to eliminate disease.
Collapse
|
19
|
Budiarti R, Kuntaman, Nasronudin, Suryokusumo, Khairunisa SQ. IN VITRO STUDIES ON HEME OXYGENASE-1 AND P24 ANTIGEN HIV-1 LEVEL AFTERHYPERBARIC OXYGEN TREATMENTOFHIV-1 INFECTED ON PERIPHERAL BLOOD MONONUCLEAR CELLS (PBMCS). Afr J Infect Dis 2018; 12:1-6. [PMID: 29619425 PMCID: PMC5876773 DOI: 10.21010/ajid.12v1s.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 08/24/2017] [Accepted: 08/25/2017] [Indexed: 12/04/2022] Open
Abstract
Background: Heme oxygenase-1 (HO-1) is a protein secreted by immune cells as a part of immune response mechanism.HO-1 can be induced by variety agents that causingoxidative stress, such as exposure to 100% oxygenat2,4 ATA pressure.It plays a vital role in maintaining cellular homeostasis.This study was conducted to identify the effect of hyperbaric oxygen exposure in cultured ofPBMCthat infected by HIV-1. Material and Methods: Primary culture of PBMCs were isolated from 16 healthy volunteers and HIV-1 infected MT4 cell line by co-culture. The PBMCs were aliquoted into two wells as control group and treatment group. The 16 samples of HIV-1 infected PBMCwere exposed to oxygen at 2,4 ATA in animal hyperbaric chamber forthree times in 30 minutes periods with 5 minutes spacing period, that called 1 session. The Treatment done on 5 sessions within 5 days. 16 samples of HIV-1 infected PMBCs that have no hyperbaric treatment became control group.The supernatant were measured the HO-1 production by ELISA andmRNA expression of HO-1 by real time PCR and the number ofantigen p24 HIV-1by ELISA. Results: The result showed that there was no increasing of HO-1 at both mRNA level and protein level, there was a decreasing number of antigen p24 HIV-1 at the treatment group. In addition, hyperbaric exposure could not increase the expression of HO-1, more over the viral replication might be reduced by other mechanism. Conclusions: Hyperbaric oxygen could increases cellular adaptive response of PBMCs infected HIV-1 through increased expression of proteins that can inhibit HIV viralreplication.
Collapse
Affiliation(s)
- Retno Budiarti
- Department of Microbiology, Faculty of Medicine, Hang Tuah University, Surabaya, Indonesia
| | - Kuntaman
- Department of Microbiology, Faculty of Medicine, Airlangga University, Surabaya, Indonesia
| | - Nasronudin
- Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya, Indonesia
| | - Suryokusumo
- Department of Hyperbaric, Faculty of Medicine, Pembangunan Nasional University, Jakarta, Indonesia
| | | |
Collapse
|
20
|
Gill AJ, Garza R, Ambegaokar SS, Gelman BB, Kolson DL. Heme oxygenase-1 promoter region (GT)n polymorphism associates with increased neuroimmune activation and risk for encephalitis in HIV infection. J Neuroinflammation 2018; 15:70. [PMID: 29510721 PMCID: PMC5838989 DOI: 10.1186/s12974-018-1102-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/21/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Heme oxygenase-1 (HO-1) is a critical cytoprotective enzyme that limits oxidative stress, inflammation, and cellular injury within the central nervous system (CNS) and other tissues. We previously demonstrated that HO-1 protein expression is decreased within the brains of HIV+ subjects and that this HO-1 reduction correlates with CNS immune activation and neurocognitive dysfunction. To define a potential CNS protective role for HO-1 against HIV, we analyzed a well-characterized HIV autopsy cohort for two common HO-1 promoter region polymorphisms that are implicated in regulating HO-1 promoter transcriptional activity, a (GT)n dinucleotide repeat polymorphism and a single nucleotide polymorphism (A(-413)T). Shorter HO-1 (GT)n repeats and the 'A' SNP allele associate with higher HO-1 promoter activity. METHODS Brain dorsolateral prefrontal cortex tissue samples from an autopsy cohort of HIV-, HIV+, and HIV encephalitis (HIVE) subjects (n = 554) were analyzed as follows: HO-1 (GT)n polymorphism allele lengths were determined by PCR and capillary electrophoresis, A(-413)T SNP alleles were determined by PCR with allele specific probes, and RNA expression of selected neuroimmune markers was analyzed by quantitative PCR. RESULTS HIV+ subjects with shorter HO-1 (GT)n alleles had a significantly lower risk of HIVE; however, shorter HO-1 (GT)n alleles did not correlate with CNS or peripheral viral loads. In HIV+ subjects without HIVE, shorter HO-1 (GT)n alleles associated significantly with lower expression of brain type I interferon response markers (MX1, ISG15, and IRF1) and T-lymphocyte activation markers (CD38 and GZMB). No significant correlations were found between the HO-1 (GT)n repeat length and brain expression of macrophage markers (CD163, CD68), endothelial markers (PECAM1, VWF), the T-lymphocyte marker CD8A, or the B-lymphocyte maker CD19. Finally, we found no significant associations between the A(-413)T SNP and HIVE diagnosis, HIV viral loads, or any neuroimmune markers. CONCLUSION Our data suggest that an individual's HO-1 promoter region (GT)n polymorphism allele repeat length exerts unique modifying risk effects on HIV-induced CNS neuroinflammation and associated neuropathogenesis. Shorter HO-1 (GT)n alleles increase HO-1 promoter activity, which could provide neuroprotection through decreased neuroimmune activation. Therapeutic strategies that induce HO-1 expression could decrease HIV-associated CNS neuroinflammation and decrease the risk for development of HIV neurological disease.
Collapse
Affiliation(s)
- Alexander J. Gill
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 280C Clinical Research Building, Philadelphia, PA 19104 USA
| | - Rolando Garza
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 280C Clinical Research Building, Philadelphia, PA 19104 USA
| | - Surendra S. Ambegaokar
- Department of Botany & Microbiology, Robbins Program in Neuroscience, Ohio Wesleyan University, Delaware, OH 43016 USA
| | - Benjamin B. Gelman
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Dennis L. Kolson
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 280C Clinical Research Building, Philadelphia, PA 19104 USA
| |
Collapse
|
21
|
Kovacsics CE, Gill AJ, Ambegaokar SS, Gelman BB, Kolson DL. Degradation of heme oxygenase-1 by the immunoproteasome in astrocytes: A potential interferon-γ-dependent mechanism contributing to HIV neuropathogenesis. Glia 2017; 65:1264-1277. [PMID: 28543773 DOI: 10.1002/glia.23160] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/28/2017] [Accepted: 04/07/2017] [Indexed: 12/16/2022]
Abstract
Induction of the detoxifying enzyme heme oxygenase-1 (HO-1) is a critical protective host response to cellular injury associated with inflammation and oxidative stress. We previously found that HO-1 protein expression is reduced in brains of HIV-infected individuals with HIV-associated neurocognitive disorders (HAND) and in HIV-infected macrophages, where this reduction associates with enhanced glutamate release and neurotoxicity. Because HIV-infected macrophages are a small component of the cellular content of the brain, the reduction of macrophage HO-1 expression likely accounts for a small portion of brain HO-1 loss in HIV infection. We therefore investigated the contribution of astrocytes, the major pool of brain HO-1. We identified immunoproteasome-mediated HO-1 degradation in astrocytes as a second possible mechanism of brain HO-1 loss in HIV infection. We demonstrate that prolonged exposure of human fetal astrocytes to interferon-gamma (IFNγ), an HIV-associated CNS immune activator, selectively reduces expression of HO-1 protein without a concomitant reduction in HO-1 RNA, increases expression of immunoproteasome subunits, and decreases expression of constitutive proteasome subunits, consistent with a shift towards increased immunoproteasome activity. In HIV-infected brain HO-1 protein reduction also associates with increased HO-1 RNA expression and increased immunoproteasome expression. Finally, we show that IFNγ treatment of astrocytic cells reduces HO-1 protein half-life in a proteasome-dependent manner. Our data thus suggest unique causal links among HIV infection, IFNγ-mediated immunoproteasome induction, and enhanced HO-1 degradation, which likely contribute to neurocognitive impairment in HAND. Such IFNγ-mediated HO-1 degradation should be further investigated for a role in neurodegeneration in inflammatory brain conditions. BRIEF SUMMARY Kovacsics et al. identify immunoproteasome degradation of heme oxygenase-1 (HO-1) in interferon gamma-stimulated astrocytes as a plausible mechanism for the observed loss of HO-1 protein expression in the brains of HIV-infected individuals, which likely contributes to the neurocognitive impairment in HIV-associated neurocognitive disorders.
Collapse
Affiliation(s)
- Colleen E Kovacsics
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104
| | - Alexander J Gill
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104
| | - Surendra S Ambegaokar
- Department of Botany & Microbiology, Robbins Program in Neuroscience, Ohio Wesleyan University, Delaware, Ohio, 43015
| | - Benjamin B Gelman
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, 77555
| | - Dennis L Kolson
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104
| |
Collapse
|
22
|
Rockwood N, Costa DL, Amaral EP, Du Bruyn E, Kubler A, Gil-Santana L, Fukutani KF, Scanga CA, Flynn JL, Jackson SH, Wilkinson KA, Bishai WR, Sher A, Wilkinson RJ, Andrade BB. Mycobacterium tuberculosis Induction of Heme Oxygenase-1 Expression Is Dependent on Oxidative Stress and Reflects Treatment Outcomes. Front Immunol 2017; 8:542. [PMID: 28553288 PMCID: PMC5427075 DOI: 10.3389/fimmu.2017.00542] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/24/2017] [Indexed: 12/17/2022] Open
Abstract
The antioxidant enzyme heme oxygenase-1 (HO-1) is implicated in the pathogenesis of tuberculosis (TB) and has been proposed as a biomarker of active disease. Nevertheless, the mechanisms by which Mycobacterium tuberculosis (Mtb) induces HO-1 as well as how its expression is affected by HIV-1 coinfection and successful antitubercular therapy (ATT) are poorly understood. We found that HO-1 expression is markedly increased in rabbits, mice, and non-human primates during experimental Mtb infection and gradually decreased during ATT. In addition, we examined circulating concentrations of HO-1 in a cohort of 130 HIV-1 coinfected and uninfected pulmonary TB patients undergoing ATT to investigate changes in expression of this biomarker in relation to HIV-1 status, radiological disease severity, and treatment outcome. We found that plasma levels of HO-1 were elevated in untreated HIV-1 coinfected TB patients and correlated positively with HIV-1 viral load and negatively with CD4+ T cell count. In both HIV-1 coinfected and Mtb monoinfected patients, HO-1 levels were substantially reduced during successful TB treatment but not in those who experienced treatment failure or subsequently relapsed. To further delineate the molecular mechanisms involved in induction of HO-1 by Mtb, we performed a series of in vitro experiments using mouse and human macrophages. We found that Mtb-induced HO-1 expression requires NADPH oxidase-dependent reactive oxygen species production induced by the early-secreted antigen ESAT-6, which in turn triggers nuclear translocation of the transcription factor NRF-2. These observations provide further insight into the utility of HO-1 as a biomarker of both disease and successful therapy in TB monoinfected and HIV-TB coinfected patients and reveal a previously undocumented pathway linking expression of the enzyme with oxidative stress.
Collapse
Affiliation(s)
- Neesha Rockwood
- Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Medicine, Imperial College, London, UK
| | - Diego L Costa
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Eduardo P Amaral
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Elsa Du Bruyn
- Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Andre Kubler
- Infectious Diseases and Immunity, Imperial College, London, UK.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leonardo Gil-Santana
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Fundação José Silveira, Salvador, Brazil.,Curso de Medicina, Faculdade de Tecnologia e Ciências, Salvador, Brazil
| | | | - Charles A Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sharon H Jackson
- Division of Intramural Research, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Katalin A Wilkinson
- Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,The Francis Crick Institute, London, UK
| | - William R Bishai
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert J Wilkinson
- Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Medicine, Imperial College, London, UK.,The Francis Crick Institute, London, UK
| | - Bruno B Andrade
- Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Fundação José Silveira, Salvador, Brazil.,Curso de Medicina, Faculdade de Tecnologia e Ciências, Salvador, Brazil.,Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.,Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| |
Collapse
|
23
|
Patton SM, Wang Q, Hulgan T, Connor JR, Jia P, Zhao Z, Letendre SL, Ellis RJ, Bush WS, Samuels DC, Franklin DR, Kaur H, Iudicello J, Grant I, Kallianpur AR. Cerebrospinal fluid (CSF) biomarkers of iron status are associated with CSF viral load, antiretroviral therapy, and demographic factors in HIV-infected adults. Fluids Barriers CNS 2017; 14:11. [PMID: 28427421 PMCID: PMC5399327 DOI: 10.1186/s12987-017-0058-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 04/01/2017] [Indexed: 02/06/2023] Open
Abstract
Background HIV-associated neurocognitive disorder (HAND) remains common, despite antiretroviral therapy (ART). HIV dysregulates iron metabolism, but cerebrospinal fluid (CSF) levels of iron and iron-transport proteins in HIV-infected (HIV+) persons are largely unknown. The objectives of this study were to characterize CSF iron-related biomarkers in HIV+ adults and explore their relationships to known predictors of HAND. Methods We quantified total iron, transferrin and heavy-chain (H)-ferritin by immunoassay in CSF sampled by lumbar puncture in 403 HIV+ participants in a multi-center, observational study and evaluated biomarker associations with demographic and HIV-related correlates of HAND [e.g., age, sex, self-reported race/ethnicity, ART, and detectable plasma virus and CSF viral load (VL)] by multivariable regression. In a subset (N = 110) with existing CSF: serum albumin (QAlb) measurements, QAlb and comorbidity severity were also included as covariates to account for variability in the blood–CSF-barrier. Results Among 403 individuals (median age 43 years, 19% women, 56% non-Whites, median nadir CD4+ T cell count 180 cells/µL, 46% with undetectable plasma virus), men had 25% higher CSF transferrin (median 18.1 vs. 14.5 µg/mL), and 71% higher H-ferritin (median 2.9 vs. 1.7 ng/mL) than women (both p-values ≤0.01). CSF iron was 41% higher in self-reported Hispanics and 27% higher in (non-Hispanic) Whites than in (non-Hispanic) Blacks (median 5.2 and 4.7 µg/dL in Hispanics and Whites, respectively, vs. 3.7 µg/dL in Blacks, both p ≤ 0.01); these findings persisted after adjustment for age, sex, and HIV-specific factors. Median H-ferritin was 25% higher (p < 0.05), and transferrin 14% higher (p = 0.06), in Whites than Blacks. Transferrin and H-ferritin were 33 and 50% higher, respectively, in older (age > 50 years) than in younger persons (age ≤ 35 years; both p < 0.01), but these findings lost statistical significance in subset analyses that adjusted for QAlb and comorbidity. After these additional adjustments, associations were observed for CSF iron and transferrin with race/ethnicity as well as CSF VL, for transferrin with sex and ART, and for H-ferritin with plasma virus detectability and significant comorbidity (all p < 0.05). Conclusions CSF iron biomarkers are associated with demographic factors, ART, and CSF VL in HIV+ adults. Future studies should investigate a role for CNS iron dysregulation, to which an altered blood-CSF barrier may contribute, in HAND. Electronic supplementary material The online version of this article (doi:10.1186/s12987-017-0058-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stephanie M Patton
- Department of Neurosurgery, Penn State Hershey Medical Center, 500 University Drive, Mailbox H110, Hershey, PA, 17033, USA.
| | - Quan Wang
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Todd Hulgan
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - James R Connor
- Department of Neurosurgery, Penn State Hershey Medical Center, 500 University Drive, Mailbox H110, Hershey, PA, 17033, USA
| | - Peilin Jia
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhongming Zhao
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Scott L Letendre
- Department of Medicine, University of California-San Diego, San Diego, CA, USA
| | - Ronald J Ellis
- Department of Neurology, University of California-San Diego, San Diego, CA, USA
| | - William S Bush
- Department of Epidemiology and Biostatistics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - David C Samuels
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Donald R Franklin
- Department of Medicine, University of California-San Diego, San Diego, CA, USA
| | - Harpreet Kaur
- Genomic Medicine Institute, Cleveland Clinic/Lerner Research Institute, Cleveland, OH, USA
| | - Jennifer Iudicello
- Department of Psychiatry, University of California-San Diego, San Diego, CA, USA
| | - Igor Grant
- Department of Psychiatry, University of California-San Diego, San Diego, CA, USA
| | - Asha R Kallianpur
- Genomic Medicine Institute, Cleveland Clinic/Lerner Research Institute, Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
24
|
Ivanov AV, Valuev-Elliston VT, Ivanova ON, Kochetkov SN, Starodubova ES, Bartosch B, Isaguliants MG. Oxidative Stress during HIV Infection: Mechanisms and Consequences. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8910396. [PMID: 27829986 PMCID: PMC5088339 DOI: 10.1155/2016/8910396] [Citation(s) in RCA: 234] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/18/2016] [Indexed: 12/15/2022]
Abstract
It is generally acknowledged that reactive oxygen species (ROS) play crucial roles in a variety of natural processes in cells. If increased to levels which cannot be neutralized by the defense mechanisms, they damage biological molecules, alter their functions, and also act as signaling molecules thus generating a spectrum of pathologies. In this review, we summarize current data on oxidative stress markers associated with human immunodeficiency virus type-1 (HIV-1) infection, analyze mechanisms by which this virus triggers massive ROS production, and describe the status of various defense mechanisms of the infected host cell. In addition, we have scrutinized scarce data on the effect of ROS on HIV-1 replication. Finally, we present current state of knowledge on the redox alterations as crucial factors of HIV-1 pathogenicity, such as neurotoxicity and dementia, exhaustion of CD4+/CD8+ T-cells, predisposition to lung infections, and certain side effects of the antiretroviral therapy, and compare them to the pathologies associated with the nitrosative stress.
Collapse
Affiliation(s)
- Alexander V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
| | - Vladimir T. Valuev-Elliston
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
| | - Olga N. Ivanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
| | - Sergey N. Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
| | - Elizaveta S. Starodubova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
- M. P. Chumakov Institute of Poliomyelitis and Viral Encephalitides, Moscow 142782, Russia
| | - Birke Bartosch
- Cancer Research Center Lyon, INSERM U1052 and CNRS 5286, Lyon University, 69003 Lyon, France
- DevWeCan Laboratories of Excellence Network (Labex), France
| | - Maria G. Isaguliants
- Riga Stradins University, Riga LV-1007, Latvia
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
- N. F. Gamaleya Research Center of Epidemiology and Microbiology, Moscow 123098, Russia
| |
Collapse
|
25
|
Tseng CK, Lin CK, Wu YH, Chen YH, Chen WC, Young KC, Lee JC. Human heme oxygenase 1 is a potential host cell factor against dengue virus replication. Sci Rep 2016; 6:32176. [PMID: 27553177 PMCID: PMC4995454 DOI: 10.1038/srep32176] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 08/03/2016] [Indexed: 12/17/2022] Open
Abstract
Dengue virus (DENV) infection and replication induces oxidative stress, which further contributes to the progression and pathogenesis of the DENV infection. Modulation of host antioxidant molecules may be a useful strategy for interfering with DENV replication. In this study, we showed that induction or exogenous overexpression of heme oxygenase-1 (HO-1), an antioxidant enzyme, effectively inhibited DENV replication in DENV-infected Huh-7 cells. This antiviral effect of HO-1 was attenuated by its inhibitor tin protoporphyrin (SnPP), suggesting that HO-1 was an important cellular factor against DENV replication. Biliverdin but not carbon monoxide and ferrous ions, which are products of the HO-1 on heme, mediated the HO-1-induced anti-DENV effect by non-competitively inhibiting DENV protease, with an inhibition constant (Ki) of 8.55 ± 0.38 μM. Moreover, HO-1 induction or its exogenous overexpression, rescued DENV-suppressed antiviral interferon response. Moreover, we showed that HO-1 induction by cobalt protoporphyrin (CoPP) and andrographolide, a natural product, as evidenced by a significant delay in the onset of disease and mortality, and virus load in the infected mice’s brains. These findings clearly revealed that a drug or therapy that induced the HO-1 signal pathway was a promising strategy for treating DENV infection.
Collapse
Affiliation(s)
- Chin-Kai Tseng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chun-Kuang Lin
- Doctoral Degree Program in Marine Biotechnology, College of Marine Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Yu-Hsuan Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen-Hsu Chen
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,School of Medicine, Graduate Institute of Medicine, Sepsis Research Center, Center for Dengue Fever Control and Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, HsinChu, Taiwan.,Center for Dengue Fever Control and Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wei-Chun Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kung-Chia Young
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jin-Ching Lee
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan.,Research Center for Natural Products and Drug Development, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
26
|
Saylor D, Dickens AM, Sacktor N, Haughey N, Slusher B, Pletnikov M, Mankowski JL, Brown A, Volsky DJ, McArthur JC. HIV-associated neurocognitive disorder--pathogenesis and prospects for treatment. Nat Rev Neurol 2016; 12:234-48. [PMID: 26965674 DOI: 10.1038/nrneurol.2016.27] [Citation(s) in RCA: 597] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the past two decades, several advancements have improved the care of HIV-infected individuals. Most importantly, the development and deployment of combination antiretroviral therapy (CART) has resulted in a dramatic decline in the rate of deaths from AIDS, so that people living with HIV today have nearly normal life expectancies if treated with CART. The term HIV-associated neurocognitive disorder (HAND) has been used to describe the spectrum of neurocognitive dysfunction associated with HIV infection. HIV can enter the CNS during early stages of infection, and persistent CNS HIV infection and inflammation probably contribute to the development of HAND. The brain can subsequently serve as a sanctuary for ongoing HIV replication, even when systemic viral suppression has been achieved. HAND can remain in patients treated with CART, and its effects on survival, quality of life and everyday functioning make it an important unresolved issue. In this Review, we describe the epidemiology of HAND, the evolving concepts of its neuropathogenesis, novel insights from animal models, and new approaches to treatment. We also discuss how inflammation is sustained in chronic HIV infection. Moreover, we suggest that adjunctive therapies--treatments targeting CNS inflammation and other metabolic processes, including glutamate homeostasis, lipid and energy metabolism--are needed to reverse or improve HAND-related neurological dysfunction.
Collapse
Affiliation(s)
- Deanna Saylor
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - Alex M Dickens
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - Ned Sacktor
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - Norman Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - Barbara Slusher
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - Mikhail Pletnikov
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - Joseph L Mankowski
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - Amanda Brown
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - David J Volsky
- The Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, New York 10029, USA
| | - Justin C McArthur
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| |
Collapse
|