1
|
Honeycutt JB, Wahl A, Files JK, League AF, Yadav-Samudrala BJ, Garcia JV, Fitting S. In situ analysis of neuronal injury and neuroinflammation during HIV-1 infection. Retrovirology 2024; 21:11. [PMID: 38945996 PMCID: PMC11215835 DOI: 10.1186/s12977-024-00644-z] [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: 01/25/2024] [Accepted: 05/10/2024] [Indexed: 07/02/2024] Open
Abstract
BACKGROUND Since the introduction of combination antiretroviral therapy (cART) the brain has become an important human immunodeficiency virus (HIV) reservoir due to the relatively low penetration of many drugs utilized in cART into the central nervous system (CNS). Given the inherent limitations of directly assessing acute HIV infection in the brains of people living with HIV (PLWH), animal models, such as humanized mouse models, offer the most effective means of studying the effects of different viral strains and their impact on HIV infection in the CNS. To evaluate CNS pathology during HIV-1 infection in the humanized bone marrow/liver/thymus (BLT) mouse model, a histological analysis was conducted on five CNS regions, including the frontal cortex, hippocampus, striatum, cerebellum, and spinal cord, to delineate the neuronal (MAP2ab, NeuN) and neuroinflammatory (GFAP, Iba-1) changes induced by two viral strains after 2 weeks and 8 weeks post-infection. RESULTS Findings reveal HIV-infected human cells in the brain of HIV-infected BLT mice, demonstrating HIV neuroinvasion. Further, both viral strains, HIV-1JR-CSF and HIV-1CH040, induced neuronal injury and astrogliosis across all CNS regions following HIV infection at both time points, as demonstrated by decreases in MAP2ab and increases in GFAP fluorescence signal, respectively. Importantly, infection with HIV-1JR-CSF had more prominent effects on neuronal health in specific CNS regions compared to HIV-1CH040 infection, with decreasing number of NeuN+ neurons, specifically in the frontal cortex. On the other hand, infection with HIV-1CH040 demonstrated more prominent effects on neuroinflammation, assessed by an increase in GFAP signal and/or an increase in number of Iba-1+ microglia, across CNS regions. CONCLUSION These findings demonstrate that CNS pathology is widespread during acute HIV infection. However, neuronal loss and the magnitude of neuroinflammation in the CNS is strain dependent indicating that strains of HIV cause differential CNS pathologies.
Collapse
Affiliation(s)
- Jenna B Honeycutt
- Division of Infectious Diseases, Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Angela Wahl
- Division of Infectious Diseases, Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, AL, 35294, USA
| | - Jacob K Files
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, AL, 35294, USA
| | - Alexis F League
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Barkha J Yadav-Samudrala
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - J Victor Garcia
- Division of Infectious Diseases, Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, AL, 35294, USA.
| | - Sylvia Fitting
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
2
|
Hamadani CM, Mahdi F, Merrell A, Flanders J, Cao R, Vashisth P, Dasanayake GS, Darlington DS, Singh G, Pride MC, Monroe WG, Taylor GR, Hunter AN, Roman G, Paris JJ, Tanner EEL. Ionic Liquid Coating-Driven Nanoparticle Delivery to the Brain: Applications for NeuroHIV. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305484. [PMID: 38572510 PMCID: PMC11186118 DOI: 10.1002/advs.202305484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/28/2023] [Indexed: 04/05/2024]
Abstract
Delivering cargo to the central nervous system (CNS) remains a pharmacological challenge. For infectious diseases such as HIV, the CNS acts as a latent reservoir that is inadequately managed by systemic antiretrovirals (ARTs). ARTs thus cannot eradicate HIV, and given CNS infection, patients experience neurological deficits collectively referred to as "neuroHIV". Herein, the development of bioinspired ionic liquid-coated nanoparticles (IL-NPs) for in situ hitchhiking on red blood cells (RBCs) is reported, which enables 48% brain delivery of intracarotid arterial- infused cargo. Moreover, IL choline trans-2-hexenoate (CA2HA 1:2) demonstrates preferential accumulation in parenchymal microglia over endothelial cells post-delivery. This study further demonstrates successful loading of abacavir (ABC), an ART that is challenging to encapsulate, into IL-NPs, and verifies retention of antiviral efficacy in vitro. IL-NPs are not cytotoxic to primary human peripheral blood mononuclear cells (PBMCs) and the CA2HA 1:2 coating itself confers notable anti-viremic capacity. In addition, in vitro cell culture assays show markedly increased uptake of IL-NPs into neural cells compared to bare PLGA nanoparticles. This work debuts bioinspired ionic liquids as promising nanoparticle coatings to assist CNS biodistribution and has the potential to revolutionize the delivery of cargos (i.e., drugs, viral vectors) through compartmental barriers such as the blood-brain-barrier (BBB).
Collapse
Affiliation(s)
- Christine M. Hamadani
- Department of Chemistry & BiochemistryThe University of MississippiUniversityMS38677USA
| | - Fakhri Mahdi
- Department of BioMolecular SciencesThe University of MississippiUniversityMS38677USA
| | - Anya Merrell
- Department of Chemistry & BiochemistryThe University of MississippiUniversityMS38677USA
| | - Jack Flanders
- Department of Chemistry & BiochemistryThe University of MississippiUniversityMS38677USA
| | - Ruofan Cao
- Department of BioMolecular SciencesThe University of MississippiUniversityMS38677USA
| | - Priyavrat Vashisth
- Department of Chemistry & BiochemistryThe University of MississippiUniversityMS38677USA
| | - Gaya S. Dasanayake
- Department of Chemistry & BiochemistryThe University of MississippiUniversityMS38677USA
| | - Donovan S. Darlington
- Department of Chemistry & BiochemistryThe University of MississippiUniversityMS38677USA
| | - Gagandeep Singh
- Department of Chemistry & BiochemistryThe University of MississippiUniversityMS38677USA
| | - Mercedes C. Pride
- Department of Chemistry & BiochemistryThe University of MississippiUniversityMS38677USA
| | - Wake G. Monroe
- Department of Chemistry & BiochemistryThe University of MississippiUniversityMS38677USA
| | - George R. Taylor
- Department of Chemistry & BiochemistryThe University of MississippiUniversityMS38677USA
| | - Alysha N. Hunter
- Department of Chemistry & BiochemistryThe University of MississippiUniversityMS38677USA
| | - Gregg Roman
- Department of BioMolecular SciencesThe University of MississippiUniversityMS38677USA
| | - Jason J. Paris
- Department of BioMolecular SciencesThe University of MississippiUniversityMS38677USA
| | - Eden E. L. Tanner
- Department of Chemistry & BiochemistryThe University of MississippiUniversityMS38677USA
| |
Collapse
|
3
|
Zhang Z, Scanlan A, Koneru R, Morrell CR, Reece MD, Edwards E, Roa S, Gavegnano C, Bimonte-Nelson H, Arbiser J, Tyor W. Honokiol hexafluoro confers reversal of neuropathological markers of HIV infection in a murine SCID model. Neurotherapeutics 2024; 21:e00329. [PMID: 38388224 PMCID: PMC10943487 DOI: 10.1016/j.neurot.2024.e00329] [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: 10/19/2023] [Revised: 01/27/2024] [Accepted: 01/28/2024] [Indexed: 02/24/2024] Open
Abstract
Cognitive impairment remains a persistent challenge in people living with HIV (PWLH) despite antiretroviral therapy (ART) due to ART's inability to eliminate brain HIV. HIV-induced cognitive dysfunction results from immune dysregulation, ongoing neuroinflammation, and the continuous virus presence, collectively contributing to cognitive deficits. Therefore, adjunctive therapies are needed to reduce cerebral HIV reservoirs, mitigate neuroinflammation, and impede cognitive dysfunction progression. Our study focused on Honokiol, known for its anti-inflammatory and neuroprotective properties, in an experimental mouse model simulating HIV-induced cognitive dysfunction. Using Honokiol Hexafluoro (HH), a synthetic analogue, we comprehensively evaluated its potential to ameliorate cognitive dysfunction and cerebral pathology in HIV-associated cognitive dysfunction. Our findings showed that HH treatment effectively reversed HIV-induced cognitive dysfunction, concurrently suppressing astrocyte activation, restoring neuronal dendritic arborization, and reducing microglial activation. Furthermore, HH remodeled the metabolic profile of HIV-infected human monocyte-derived macrophages, resulting in decreased activation and the promotion of a quiescent state in vitro.
Collapse
Affiliation(s)
- Zhan Zhang
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Aaron Scanlan
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA
| | - Rajeth Koneru
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA
| | - Chelsea Richardson Morrell
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA
| | - Monica D Reece
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Emily Edwards
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Sebastian Roa
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Christina Gavegnano
- Atlanta VA Medical Center, Decatur, GA, USA; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA; Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA; Center for the Study of Human Health, Emory College, Atlanta, GA, USA; Harvard Medical School, Center for Bioethics, Boston, MA, USA
| | | | - Jack Arbiser
- Department of Dermatology, Emory University School of Medicine, Atlanta, USA; Metroderm/United Derm Partners, Atlanta, GA, USA
| | - William Tyor
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA.
| |
Collapse
|
4
|
Abdalla AL, Guajardo-Contreras G, Mouland AJ. A Canadian Survey of Research on HIV-1 Latency-Where Are We Now and Where Are We Heading? Viruses 2024; 16:229. [PMID: 38400005 PMCID: PMC10891605 DOI: 10.3390/v16020229] [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: 01/02/2024] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Worldwide, almost 40 million people are currently living with HIV-1. The implementation of cART inhibits HIV-1 replication and reduces viremia but fails to eliminate HIV-1 from latently infected cells. These cells are considered viral reservoirs from which HIV-1 rebounds if cART is interrupted. Several efforts have been made to identify these cells and their niches. There has been little success in diminishing the pool of latently infected cells, underscoring the urgency to continue efforts to fully understand how HIV-1 establishes and maintains a latent state. Reactivating HIV-1 expression in these cells using latency-reversing agents (LRAs) has been successful, but only in vitro. This review aims to provide a broad view of HIV-1 latency, highlighting Canadian contributions toward these aims. We will summarize the research efforts conducted in Canadian labs to understand the establishment of latently infected cells and how this informs curative strategies, by reviewing how HIV latency is established, which cells are latently infected, what methodologies have been developed to characterize them, how new compounds are discovered and evaluated as potential LRAs, and what clinical trials aim to reverse latency in people living with HIV (PLWH).
Collapse
Affiliation(s)
- Ana Luiza Abdalla
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (A.L.A.); (G.G.-C.)
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Gabriel Guajardo-Contreras
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (A.L.A.); (G.G.-C.)
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Andrew J. Mouland
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (A.L.A.); (G.G.-C.)
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
5
|
Mohammadzadeh N, Chomont N, Estaquier J, Cohen EA, Power C. Is the Central Nervous System Reservoir a Hurdle for an HIV Cure? Viruses 2023; 15:2385. [PMID: 38140626 PMCID: PMC10747469 DOI: 10.3390/v15122385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/28/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
There is currently no cure for HIV infection although adherence to effective antiretroviral therapy (ART) suppresses replication of the virus in blood, increases CD4+ T-cell counts, reverses immunodeficiency, and increases life expectancy. Despite these substantial advances, ART is a lifelong treatment for people with HIV (PWH) and upon cessation or interruption, the virus quickly rebounds in plasma and anatomic sites, including the central nervous system (CNS), resulting in disease progression. With recent advances in quantifying viral burden, detection of genetically intact viral genomes, and isolation of replication-competent virus from brain tissues of PWH receiving ART, it has become apparent that the CNS viral reservoir (largely comprised of macrophage type cells) poses a substantial challenge for HIV cure strategies. Other obstacles impacting the curing of HIV include ageing populations, substance use, comorbidities, limited antiretroviral drug efficacy in CNS cells, and ART-associated neurotoxicity. Herein, we review recent findings, including studies of the proviral integration sites, reservoir decay rates, and new treatment/prevention strategies in the context of the CNS, together with highlighting the next steps for investigations of the CNS as a viral reservoir.
Collapse
Affiliation(s)
- Nazanin Mohammadzadeh
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| | - Nicolas Chomont
- Department of Immunopathology, Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada;
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3J7, Canada;
| | - Jerome Estaquier
- Department of Microbiology and Immunology, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada;
| | - Eric A. Cohen
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3J7, Canada;
- Institut de Recherches Cliniques de Montreal, Montreal, QC H2W 1R7, Canada
| | - Christopher Power
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| |
Collapse
|
6
|
Wang J, Li M, Li J, Deng R. Differences in drug resistance of HIV-1 genotypes in CSF and plasma and analysis of related factors. Virulence 2023; 14:2171632. [PMID: 36694270 PMCID: PMC9908293 DOI: 10.1080/21505594.2023.2171632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The emergence of HIV drug resistance seriously affects the quality of life of patients. However, there has been no extensive study of CSF resistance. The aim of this study is to evaluate common HIV-1 resistance in CSF and compare it with resistance in matched plasma, and analyse the influencing factors of cerebrospinal fluid drug resistance. The matched CSF and plasma samples of 62 HIV-1 patients were tested at one study site in China (Chongqing; 2019-2022). HIV genotyping and drug resistance was evaluated using the Stanford v8.7 algorithm. The diagnosis and treatment data and basic information were collected from the clinical case system, and the influencing factors of drug resistance mutations in CSF was obtained by variance analysis. CSF and matched plasma HIV-1 subtypes were confirmed in 62 patients, and the most frequent recombinant form was CRF07-BC (64.5%). Thirteen patients (21.0%) were detected with drug-resistant mutations, and the sites were consistent in both CSF and matched plasma. The drug-resistant ratios of untreated patients and treated patients were 5/51 (9.8%) and 8/11 (72.7%), respectively. The type with the highest mutation frequency was NNRTI, and no mutation was found in INSTI. Multivariate analysis indicated that ARV treatment was associated with CSF resistance (P < 0.001). The subtypes and drug resistance mutation sites are consistent in CSF and matched plasma samples of HIV-1 patients, and there is a correlation between ARV treatment and possible drug resistance, especially in CSF reservoirs. These findings highlight the concern about CSF drug resistance in HIV patients.
Collapse
Affiliation(s)
- Jie Wang
- Central lab, Chongqing Public Health Medical Center, Chongqing, China
| | - Mei Li
- Central lab, Chongqing Public Health Medical Center, Chongqing, China
| | - Jungang Li
- Central lab, Chongqing Public Health Medical Center, Chongqing, China
| | - Renni Deng
- Central lab, Chongqing Public Health Medical Center, Chongqing, China,CONTACT Renni Deng
| |
Collapse
|
7
|
Stachowicz-Kuśnierz A, Korchowiec B, Korchowiec J. Nucleoside Analog Reverse-Transcriptase Inhibitors in Membrane Environment: Molecular Dynamics Simulations. Molecules 2023; 28:6273. [PMID: 37687102 PMCID: PMC10488468 DOI: 10.3390/molecules28176273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/15/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
The behavior of four drugs from the family of nucleoside analog reverse-transcriptase inhibitors (zalcitabine, stavudine, didanosine, and apricitabine) in a membrane environment was traced using molecular dynamics simulations. The simulation models included bilayers and monolayers composed of POPC and POPG phospholipids. It was demonstrated that the drugs have a higher affinity towards POPG membranes than POPC membranes due to attractive long-range electrostatic interactions. The results obtained for monolayers were consistent with those obtained for bilayers. The drugs accumulated in the phospholipid polar headgroup region. Two adsorption modes were distinguished. They differed in the degree of penetration of the hydrophilic headgroup region. Hydrogen bonds between drug molecules and phospholipid heads were responsible for adsorption. It was shown that apricitabine penetrated the hydrophilic part of the POPC and POPG membranes more effectively than the other drugs. Van der Waals interactions between S atoms and lipids were responsible for this.
Collapse
Affiliation(s)
| | | | - Jacek Korchowiec
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland; (A.S.-K.); (B.K.)
| |
Collapse
|
8
|
Campbell GR, Rawat P, To RK, Spector SA. HIV-1 Tat Upregulates TREM1 Expression in Human Microglia. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:429-442. [PMID: 37326481 PMCID: PMC10352590 DOI: 10.4049/jimmunol.2300152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/30/2023] [Indexed: 06/17/2023]
Abstract
Because microglia are a reservoir for HIV and are resistant to the cytopathic effects of HIV infection, they are a roadblock for any HIV cure strategy. We have previously identified that triggering receptor expressed on myeloid cells 1 (TREM1) plays a key role in human macrophage resistance to HIV-mediated cytopathogenesis. In this article, we show that HIV-infected human microglia express increased levels of TREM1 and are resistant to HIV-induced apoptosis. Moreover, upon genetic inhibition of TREM1, HIV-infected microglia undergo cell death in the absence of increased viral or proinflammatory cytokine expression or the targeting of uninfected cells. We also show that the expression of TREM1 is mediated by HIV Tat through a TLR4, TICAM1, PG-endoperoxide synthase 2, PGE synthase, and PGE2-dependent manner. These findings highlight the potential of TREM1 as a therapeutic target to eradicate HIV-infected microglia without inducing a proinflammatory response.
Collapse
Affiliation(s)
- Grant R. Campbell
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD
| | - Pratima Rawat
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Rachel K. To
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Stephen A. Spector
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA
- Rady Children’s Hospital, San Diego, CA
| |
Collapse
|
9
|
Abbate MTA, Ramöller IK, Sabri AH, Paredes AJ, Hutton AJ, McKenna PE, Peng K, Hollett JA, McCarthy HO, Donnelly RF. Formulation of antiretroviral nanocrystals and development into a microneedle delivery system for potential treatment of HIV-associated neurocognitive disorder (HAND). Int J Pharm 2023; 640:123005. [PMID: 37142137 DOI: 10.1016/j.ijpharm.2023.123005] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023]
Abstract
HIV/AIDS remains a major global public health issue. While antiretroviral therapy is effective at reducing the viral load in the blood, up to 50% of those with HIV suffer from some degree of HIV-associated neurocognitive disorder, due to the presence of the blood-brain barrier restricting drugs from crossing into the central nervous system and treating the viral reservoir there. One way to circumvent this is the nose-to-brain pathway. This pathway can also be accessed via a facial intradermal injection. Certain parameters can increase delivery via this route, including using nanoparticles with a positive zeta potential and an effective diameter of 200 nm or less. Microneedle arrays offer a minimally invasive, pain-free alternative to traditional hypodermic injections. This study shows the formulation of nanocrystals of both rilpivirine (RPV) and cabotegravir, followed by incorporation into separate microneedle delivery systems for application to either side of the face. Following an in vivo study in rats, delivery to the brain was seen for both drugs. For RPV, a Cmax was seen at 21 days of 619.17 ± 73.32 ng/g, above that of recognised plasma IC90 levels, and potentially therapeutically relevant levels were maintained for 28 days. For CAB, a Cmax was seen at 28 days of 478.31 ± 320.86 ng/g, and while below recognised 4IC90 levels, does indicate that therapeutically relevant levels could be achieved by manipulating final microaaray patch size in humans.
Collapse
Affiliation(s)
- Marco T A Abbate
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL
| | - Inken K Ramöller
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL
| | - Akmal H Sabri
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL
| | | | - Aaron J Hutton
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL
| | - Peter E McKenna
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL
| | - Ke Peng
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL
| | - Jessica A Hollett
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL
| |
Collapse
|
10
|
Kumar A, Zhou L, Godse S, Sinha N, Ma D, Parmar K, Kumar S. Intranasal delivery of darunavir improves brain drug concentrations in mice for effective HIV treatment. Biochem Biophys Rep 2023; 33:101408. [DOI: 10.1016/j.bbrep.2022.101408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
|
11
|
Hamadani CM, Mahdi F, Merrell A, Flanders J, Cao R, Vashisth P, Pride MC, Hunter AN, Singh G, Roman G, Paris JJ, Tanner EEL. Ionic Liquid Coating-Driven Nanoparticle Delivery to the Brain: Applications for NeuroHIV. RESEARCH SQUARE 2023:rs.3.rs-2574352. [PMID: 36824802 PMCID: PMC9949257 DOI: 10.21203/rs.3.rs-2574352/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Delivering cargo to the central nervous system (CNS) remains a pharmacological challenge. For infectious diseases such as HIV, the CNS acts as a latent reservoir that is inadequately managed by systemic antiretrovirals (ARTs). ARTs thus cannot eradicate HIV, and given CNS infection, patients experience an array of neurological deficits that are collectively referred to as 'neuroHIV'. Herein we report the development of bioinspired ionic liquid-coated nanoparticles (IL-NPs) for in situ hitchhiking on red blood cells (RBCs), which enabled 48% delivery of intravenously infused cargo to the brain. Moreover, the ionic liquid (IL) choline trans-2-hexenoate (CA2HA 1:2) demonstrated preferential accumulation in parenchymal microglia over endothelial cells post-delivery. We further demonstrate the successful loading of abacavir (ABC), an ART that is challenging to encapsulate, into the IL-coated NPs and verify the retention of antiviral efficacy in vitro. IL-NPs were not cytotoxic to primary human peripheral blood mononuclear cells (PBMCs) and the CA2HA 1:2 coating conferred notable anti-viremic capacity on its own. In addition, in vitro cell culture assays showed markedly increased uptake of IL-coated nanoparticles into neuronal cells compared to bare nanoparticles. This work debuts bioinspired ionic liquids as promising nanoparticle coatings to assist CNS biodistribution and has the potential to revolutionize the delivery of cargos (i.e., drugs, viral vectors) through compartmental barriers such as the blood-brain-barrier (BBB), illustrated in the graphical abstract below.
Collapse
|
12
|
Overmars RJ, Krullaars Z, Mesplède T. Investigational drugs for HIV: trends, opportunities and key players. Expert Opin Investig Drugs 2023; 32:127-139. [PMID: 36751107 DOI: 10.1080/13543784.2023.2178415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Since the first antiretroviral drug was described, the field of HIV treatment and prevention has undergone two drug-based revolutions: the first one, enabled by the virtually concomitant discovery of non-nucleoside reverse transcriptase and protease inhibitors, was the inception of combined antiretroviral therapy. The second followed the creation of integrase strand-transfer inhibitors with improved safety, potency, and resistance profiles. Long-acting antiretroviral drugs, including broadly neutralizing antibodies, now offer the opportunity for a third transformational change in HIV management. AREAS COVERED Our review focused on HIV treatment and prevention with investigational drugs that offer the potential for infrequent dosing, including drugs not yet approved for clinical use. We also discussed approved drugs for which administration modalities or formulations are being optimized. We performed a literature search in published manuscripts, conference communications, and registered clinical trials. EXPERT OPINION While the field focuses on extending dosing intervals, we identify drug tissue penetration as an understudied opportunity to improve HIV care. We repeat that self-administration remains an essential milestone to reach the full potential of long-acting drugs. Treatments and prevention strategies based on broadly neutralizing antibodies require a deeper understanding of their antiretroviral properties.
Collapse
Affiliation(s)
- Ronald J Overmars
- Viroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Zoë Krullaars
- Viroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Thibault Mesplède
- Viroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
13
|
Mohammadzadeh N, Zhang N, Branton WG, Zghidi-Abouzid O, Cohen EA, Gelman BB, Estaquier J, Kong L, Power C. The HIV Restriction Factor Profile in the Brain Is Associated with the Clinical Status and Viral Quantities. Viruses 2023; 15:316. [PMID: 36851531 PMCID: PMC9962287 DOI: 10.3390/v15020316] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
HIV-encoded DNA, RNA and proteins persist in the brain despite effective antiretroviral therapy (ART), with undetectable plasma and cerebrospinal fluid viral RNA levels, often in association with neurocognitive impairments. Although the determinants of HIV persistence have garnered attention, the expression and regulation of antiretroviral host restriction factors (RFs) in the brain for HIV and SIV remain unknown. We investigated the transcriptomic profile of antiretroviral RF genes by RNA-sequencing with confirmation by qRT-PCR in the cerebral cortex of people who are uninfected (HIV[-]), those who are HIV-infected without pre-mortem brain disease (HIV[+]), those who are HIV-infected with neurocognitive disorders (HIV[+]/HAND) and those with neurocognitive disorders with encephalitis (HIV[+]/HIVE). We observed significant increases in RF expression in the brains of HIV[+]/HIVE in association with the brain viral load. Machine learning techniques identified MAN1B1 as a key gene that distinguished the HIV[+] group from the HIV[+] groups with HAND. Analyses of SIV-associated RFs in brains from SIV-infected Chinese rhesus macaques with different ART regimens revealed diminished RF expression among ART-exposed SIV-infected animals, although ART interruption resulted in an induced expression of several RF genes including OAS3, RNASEL, MX2 and MAN1B1. Thus, the brain displays a distinct expression profile of RFs that is associated with the neurological status as well as the brain viral burden. Moreover, ART interruption can influence the brain's RF profile, which might contribute to disease outcomes.
Collapse
Affiliation(s)
- Nazanin Mohammadzadeh
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Na Zhang
- Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - William G. Branton
- Department of Medicine (Neurology) University of Alberta, 6-11 Heritage Medical Research Centre, Edmonton, AB T6G 2R3, Canada
| | - Ouafa Zghidi-Abouzid
- Department of Microbiology and Immunology, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada
| | - Eric A. Cohen
- Institut de Recherches Cliniques de Montreal and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC J2S 2M2, Canada
| | - Benjamin B. Gelman
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jerome Estaquier
- Department of Microbiology and Immunology, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada
| | - Linglong Kong
- Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Christopher Power
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Department of Medicine (Neurology) University of Alberta, 6-11 Heritage Medical Research Centre, Edmonton, AB T6G 2R3, Canada
| |
Collapse
|
14
|
Scanlan A, Zhang Z, Koneru R, Reece M, Gavegnano C, Anderson AM, Tyor W. A Rationale and Approach to the Development of Specific Treatments for HIV Associated Neurocognitive Impairment. Microorganisms 2022; 10:2244. [PMID: 36422314 PMCID: PMC9699382 DOI: 10.3390/microorganisms10112244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 05/22/2024] Open
Abstract
Neurocognitive impairment (NCI) associated with HIV infection of the brain impacts a large proportion of people with HIV (PWH) regardless of antiretroviral therapy (ART). While the number of PWH and severe NCI has dropped considerably with the introduction of ART, the sole use of ART is not sufficient to prevent or arrest NCI in many PWH. As the HIV field continues to investigate cure strategies, adjunctive therapies are greatly needed. HIV imaging, cerebrospinal fluid, and pathological studies point to the presence of continual inflammation, and the presence of HIV RNA, DNA, and proteins in the brain despite ART. Clinical trials exploring potential adjunctive therapeutics for the treatment of HIV NCI over the last few decades have had limited success. Ideally, future research and development of novel compounds need to address both the HIV replication and neuroinflammation associated with HIV infection in the brain. Brain mononuclear phagocytes (MPs) are the primary instigators of inflammation and HIV protein expression; therefore, adjunctive treatments that act on MPs, such as immunomodulating agents, look promising. In this review, we will highlight recent developments of innovative therapies and discuss future approaches for HIV NCI treatment.
Collapse
Affiliation(s)
- Aaron Scanlan
- Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Zhan Zhang
- Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Rajeth Koneru
- Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
| | - Monica Reece
- Department of Pathology, Division of Experimental Pathology, Emory University, Atlanta, GA 30322, USA
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA 30322, USA
| | - Christina Gavegnano
- Department of Pathology, Division of Experimental Pathology, Emory University, Atlanta, GA 30322, USA
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA 30322, USA
| | - Albert M. Anderson
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - William Tyor
- Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
15
|
Sreeram S, Ye F, Garcia-Mesa Y, Nguyen K, El Sayed A, Leskov K, Karn J. The potential role of HIV-1 latency in promoting neuroinflammation and HIV-1-associated neurocognitive disorder. Trends Immunol 2022; 43:630-639. [PMID: 35840529 PMCID: PMC9339484 DOI: 10.1016/j.it.2022.06.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 11/29/2022]
Abstract
Despite potent suppression of HIV-1 viral replication in the central nervous system (CNS) by antiretroviral therapy (ART), between 15% and 60% of HIV-1-infected patients receiving ART exhibit neuroinflammation and symptoms of HIV-1-associated neurocognitive disorder (HAND) - a significant unmet challenge. We propose that the emergence of HIV-1 from latency in microglia underlies both neuroinflammation in the CNS and the progression of HAND. Recent molecular studies of cellular silencing mechanisms of HIV-1 in microglia show that HIV-1 latency can be reversed both by proinflammatory cytokines and by signals from damaged neurons, potentially creating intermittent cycles of HIV-1 reactivation and silencing in the brain. We posit that anti-inflammatory agents that also block HIV-1 reactivation, such as nuclear receptor agonists, might provide new putative therapeutic avenues for the treatment of HAND.
Collapse
Affiliation(s)
- Sheetal Sreeram
- Department of Molecular Biology and Microbiology. Case Western Reserve University, Cleveland, OH, USA
| | - Fengchun Ye
- Department of Molecular Biology and Microbiology. Case Western Reserve University, Cleveland, OH, USA
| | - Yoelvis Garcia-Mesa
- Department of Molecular Biology and Microbiology. Case Western Reserve University, Cleveland, OH, USA
| | - Kien Nguyen
- Department of Molecular Biology and Microbiology. Case Western Reserve University, Cleveland, OH, USA
| | - Ahmed El Sayed
- Department of Molecular Biology and Microbiology. Case Western Reserve University, Cleveland, OH, USA
| | - Konstantin Leskov
- Department of Molecular Biology and Microbiology. Case Western Reserve University, Cleveland, OH, USA
| | - Jonathan Karn
- Department of Molecular Biology and Microbiology. Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
16
|
HIV drug resistance in various body compartments. Curr Opin HIV AIDS 2022; 17:205-212. [PMID: 35762375 DOI: 10.1097/coh.0000000000000741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW HIV drug resistance testing using blood plasma or dried blood spots forms part of international guidelines. However, as the clinical utility of assessing drug resistance in other body compartments is less well established, we review this for blood cells and samples from other body compartments. RECENT EVIDENCE Although clinical benefit is not clear, drug resistance testing in blood cells is often performed when patients with suppressed plasma viral loads require a treatment substitution. In patients with HIV neurocognitive disease, cerebral spinal fluid (CSF) drug resistance is rarely discordant with plasma but has nevertheless been used to guide antiretroviral drug substitutions. Cases with HIV drug resistance in genital fluids have been documented but this does not appear to indicate transmission risk when blood plasma viral loads are suppressed. SUMMARY Drug-resistant variants, which may be selected in tissues under conditions of variable adherence and drug penetration, appear to disseminate quickly, and become detectable in blood. This may explain why drug resistance discordance between plasma and these compartments is rarely found. Partial compartmentalization of HIV populations is well established for the CSF and the genital tract but other than blood plasma, evidence is lacking to support drug resistance testing in body compartments.
Collapse
|
17
|
Li W, Pandya D, Pasternack N, Garcia-Montojo M, Henderson L, Kozak CA, Nath A. Retroviral Elements in Pathophysiology and as Therapeutic Targets for Amyotrophic Lateral Sclerosis. Neurotherapeutics 2022; 19:1085-1101. [PMID: 35415778 PMCID: PMC9587200 DOI: 10.1007/s13311-022-01233-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2022] [Indexed: 10/18/2022] Open
Abstract
The study of the role of retroviruses in amyotrophic lateral sclerosis (ALS) dates back to the 1960s shortly after transposable elements themselves were first discovered. It was quickly realized that in wild mice both horizontal and vertical transmissions of retroviral elements were key to the development of an ALS-like syndrome leading to the postulate that endogenous retroviruses (ERVs) contribute significantly to the pathogenicity of this disease. Subsequent studies identified retroviral reverse transcriptase activity in brains of individuals with ALS from Guam. However, except for a single study from the former Soviet Union, ALS could not be transmitted to rhesus macaques. The discovery of an ALS-like syndrome in human immunodeficiency virus (HIV) and human T cell leukemia virus infected individuals led to renewed interest in the field and reverse transcriptase activity was found in the blood and cerebrospinal fluid of individuals with sporadic ALS. However, exogenous retroviruses could not be found in individuals with ALS which further reinforced the possibility of involvement of a human ERV (HERV). The first demonstration of the involvement of a HERV was the discovery of the activation of human endogenous retrovirus-K subtype HML-2 in the brains of individuals with ALS. The envelope protein of HML-2 is neurotoxic and transgenic animals expressing the envelope protein develop an ALS-like syndrome. Activation of HML-2 occurs in the context of generalized transposable element activation and is not specific for ALS. Individuals with HIV-associated ALS show a remarkable response to antiretroviral therapy; however, antiretroviral trials in ALS down-regulate HML-2 without ameliorating the disease. This highlights the need for specific drugs to be developed against HML-2 as a novel therapeutic target for ALS. Other approaches might include antisense oligonucleotides, shRNA targeted against the envelope gene or antibodies that can target the extracellular envelope protein. Future clinical trials in ALS should consider combination therapies to control these ERVs.
Collapse
Affiliation(s)
- Wenxue Li
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Darshan Pandya
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Nicholas Pasternack
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Marta Garcia-Montojo
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Lisa Henderson
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Christine A Kozak
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
| |
Collapse
|
18
|
Millon EM, Lehrer PM, Shors TJ. Meditation and Aerobic Exercise Enhance Mental Health Outcomes and Pattern Separation Learning Without Changing Heart Rate Variability in Women with HIV. Appl Psychophysiol Biofeedback 2022; 47:27-42. [PMID: 35040014 PMCID: PMC8763305 DOI: 10.1007/s10484-021-09530-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2021] [Indexed: 01/01/2023]
Abstract
Mental and physical (MAP) training targets the brain and the body through a combination of focused-attention meditation and aerobic exercise. The following feasibility pilot study tested whether 6 weeks of MAP training improves mental health outcomes, while enhancing discrimination learning and heart rate variability (HRV) in a group of women living with human immunodeficiency virus (HIV) and other stress-related conditions. Participants were assigned to training (n = 18) or no-training control (n = 8) groups depending on their ability and willingness to participate, and if their schedule allowed. Training sessions were held once a week for 6 weeks with 30 min of meditation followed by 30 min of aerobic exercise. Before and after 6 weeks of training, participants completed the Behavioral Pattern Separation Task as a measure of discrimination learning, self-report questionnaires of ruminative and trauma-related thoughts, depression, anxiety, and perceived stress, and an assessment of HRV at rest. After training, participants reported fewer ruminative and trauma-related thoughts, fewer depressive and anxiety symptoms, and less perceived stress (p's < 0.05). The positive impact on ruminative thoughts and depressive symptoms persisted 6 months after training. They also demonstrated enhanced discrimination of similar patterns of information (p < 0.05). HRV did not change after training (p > 0.05). Combining mental and physical training is an effective program for enhancing mental health and aspects of cognition in women living with HIV, although not necessarily through variance in heart rate.
Collapse
Affiliation(s)
- Emma M Millon
- Behavioral and Systems Neuroscience, Department of Psychology, Rutgers University, Piscataway, New Jersey, USA.
| | - Paul M Lehrer
- Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Tracey J Shors
- Behavioral and Systems Neuroscience, Department of Psychology, Rutgers University, Piscataway, New Jersey, USA
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
19
|
Brew BJ. Is HIV Brain Disease Preventable? NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:9/2/e1145. [PMID: 35140143 PMCID: PMC8904082 DOI: 10.1212/nxi.0000000000001145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 11/24/2022]
Affiliation(s)
- Bruce J Brew
- From the Faculty of Medicine (B.J.B.), University of New South Wales, and University of Notre Dame Sydney; and Departments of Neurology and HIV Medicine (B.J.B.), St Vincent's Hospital, and Peter Duncan Neurosciences Unit, St Vincent's Centre for Applied Medical Research, Sydney, Australia.
| |
Collapse
|
20
|
Brookes A, Ji L, Bradshaw TD, Stocks M, Gray D, Butler J, Gershkovich P. Is Oral Lipid-Based Delivery for Drug Targeting to the Brain Feasible? Eur J Pharm Biopharm 2022; 172:112-122. [PMID: 35149190 DOI: 10.1016/j.ejpb.2022.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/21/2022] [Accepted: 02/05/2022] [Indexed: 12/12/2022]
Abstract
This review outlines the feasibility of oral lipid-based targeted delivery of drugs to the brain, including permeation of the central nervous system's (CNS) protective blood-brain barrier (BBB). The structure of the BBB and disruption caused by varying disease states highlights the need for disease-specific approaches to alter permeation. Disruption during disease state, and the effects of certain molecules on the barrier, demonstrate the possibility of exploiting such BBB disruption for drug delivery. Many administration methods can be used to target the brain, but oral administration is considered ideal for chronic, long-term illnesses. Several lipids that have been shown to facilitate drug delivery into the brain after systemic administration, but could also be delivered orally are discussed, including oleic acid, triolein, alkylglycerol, and conjugates of linoleic and myristic acids. Current data reveal the potential for the use of such lipids as part of oral formulations for delivery to the brain by reaching sufficient plasma levels after administration to increase the permeability of the BBB. However, gaps in the literature remain regarding the concentrations and form of most lipids required to produce the desired effects. The use of lipids via oral delivery for brain targeting has not been investigated thoroughly enough to determine with certainty if similar permeability-enhancing effects would be observed as for parenteral administration. In conclusion, further research to fill research gaps is needed, but the limited evidence suggests that oral lipid-based drug delivery for brain targeting is potentially feasible.
Collapse
Affiliation(s)
- Alice Brookes
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Liuhang Ji
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Tracey D Bradshaw
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Michael Stocks
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - David Gray
- Division of Food, Nutrition and Dietetics, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire, UK, LE12 5RD
| | - James Butler
- GlaxoSmithKline Research and Development, Park Road, Ware, Hertfordshire, UK, SG12 0DP
| | - Pavel Gershkovich
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD.
| |
Collapse
|
21
|
Gumbs SBH, Kübler R, Gharu L, Schipper PJ, Borst AL, Snijders GJLJ, Ormel PR, van Berlekom AB, Wensing AMJ, de Witte LD, Nijhuis M. Human microglial models to study HIV infection and neuropathogenesis: a literature overview and comparative analyses. J Neurovirol 2022; 28:64-91. [PMID: 35138593 PMCID: PMC9076745 DOI: 10.1007/s13365-021-01049-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/03/2021] [Accepted: 12/18/2021] [Indexed: 02/08/2023]
Abstract
HIV persistence in the CNS despite antiretroviral therapy may cause neurological disorders and poses a critical challenge for HIV cure. Understanding the pathobiology of HIV-infected microglia, the main viral CNS reservoir, is imperative. Here, we provide a comprehensive comparison of human microglial culture models: cultured primary microglia (pMG), microglial cell lines, monocyte-derived microglia (MDMi), stem cell-derived microglia (iPSC-MG), and microglia grown in 3D cerebral organoids (oMG) as potential model systems to advance HIV research on microglia. Functional characterization revealed phagocytic capabilities and responsiveness to LPS across all models. Microglial transcriptome profiles of uncultured pMG showed the highest similarity to cultured pMG and oMG, followed by iPSC-MG and then MDMi. Direct comparison of HIV infection showed a striking difference, with high levels of viral replication in cultured pMG and MDMi and relatively low levels in oMG resembling HIV infection observed in post-mortem biopsies, while the SV40 and HMC3 cell lines did not support HIV infection. Altogether, based on transcriptional similarities to uncultured pMG and susceptibility to HIV infection, MDMi may serve as a first screening tool, whereas oMG, cultured pMG, and iPSC-MG provide more representative microglial culture models for HIV research. The use of current human microglial cell lines (SV40, HMC3) is not recommended.
Collapse
Affiliation(s)
- Stephanie B H Gumbs
- Translational Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Raphael Kübler
- Translational Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Psychiatry, Icahn School of Medicine, New York, NY, USA
| | - Lavina Gharu
- Translational Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pauline J Schipper
- Translational Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anne L Borst
- Translational Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gijsje J L J Snijders
- Department of Psychiatry, Icahn School of Medicine, New York, NY, USA
- Department of Psychiatry, University Medical Center Utrecht, Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Paul R Ormel
- Department of Psychiatry, University Medical Center Utrecht, Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Amber Berdenis van Berlekom
- Department of Psychiatry, University Medical Center Utrecht, Brain Center, Utrecht University, Utrecht, The Netherlands
- Department of Translational Neuroscience, University Medical Center Utrecht, Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Annemarie M J Wensing
- Translational Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lot D de Witte
- Department of Psychiatry, Icahn School of Medicine, New York, NY, USA
- Department of Psychiatry, University Medical Center Utrecht, Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Monique Nijhuis
- Translational Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
22
|
Adeola AO, Forbes PBC. Antiretroviral Drugs in African Surface Waters: Prevalence, Analysis, and Potential Remediation. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2022; 41:247-262. [PMID: 34033688 DOI: 10.1002/etc.5127] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/24/2021] [Accepted: 05/18/2021] [Indexed: 06/12/2023]
Abstract
The sources, ecotoxicological impact, and potential remediation strategies of antiretroviral drugs (ARVDs) as emerging contaminants in surface waters are reviewed based on recent literature. The occurrence of ARVDs in water bodies raises concern because many communities in Africa depend on rivers for water resources. Southern Africa is a potential hotspot regarding ARVD contamination due to relatively high therapeutic application and detection thereof in water bodies. Efavirenz and nevirapine are the most persistent in effluents and are prevalent in surface water based on environmental concentrations. Whereas the highest concentration of efavirenz reported in Kenya was 12.4 µg L-1 , concentrations as high as 119 and 140 µg L-1 have been reported in Zambia and South Africa, respectively. Concentrations of ARVDs ranging from 670 to 34 000 ng L-1 (influents) and 540 to 34 000 ng L-1 (effluents) were determined in wastewater treatment plants in South Africa, compared with Europe, where reported concentrations range from less than limit of detection (LOD) to 32 ng L-1 (influents) and less than LOD to 22 ng L-1 (effluents). The present African-based review suggests the need for comprehensive toxicological and risk assessment of these emerging pollutants in Africa, with the intent of averting environmental hazards and the development of sustainable remediation strategies. Environ Toxicol Chem 2022;41:247-262. © 2021 SETAC.
Collapse
Affiliation(s)
- Adedapo O Adeola
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, University of Pretoria, Hatfield, Pretoria, South Africa
| | - Patricia B C Forbes
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, University of Pretoria, Hatfield, Pretoria, South Africa
| |
Collapse
|
23
|
Kumar G, Cottalorda-Dufayard J, Garraffo R, De Salvador-Guillouët F, Cua E, Roger PM. Raltegravir Inclusion Decreases CD4 T-Cells Intra-Cellular Viral Load and Increases CD4 and CD28 Positive T-Cells in Selected HIV Patients. Cells 2022; 11:cells11020208. [PMID: 35053324 PMCID: PMC8773801 DOI: 10.3390/cells11020208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/27/2021] [Accepted: 12/30/2021] [Indexed: 02/04/2023] Open
Abstract
Raltegravir (RLT) prevents the integration of HIV DNA in the nucleus, but published studies remain controversial, suggesting that it does not decrease proviral DNA. However, there are only a few studies focused on virus-targeted cells. We aimed our study on the impact of RLT inclusion on total intra-cellular viral DNA (TID) in cellular subsets and immune effects in patients with newly acquired undetectable plasmatic viral load (UVL). Six patients having UVL using an antiretroviral combination for 6 months and CD4 T-cells > 350/mL and <500/mL were selected to receive RLT for 3 months from M0 to M3. Patients had 7 sequential viro-immunological determinations from M-1 to M5. Immune phenotypes were determined by flow cytometry and TID quantification was performed using PCR assay on purified cells. TID (median values) at the initiation of RLT in CD4 T-cells was 117 copies/millions of cells, decreased to 27.5 on M3, and remained thereafter permanently under the cut-off (<10 copies/millions of cells) in 4 out of 6 patients. This was associated with an increase of CD4 and CD4 + CD28+ T-cells and a decrease of HLA-DR expression and apoptosis of CD4 T-cells. RLT inclusion led to decreases in the viral load along with positive immune reconstitution, mainly for CD4 T-cells in HIV patients.
Collapse
Affiliation(s)
- Gaurav Kumar
- Unité 576, Centre Hospitalier Universitaire de Nice, Institut National de la Sante et de la Recherche Medicale, Universite de Nice-Sophia-Antipolis, 06200 Nice, France;
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Correspondence: ; Tel.: +1-(405)-271-2907; Fax: +1-(405)-271-4110
| | - Jacqueline Cottalorda-Dufayard
- Virologie, Hopital l’Archet 2, Centre Hospitalier Universitaire de Nice, Universite de Nice-Sophia-Antipolis, 06200 Nice, France;
| | - Rodolphe Garraffo
- Pharmacologie, Hopital Pasteur, Centre Hospitalier Universitaire de Nice, Universite de Nice-Sophia-Antipolis, 06200 Nice, France;
| | - Francine De Salvador-Guillouët
- Infectiologie, Hopital l’Archet 1, Centre Hospitalier Universitaire de Nice, Universite de Nice-Sophia-Antipolis, 06200 Nice, France; (F.D.S.-G.); (E.C.)
| | - Eric Cua
- Infectiologie, Hopital l’Archet 1, Centre Hospitalier Universitaire de Nice, Universite de Nice-Sophia-Antipolis, 06200 Nice, France; (F.D.S.-G.); (E.C.)
| | - Pierre-Marie Roger
- Unité 576, Centre Hospitalier Universitaire de Nice, Institut National de la Sante et de la Recherche Medicale, Universite de Nice-Sophia-Antipolis, 06200 Nice, France;
- Infectiologie, Hopital l’Archet 1, Centre Hospitalier Universitaire de Nice, Universite de Nice-Sophia-Antipolis, 06200 Nice, France; (F.D.S.-G.); (E.C.)
- Service Des Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire de Pointe-à-Pitre, 97159 Pointe-à-Pitre, France
| |
Collapse
|
24
|
Lentiviral Infections Persist in Brain despite Effective Antiretroviral Therapy and Neuroimmune Activation. mBio 2021; 12:e0278421. [PMID: 34903055 PMCID: PMC8669467 DOI: 10.1128/mbio.02784-21] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
HIV infection persists in different tissue reservoirs among people with HIV (PWH) despite effective antiretroviral therapy (ART). In the brain, lentiviruses replicate principally in microglia and trafficking macrophages. The impact of ART on this viral reservoir is unknown. We investigated the activity of contemporary ART in various models of lentivirus brain infection. HIV-1 RNA and total and integrated DNA were detected in cerebral cortex from all PWH (n = 15), regardless of ART duration or concurrent plasma viral quantity and, interestingly, integrated proviral DNA levels in brain were significantly higher in the aviremic ART-treated group (P < 0.005). Most ART drugs tested (dolutegravir, ritonavir, raltegravir, and emtricitabine) displayed significantly lower 50% effective concentration (EC50) values in lymphocytes than in microglia, except tenofovir, which showed 1.5-fold greater activity in microglia (P < 0.05). In SIV-infected Chinese rhesus macaques, despite receiving suppressive (n = 7) or interrupted (n = 8) ART, brain tissues had similar SIV-encoded RNA and total and integrated DNA levels compared to brains from infected animals without ART (n = 3). SIV and HIV-1 capsid antigens were immunodetected in brain, principally in microglia/macrophages, regardless of ART duration and outcome. Antiviral immune responses were comparable in the brains of ART-treated and untreated HIV- and SIV-infected hosts. Both HIV-1 and SIV persist in brain tissues despite contemporary ART, with undetectable virus in blood. ART interruption exerted minimal effect on the SIV brain reservoir and did not alter the neuroimmune response profile. These studies underscore the importance of augmenting ART potency in different tissue compartments. IMPORTANCE Antiretroviral therapy (ART) suppresses HIV-1 in plasma and CSF to undetectable levels. However, the impact of contemporary ART on HIV-1 brain reservoirs remains uncertain. An active viral reservoir in the brain during ART could lead to rebound systemic infection after cessation of therapy, development of drug resistance mutations, and neurological disease. ART's impact, including its interruption, on brain proviral DNA remains unclear. The present studies show that in different experimental platforms, contemporary ART did not suppress viral burden in the brain, regardless of ART component regimen, the duration of therapy, and its interruption. Thus, new strategies for effective HIV-1 suppression in the brain are imperative to achieve sustained HIV suppression.
Collapse
|
25
|
Hokello J, Sharma AL, Tyagi P, Bhushan A, Tyagi M. Human Immunodeficiency Virus Type-1 (HIV-1) Transcriptional Regulation, Latency and Therapy in the Central Nervous System. Vaccines (Basel) 2021; 9:vaccines9111272. [PMID: 34835203 PMCID: PMC8618135 DOI: 10.3390/vaccines9111272] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
The central nervous system (CNS) is highly compartmentalized and serves as a specific site of human immunodeficiency virus (HIV) infection. Therefore, an understanding of the cellular populations that are infected by HIV or that harbor latent HIV proviruses is imperative in the attempts to address cure strategies, taking into account that HIV infection and latency in the CNS may differ considerably from those in the periphery. HIV replication in the CNS is reported to persist despite prolonged combination antiretroviral therapy due to the inability of the current antiretroviral drugs to penetrate and cross the blood–brain barrier. Consequently, as a result of sustained HIV replication in the CNS even in the face of combination antiretroviral therapy, there is a high incidence of HIV-associated neurocognitive disorders (HAND). This article, therefore, provides a comprehensive review of HIV transcriptional regulation, latency, and therapy in the CNS.
Collapse
Affiliation(s)
- Joseph Hokello
- Department of Biology, Faculty of Science and Education, Busitema University, Tororo P.O. Box 236, Uganda;
| | | | - Priya Tyagi
- Cherry Hill East High School, 1750 Kresson Rd, Cherry Hill, NJ 08003, USA;
| | - Alok Bhushan
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Mudit Tyagi
- Center for Translational Medicine, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA;
- Correspondence:
| |
Collapse
|
26
|
Nickoloff-Bybel EA, Festa L, Meucci O, Gaskill PJ. Co-receptor signaling in the pathogenesis of neuroHIV. Retrovirology 2021; 18:24. [PMID: 34429135 PMCID: PMC8385912 DOI: 10.1186/s12977-021-00569-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
The HIV co-receptors, CCR5 and CXCR4, are necessary for HIV entry into target cells, interacting with the HIV envelope protein, gp120, to initiate several signaling cascades thought to be important to the entry process. Co-receptor signaling may also promote the development of neuroHIV by contributing to both persistent neuroinflammation and indirect neurotoxicity. But despite the critical importance of CXCR4 and CCR5 signaling to HIV pathogenesis, there is only one therapeutic (the CCR5 inhibitor Maraviroc) that targets these receptors. Moreover, our understanding of co-receptor signaling in the specific context of neuroHIV is relatively poor. Research into co-receptor signaling has largely stalled in the past decade, possibly owing to the complexity of the signaling cascades and functions mediated by these receptors. Examining the many signaling pathways triggered by co-receptor activation has been challenging due to the lack of specific molecular tools targeting many of the proteins involved in these pathways and the wide array of model systems used across these experiments. Studies examining the impact of co-receptor signaling on HIV neuropathogenesis often show activation of multiple overlapping pathways by similar stimuli, leading to contradictory data on the effects of co-receptor activation. To address this, we will broadly review HIV infection and neuropathogenesis, examine different co-receptor mediated signaling pathways and functions, then discuss the HIV mediated signaling and the differences between activation induced by HIV and cognate ligands. We will assess the specific effects of co-receptor activation on neuropathogenesis, focusing on neuroinflammation. We will also explore how the use of substances of abuse, which are highly prevalent in people living with HIV, can exacerbate the neuropathogenic effects of co-receptor signaling. Finally, we will discuss the current state of therapeutics targeting co-receptors, highlighting challenges the field has faced and areas in which research into co-receptor signaling would yield the most therapeutic benefit in the context of HIV infection. This discussion will provide a comprehensive overview of what is known and what remains to be explored in regard to co-receptor signaling and HIV infection, and will emphasize the potential value of HIV co-receptors as a target for future therapeutic development. ![]()
Collapse
Affiliation(s)
- E A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - L Festa
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 S. 40th Street, Philadelphia, PA, 19104, USA
| | - O Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.,Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - P J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
27
|
HIV in the Brain: Identifying Viral Reservoirs and Addressing the Challenges of an HIV Cure. Vaccines (Basel) 2021; 9:vaccines9080867. [PMID: 34451992 PMCID: PMC8402376 DOI: 10.3390/vaccines9080867] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 12/13/2022] Open
Abstract
Advances in antiretroviral therapy have prolonged the life of people living with HIV and diminished the level of virus in these individuals. Yet, HIV quickly rebounds after disruption and/or cessation of treatment due to significant cellular and anatomical reservoirs for HIV, which underscores the challenge for HIV cure strategies. The central nervous system (CNS), in particular, is seeded with HIV within 1–2 weeks of infection and is a reservoir for HIV. In this review, we address the paradigm of HIV reservoirs in the CNS and the relevant cell types, including astrocytes and microglia, that have been shown to harbor viral infection even with antiretroviral treatment. In particular, we focus on developmental aspects of astrocytes and microglia that lead to their susceptibility to infection, and how HIV infection propagates among these cells. We also address challenges of measuring the HIV latent reservoir, advances in viral detection assays, and how curative strategies have evolved in regard to the CNS reservoir. Current curative strategies still require optimization to reduce or eliminate the HIV CNS reservoir, and may also contribute to levels of neuroinflammation that lead to cognitive decline. With this in mind, the latent HIV reservoir in the brain should remain a prominent focus when assessing treatment options and overall viral burden in the clinic, especially in the context of HIV-associated neurocognitive disorders (HAND).
Collapse
|
28
|
Borrajo López A, Penedo MA, Rivera-Baltanas T, Pérez-Rodríguez D, Alonso-Crespo D, Fernández-Pereira C, Olivares JM, Agís-Balboa RC. Microglia: The Real Foe in HIV-1-Associated Neurocognitive Disorders? Biomedicines 2021; 9:925. [PMID: 34440127 PMCID: PMC8389599 DOI: 10.3390/biomedicines9080925] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/19/2021] [Accepted: 07/26/2021] [Indexed: 12/21/2022] Open
Abstract
The current use of combined antiretroviral therapy (cART) is leading to a significant decrease in deaths and comorbidities associated with human immunodeficiency virus type 1 (HIV-1) infection. Nonetheless, none of these therapies can extinguish the virus from the long-lived cellular reservoir, including microglia, thereby representing an important obstacle to curing HIV. Microglia are the foremost cells infected by HIV-1 in the central nervous system (CNS) and are believed to be involved in the development of HIV-1-associated neurocognitive disorder (HAND). At present, the pathological mechanisms contributing to HAND remain unclear, but evidence suggests that removing these infected cells from the brain, as well as obtaining a better understanding of the specific molecular mechanisms of HIV-1 latency in these cells, should help in the design of new strategies to prevent HAND and achieve a cure for these diseases. The goal of this review was to study the current state of knowledge of the neuropathology and research models of HAND containing virus susceptible target cells (microglial cells) and potential pharmacological treatment approaches under investigation.
Collapse
Affiliation(s)
- Ana Borrajo López
- Department of Microbiology and Parasitology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Roma, Italy
| | - Maria Aránzazu Penedo
- Translational Neuroscience Group-CIBERSAM, Galicia Sur Health Research Institute (IIS Galicia Sur), Área Sanitaria de Vigo-Hospital Álvaro Cunqueiro, SERGAS-UVIGO, 36213 Vigo, Spain; (M.A.P.); (T.R.-B.); (D.P.-R.); (C.F.-P.); (J.M.O.)
- Neuro Epigenetics Laboratory, University Hospital Complex of Vigo, SERGAS-UVIGO, 36213 Virgo, Spain
| | - Tania Rivera-Baltanas
- Translational Neuroscience Group-CIBERSAM, Galicia Sur Health Research Institute (IIS Galicia Sur), Área Sanitaria de Vigo-Hospital Álvaro Cunqueiro, SERGAS-UVIGO, 36213 Vigo, Spain; (M.A.P.); (T.R.-B.); (D.P.-R.); (C.F.-P.); (J.M.O.)
| | - Daniel Pérez-Rodríguez
- Translational Neuroscience Group-CIBERSAM, Galicia Sur Health Research Institute (IIS Galicia Sur), Área Sanitaria de Vigo-Hospital Álvaro Cunqueiro, SERGAS-UVIGO, 36213 Vigo, Spain; (M.A.P.); (T.R.-B.); (D.P.-R.); (C.F.-P.); (J.M.O.)
- Neuro Epigenetics Laboratory, University Hospital Complex of Vigo, SERGAS-UVIGO, 36213 Virgo, Spain
| | - David Alonso-Crespo
- Nursing Team-Intensive Care Unit, Área Sanitaria de Vigo, Estrada de Clara Campoamor 341, SERGAS-UVigo, 36312 Virgo, Spain;
| | - Carlos Fernández-Pereira
- Translational Neuroscience Group-CIBERSAM, Galicia Sur Health Research Institute (IIS Galicia Sur), Área Sanitaria de Vigo-Hospital Álvaro Cunqueiro, SERGAS-UVIGO, 36213 Vigo, Spain; (M.A.P.); (T.R.-B.); (D.P.-R.); (C.F.-P.); (J.M.O.)
- Neuro Epigenetics Laboratory, University Hospital Complex of Vigo, SERGAS-UVIGO, 36213 Virgo, Spain
| | - José Manuel Olivares
- Translational Neuroscience Group-CIBERSAM, Galicia Sur Health Research Institute (IIS Galicia Sur), Área Sanitaria de Vigo-Hospital Álvaro Cunqueiro, SERGAS-UVIGO, 36213 Vigo, Spain; (M.A.P.); (T.R.-B.); (D.P.-R.); (C.F.-P.); (J.M.O.)
- Department of Psychiatry, Área Sanitaria de Vigo, Estrada de Clara Campoamor 341, SERGAS-UVigo, 36312 Vigo, Spain
| | - Roberto Carlos Agís-Balboa
- Translational Neuroscience Group-CIBERSAM, Galicia Sur Health Research Institute (IIS Galicia Sur), Área Sanitaria de Vigo-Hospital Álvaro Cunqueiro, SERGAS-UVIGO, 36213 Vigo, Spain; (M.A.P.); (T.R.-B.); (D.P.-R.); (C.F.-P.); (J.M.O.)
| |
Collapse
|
29
|
Li M, Brokaw A, Furuta AM, Coler B, Obregon-Perko V, Chahroudi A, Wang HY, Permar SR, Hotchkiss CE, Golos TG, Rajagopal L, Adams Waldorf KM. Non-human Primate Models to Investigate Mechanisms of Infection-Associated Fetal and Pediatric Injury, Teratogenesis and Stillbirth. Front Genet 2021; 12:680342. [PMID: 34290739 PMCID: PMC8287178 DOI: 10.3389/fgene.2021.680342] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/25/2021] [Indexed: 12/25/2022] Open
Abstract
A wide array of pathogens has the potential to injure the fetus and induce teratogenesis, the process by which mutations in fetal somatic cells lead to congenital malformations. Rubella virus was the first infectious disease to be linked to congenital malformations due to an infection in pregnancy, which can include congenital cataracts, microcephaly, hearing impairment and congenital heart disease. Currently, human cytomegalovirus (HCMV) is the leading infectious cause of congenital malformations globally, affecting 1 in every 200 infants. However, our knowledge of teratogenic viruses and pathogens is far from complete. New emerging infectious diseases may induce teratogenesis, similar to Zika virus (ZIKV) that caused a global pandemic in 2016-2017; thousands of neonates were born with congenital microcephaly due to ZIKV exposure in utero, which also included a spectrum of injuries to the brain, eyes and spinal cord. In addition to congenital anomalies, permanent injury to fetal and neonatal organs, preterm birth, stillbirth and spontaneous abortion are known consequences of a broader group of infectious diseases including group B streptococcus (GBS), Listeria monocytogenes, Influenza A virus (IAV), and Human Immunodeficiency Virus (HIV). Animal models are crucial for determining the mechanism of how these various infectious diseases induce teratogenesis or organ injury, as well as testing novel therapeutics for fetal or neonatal protection. Other mammalian models differ in many respects from human pregnancy including placentation, labor physiology, reproductive tract anatomy, timeline of fetal development and reproductive toxicology. In contrast, non-human primates (NHP) most closely resemble human pregnancy and exhibit key similarities that make them ideal for research to discover the mechanisms of injury and for testing vaccines and therapeutics to prevent teratogenesis, fetal and neonatal injury and adverse pregnancy outcomes (e.g., stillbirth or spontaneous abortion). In this review, we emphasize key contributions of the NHP model pre-clinical research for ZIKV, HCMV, HIV, IAV, L. monocytogenes, Ureaplasma species, and GBS. This work represents the foundation for development and testing of preventative and therapeutic strategies to inhibit infectious injury of human fetuses and neonates.
Collapse
Affiliation(s)
- Miranda Li
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
- Department of Biological Sciences, Columbia University, New York, NY, United States
| | - Alyssa Brokaw
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Anna M. Furuta
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Brahm Coler
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Veronica Obregon-Perko
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, GA, United States
| | - Hsuan-Yuan Wang
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Charlotte E. Hotchkiss
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Thaddeus G. Golos
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Lakshmi Rajagopal
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Kristina M. Adams Waldorf
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
30
|
Roda WC, Liu S, Power C, Li MY. Modeling the Effects of Latency Reversing Drugs During HIV-1 and SIV Brain Infection with Implications for the "Shock and Kill" Strategy. Bull Math Biol 2021; 83:39. [PMID: 33712983 DOI: 10.1007/s11538-021-00875-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 02/25/2021] [Indexed: 11/30/2022]
Abstract
Combination antiretroviral therapy (cART) has greatly increased life expectancy for human immunodeficiency virus-1 (HIV-1)-infected patients. Even given the remarkable success of cART, the virus persists in many different cells and tissues. The presence of viral reservoirs represents a major obstacle to HIV-1 eradication. These viral reservoirs contain latently infected long-lived cells. The "Shock and Kill" therapeutic strategy aims to reactivate latently infected cells by latency reversing agents (LRAs) and kill these reactivated cells by strategies involving the host immune system. The brain is a natural anatomical reservoir for HIV-1 infection. Brain macrophages, including microglia and perivascular macrophages, display productive HIV-1 infection. A mathematical model was used to analyze the dynamics of latently and productively infected brain macrophages during viral infection and this mathematical model enabled prediction of the effects of LRAs applied to the "Shock and Kill" strategy in the brain. The model was calibrated using reported data from simian immunodeficiency virus (SIV) studies. Our model produces the overarching observation that effective cART can suppress productively infected brain macrophages but leaves a residual latent viral reservoir in brain macrophages. In addition, our model demonstrates that there exists a parameter regime wherein the "Shock and Kill" strategy can be safe and effective for SIV infection in the brain. The results indicate that the "Shock and Kill" strategy can restrict brain viral RNA burden associated with severe neuroinflammation and can lead to the eradication of the latent reservoir of brain macrophages.
Collapse
Affiliation(s)
- Weston C Roda
- Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, AB, T6G 2G1, Canada.
| | - Suli Liu
- School of Mathematics, Jilin University, Changchun, 130012, Jilin Province, China
| | - Christopher Power
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Michael Y Li
- Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, AB, T6G 2G1, Canada
| |
Collapse
|
31
|
HIV Infection and Related Mental Disorders. Brain Sci 2021; 11:brainsci11020248. [PMID: 33671125 PMCID: PMC7922767 DOI: 10.3390/brainsci11020248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 01/13/2023] Open
Abstract
Over the more than thirty-year period of the human immunodeficiency virus type 1 (HIV-1) epidemic, many data have been accumulated indicating that HIV infection predisposes one to the development of mental pathologies. It has been proven that cognitive disorders in HIV-positive individuals are the result of the direct exposure of the virus to central nervous system (CNS) cells. The use of antiretroviral therapy has significantly reduced the number of cases of mental disorders among people infected with HIV. However, the incidence of moderate to mild cognitive impairment at all stages of HIV infection is still quite high. This review describes the most common forms of mental pathology that occur in people living with HIV and presents the current concepts on the possible pathogenetic mechanisms of the influence of human immunodeficiency virus (HIV-1) and its viral proteins on the cells of the CNS and the CNS’s functions. This review also provides the current state of knowledge on the impact of the antiretroviral therapy on the development of mental pathologies in people living with HIV, as well as current knowledge on the interactions between antiretroviral and psychotropic drugs that occur under their simultaneous administration.
Collapse
|
32
|
Alamer E, Zhong C, Hajnik R, Soong L, Hu H. Modulation of BRD4 in HIV epigenetic regulation: implications for finding an HIV cure. Retrovirology 2021; 18:3. [PMID: 33413475 PMCID: PMC7792063 DOI: 10.1186/s12977-020-00547-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/30/2020] [Accepted: 12/23/2020] [Indexed: 12/16/2022] Open
Abstract
Following reverse transcription, HIV viral DNA is integrated into host cell genomes and establishes a stable latent infection, which has posed a major obstacle for obtaining a cure for HIV. HIV proviral transcription is regulated in cellular reservoirs by complex host epigenetic and transcriptional machineries. The Bromodomain (BD) and Extra-Terminal Domain (ET) protein, BRD4, is an important epigenetic reader that interacts with acetyl-histones and a variety of chromatin and transcriptional regulators to control gene expression, including HIV. Modulation of BRD4 by a pan BET inhibitor (JQ1) has been shown to activate HIV transcription. Recent studies by my group and others indicate that the function of BRD4 is versatile and its effects on HIV transcription may depend on the partner proteins or pathways engaged by BRD4. Our studies have reported a novel class of small-molecule modulators that are distinct from JQ1 but induce HIV transcriptional suppression through BRD4. Herein, we reviewed recent research on the modulation of BRD4 in HIV epigenetic regulation and discussed their potential implications for finding an HIV cure.
Collapse
Affiliation(s)
- Edrous Alamer
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), MRB 4.142A, 301 University Blvd, Galveston, TX, 77555, USA.,Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia.,Medical Research Center, Jazan University, Jazan, 45142, Saudi Arabia
| | - Chaojie Zhong
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), MRB 4.142A, 301 University Blvd, Galveston, TX, 77555, USA
| | - Renee Hajnik
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), MRB 4.142A, 301 University Blvd, Galveston, TX, 77555, USA
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), MRB 4.142A, 301 University Blvd, Galveston, TX, 77555, USA.,Institute for Human Infections and Immunity, Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Haitao Hu
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), MRB 4.142A, 301 University Blvd, Galveston, TX, 77555, USA. .,Institute for Human Infections and Immunity, Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
33
|
Fitting S, McRae M, Hauser KF. Opioid and neuroHIV Comorbidity - Current and Future Perspectives. J Neuroimmune Pharmacol 2020; 15:584-627. [PMID: 32876803 PMCID: PMC7463108 DOI: 10.1007/s11481-020-09941-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 07/02/2020] [Indexed: 12/14/2022]
Abstract
With the current national opioid crisis, it is critical to examine the mechanisms underlying pathophysiologic interactions between human immunodeficiency virus (HIV) and opioids in the central nervous system (CNS). Recent advances in experimental models, methodology, and our understanding of disease processes at the molecular and cellular levels reveal opioid-HIV interactions with increasing clarity. However, despite the substantial new insight, the unique impact of opioids on the severity, progression, and prognosis of neuroHIV and HIV-associated neurocognitive disorders (HAND) are not fully understood. In this review, we explore, in detail, what is currently known about mechanisms underlying opioid interactions with HIV, with emphasis on individual HIV-1-expressed gene products at the molecular, cellular and systems levels. Furthermore, we review preclinical and clinical studies with a focus on key considerations when addressing questions of whether opioid-HIV interactive pathogenesis results in unique structural or functional deficits not seen with either disease alone. These considerations include, understanding the combined consequences of HIV-1 genetic variants, host variants, and μ-opioid receptor (MOR) and HIV chemokine co-receptor interactions on the comorbidity. Lastly, we present topics that need to be considered in the future to better understand the unique contributions of opioids to the pathophysiology of neuroHIV. Graphical Abstract Blood-brain barrier and the neurovascular unit. With HIV and opiate co-exposure (represented below the dotted line), there is breakdown of tight junction proteins and increased leakage of paracellular compounds into the brain. Despite this, opiate exposure selectively increases the expression of some efflux transporters, thereby restricting brain penetration of specific drugs.
Collapse
Affiliation(s)
- Sylvia Fitting
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-3270, USA
| | - MaryPeace McRae
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 1217 East Marshall Street, Richmond, VA, 23298-0613, USA.
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298-0709, USA.
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, 203 East Cary Street, Richmond, VA, 23298-0059, USA.
| |
Collapse
|
34
|
HIV Infection and Persistence in Pulmonary Mucosal Double Negative T Cells In Vivo. J Virol 2020; 94:JVI.01788-20. [PMID: 32967958 PMCID: PMC7925170 DOI: 10.1128/jvi.01788-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 09/12/2020] [Indexed: 11/20/2022] Open
Abstract
The lungs are relatively unexplored anatomical human immunodeficiency virus (HIV) reservoirs in the antiretroviral therapy (ART) era. Double negative (DN) T cells are a subset of T cells that lack expression of CD4 and CD8 (CD4- CD8-) and may have both regulatory and effector functions during HIV infection. Notably, circulating DN T cells were previously described as cellular HIV reservoirs. Here, we undertook a thorough analysis of pulmonary versus blood DN T cells of people living with HIV (PLWH) under ART. Bronchoalveolar lavage (BAL) fluid and matched peripheral blood were collected from 35 PLWH on ART and 16 uninfected volunteers without respiratory symptoms. Both PLWH and HIV-negative (HIV-) adults displayed higher frequencies of DN T cells in BAL versus blood, and these cells mostly exhibited an effector memory phenotype. In PLWH, pulmonary mucosal DN T cells expressed higher levels of HLA-DR and several cellular markers associated with HIV persistence (CCR6, CXCR3, and PD-1) than blood. We also observed that DN T cells were less senescent (CD28- CD57+) and expressed less immunosuppressive ectonucleotidase (CD73/CD39), granzyme B, and perforin in the BAL fluid than in the blood of PLWH. Importantly, fluorescence-activated cell sorter (FACS)-sorted DN T cells from the BAL fluid of PLWH under suppressive ART harbored HIV DNA. Using the humanized bone marrow-liver-thymus (hu-BLT) mouse model of HIV infection, we observed higher infection frequencies of lung DN T cells than those of the blood and spleen in both early and late HIV infection. Overall, our findings show that HIV is seeded in pulmonary mucosal DN T cells early following infection and persists in these potential cellular HIV reservoirs even during long-term ART.IMPORTANCE Reservoirs of HIV during ART are the primary reasons why HIV/AIDS remains an incurable disease. Indeed, HIV remains latent and unreachable by antiretrovirals in cellular and anatomical sanctuaries, preventing its eradication. The lungs have received very little attention compared to other anatomical reservoirs despite being immunological effector sites exhibiting characteristics ideal for HIV persistence. Furthermore, PLWH suffer from a high burden of pulmonary non-opportunistic infections, suggesting impaired pulmonary immunity despite ART. Meanwhile, various immune cell populations have been proposed to be cellular reservoirs in blood, including CD4- CD8- DN T cells, a subset that may originate from CD4 downregulation by HIV proteins. The present study aims to describe DN T cells in human and humanized mice lungs in relation to intrapulmonary HIV burden. The characterization of DN T cells as cellular HIV reservoirs and the lungs as an anatomical HIV reservoir will contribute to the development of targeted HIV eradication strategies.
Collapse
|
35
|
Whyte-Allman SK, Bendayan R. HIV-1 Sanctuary Sites-the Role of Membrane-Associated Drug Transporters and Drug Metabolic Enzymes. AAPS JOURNAL 2020; 22:118. [PMID: 32875457 DOI: 10.1208/s12248-020-00498-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/06/2020] [Indexed: 02/08/2023]
Abstract
Despite significant advances in the treatment of human immunodeficiency virus-1 (HIV) infection with highly active antiretroviral drug therapy, the persistence of the virus in cellular and anatomic reservoirs is a major obstacle preventing total HIV eradication. Viral persistence could result from a variety of contributing factors including, but not limited to, non-adherence to treatment and adverse drug reactions, latently infected cells carrying replication-competent virus, drug-drug interactions, and inadequate antiretroviral drug (ARV) concentrations reached in several anatomic sites such as the brain, testis, and gut-associated lymphoid tissues. The distribution of ARVs at specific sites of infection is primarily dependent on drug physicochemical properties and drug plasma protein binding, as well as drug efflux, influx, and metabolic processes. A thorough understanding of the functional roles of drug transporters and metabolic enzymes in the disposition of ARVs in immune cell types and tissues that are characterized as HIV reservoirs and sanctuaries is critical to overcome the challenge of suboptimal drug distribution at sites of persistent HIV infection. This review summarizes the current knowledge related to the expression and function of drug transporters and metabolic enzymes in HIV cellular and anatomic reservoirs, and their potential contribution to drug-drug interactions and insufficient drug concentration at these sites.
Collapse
Affiliation(s)
- Sana-Kay Whyte-Allman
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario, M5S 3M2, Canada
| | - Reina Bendayan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario, M5S 3M2, Canada.
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Perinatal HIV-1 infection is associated with an increased risk for neurologic impairments. With limited access to clinical specimens, animal models could advance our understanding of pediatric central nervous system (CNS) disease and viral persistence. Here, we summarize current findings on HIV-1 CNS infection from nonhuman primate (NHP) models and discuss their implications for improving pediatric clinical outcomes. RECENT FINDINGS SIV/SHIV can be found in the CNS of infant macaques within 48 h of challenge. Recent studies show an impermeable BBB during SIV infection, suggesting neuroinvasion in post-partum infection is likely not wholly attributed to barrier dysfunction. Histopathological findings reveal dramatic reductions in hippocampal neuronal populations and myelination in infected infant macaques, providing a link for cognitive impairments seen in pediatric cases. Evidence from humans and NHPs support the CNS as a functional latent reservoir, harbored in myeloid cells that may require unique eradication strategies. Studies in NHP models are uncovering early events, causes, and therapeutic targets of CNS disease as well as highlighting the importance of age-specific studies that capture the distinct features of pediatric HIV-1 infection.
Collapse
Affiliation(s)
| | - Katherine Bricker
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.
- Emory+Children's Center for Childhood Infections and Vaccines, Atlanta, GA, USA.
| |
Collapse
|
37
|
Omeragic A, Kayode O, Hoque MT, Bendayan R. Potential pharmacological approaches for the treatment of HIV-1 associated neurocognitive disorders. Fluids Barriers CNS 2020; 17:42. [PMID: 32650790 PMCID: PMC7350632 DOI: 10.1186/s12987-020-00204-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023] Open
Abstract
HIV associated neurocognitive disorders (HAND) are the spectrum of cognitive impairments present in patients infected with human immunodeficiency virus type 1 (HIV-1). The number of patients affected with HAND ranges from 30 to 50% of HIV infected individuals and although the development of combinational antiretroviral therapy (cART) has improved longevity, HAND continues to pose a significant clinical problem as the current standard of care does not alleviate or prevent HAND symptoms. At present, the pathological mechanisms contributing to HAND remain unclear, but evidence suggests that it stems from neuronal injury due to chronic release of neurotoxins, chemokines, viral proteins, and proinflammatory cytokines secreted by HIV-1 activated microglia, macrophages and astrocytes in the central nervous system (CNS). Furthermore, the blood-brain barrier (BBB) not only serves as a route for HIV-1 entry into the brain but also prevents cART therapy from reaching HIV-1 brain reservoirs, and therefore could play an important role in HAND. The goal of this review is to discuss the current data on the epidemiology, pathology and research models of HAND as well as address the potential pharmacological treatment approaches that are being investigated.
Collapse
Affiliation(s)
- Amila Omeragic
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Room 1001, Toronto, ON, M5S 3M2, Canada
| | - Olanre Kayode
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Room 1001, Toronto, ON, M5S 3M2, Canada
| | - Md Tozammel Hoque
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Room 1001, Toronto, ON, M5S 3M2, Canada
| | - Reina Bendayan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Room 1001, Toronto, ON, M5S 3M2, Canada.
| |
Collapse
|
38
|
HIV-1 Persistence and Chronic Induction of Innate Immune Responses in Macrophages. Viruses 2020; 12:v12070711. [PMID: 32630058 PMCID: PMC7412260 DOI: 10.3390/v12070711] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
A hallmark of HIV-1 infection is chronic inflammation, which plays a significant role in disease pathogenesis. Acute HIV infection induces robust inflammatory responses, which are insufficient to prevent or eliminate virus in mucosal tissues. While establishment of viral set-point is coincident with downregulation of acute innate responses, systemic inflammatory responses persist during the course of chronic HIV infection. Since the introduction of combination antiviral therapy (cART), most HIV-1+ individuals can suppress viremia under detection levels for decades. However, chronic immune activation persists and has been postulated to cause HIV associated non-AIDS complications (HANA). Importantly, inflammatory cytokines and activation markers associated with macrophages are strongly and selectively correlated with the incidence of HIV-associated neurocognitive disorder (HAND), cardiovascular dysfunctions (CVD) and other HANA conditions. In this review, we discuss the roles of macrophages in facilitating viral persistence and contributing to generation of persistent inflammatory responses.
Collapse
|
39
|
Epigenetic Suppression of HIV in Myeloid Cells by the BRD4-Selective Small Molecule Modulator ZL0580. J Virol 2020; 94:JVI.01880-19. [PMID: 32188727 DOI: 10.1128/jvi.01880-19] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 03/08/2020] [Indexed: 01/06/2023] Open
Abstract
Brain-resident microglia and myeloid cells (perivascular macrophages) are important HIV reservoirs in vivo, especially in the central nervous system (CNS). Despite antiretroviral therapy (ART), low-level persistent HIV replication in these reservoirs remains detectable, which contributes to neuroinflammation and neurological disorders in HIV-infected patients. New approaches complementary to ART to repress residual HIV replication in CNS reservoirs are needed. Our group has recently identified a BRD4-selective small molecule modulator (ZL0580) that induces the epigenetic suppression of HIV. Here, we examined the effects of this compound on HIV in human myeloid cells. We found that ZL0580 induces potent and durable suppression of both induced and basal HIV transcription in microglial cells (HC69) and monocytic cell lines (U1 and OM10.1). Pretreatment of microglia with ZL0580 renders them more refractory to latent HIV reactivation, indicating an epigenetic reprogramming effect of ZL0580 on HIV long terminal repeat (LTR) in microglia. We also demonstrate that ZL0580 induces repressive effect on HIV in human primary monocyte-derived macrophages (MDMs) by promoting HIV suppression during ART treatment. Mechanistically, ZL0580 inhibits Tat transactivation in microglia by disrupting binding of Tat to CDK9, a process key to HIV transcription elongation. High-resolution micrococcal nuclease mapping showed that ZL0580 induces a repressive chromatin structure at the HIV LTR. Taken together, our data suggest that ZL0580 represents a potential approach that could be used in combination with ART to suppress residual HIV replication and/or latent HIV reactivation in CNS reservoirs, thereby reducing HIV-associated neuroinflammation.IMPORTANCE Brain-resident microglia and perivascular macrophages are important HIV reservoirs in the CNS. Persistent viral replication and latent HIV reactivation in the CNS, even under ART, are believed to occur, causing neuroinflammation and neurological disorders in HIV-infected patients. It is critical to identify new approaches that can control residual HIV replication and/or latent HIV reactivation in these reservoirs. We here report that the BRD4-selective small molecule modulator, ZL0580, induces potent and durable suppression of HIV in human microglial and monocytic cell lines. Using an in vitro HIV-infected, ART-treated MDM model, we show that ZL0580 also induces suppressive effect on HIV in human primary macrophages. The significance of our research is that it suggests a potential new approach that has utility in combination with ART to suppress residual HIV replication and/or HIV reactivation in CNS reservoirs, thereby reducing neuroinflammation and neurological disorders in HIV-infected individuals.
Collapse
|
40
|
Agarwal Y, Beatty C, Biradar S, Castronova I, Ho S, Melody K, Bility MT. Moving beyond the mousetrap: current and emerging humanized mouse and rat models for investigating prevention and cure strategies against HIV infection and associated pathologies. Retrovirology 2020; 17:8. [PMID: 32276640 PMCID: PMC7149862 DOI: 10.1186/s12977-020-00515-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 03/31/2020] [Indexed: 12/22/2022] Open
Abstract
The development of safe and effective combination antiretroviral therapies for human immunodeficiency virus (HIV) infection over the past several decades has significantly reduced HIV-associated morbidity and mortality. Additionally, antiretroviral drugs have provided an effective means of protection against HIV transmission. Despite these advances, significant limitations exist; namely, the inability to eliminate HIV reservoirs, the inability to reverse lymphoid tissues damage, and the lack of an effective vaccine for preventing HIV transmission. Evaluation of the safety and efficacy of therapeutics and vaccines for eliminating HIV reservoirs and preventing HIV transmission requires robust in vivo models. Since HIV is a human-specific pathogen, that targets hematopoietic lineage cells and lymphoid tissues, in vivo animal models for HIV-host interactions require incorporation of human hematopoietic lineage cells and lymphoid tissues. In this review, we will discuss the construction of mouse models with human lymphoid tissues and/or hematopoietic lineage cells, termed, human immune system (HIS)-humanized mice. These HIS-humanized mouse models can support the development of functional human innate and adaptive immune cells, along with primary (thymus) and secondary (spleen) lymphoid tissues. We will discuss applications of HIS-humanized mouse models in evaluating the safety and efficacy of therapeutics against HIV reservoirs and associated immunopathology, and delineate the human immune response elicited by candidate HIV vaccines. In addition to focusing on how these HIS-humanized mouse models have already furthered our understanding of HIV and contributed to HIV therapeutics development, we discuss how emerging HIS-humanized rat models could address the limitations of HIS-mouse models.
Collapse
Affiliation(s)
- Yash Agarwal
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cole Beatty
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shivkumar Biradar
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Isabella Castronova
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sara Ho
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kevin Melody
- Galveston National Laboratory and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Moses Turkle Bility
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
41
|
Omeragic A, Saikali MF, Currier S, Volsky DJ, Cummins CL, Bendayan R. Selective peroxisome proliferator-activated receptor-gamma modulator, INT131 exhibits anti-inflammatory effects in an EcoHIV mouse model. FASEB J 2019; 34:1996-2010. [PMID: 31907999 DOI: 10.1096/fj.201901874r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/24/2019] [Accepted: 11/01/2019] [Indexed: 12/13/2022]
Abstract
Despite the use of antiretroviral therapy for the treatment of HIV-1 infection, cognitive impairments, that is, HIV-1-associated neurocognitive disorders remain prevalent potentially due to persistent viral replication, production of viral proteins, associated brain inflammation or in certain instances, antiretroviral neurotoxicity. Cellular targets in the brain include microglia which in response to infection release inflammatory markers and viral proteins. Evidence suggests that PPARγ agonists exert anti-inflammatory properties in neurological disorders. However, these agonists namely, thiazolidinediones have limited use in the clinic due to reported adverse side effects. INT131 is a novel non-thiazolidinedione compound that belongs to a new class of drugs known as selective PPARγ modulators. INT131 is considered to have a safer profile; however, its neuroprotective role in vivo is not known.The goal of this study was to examine the effect of INT131 in the context of EcoHIV-induced inflammation in vitro, in primary cultures of mouse glial cells and in vivo, in a mouse model of EcoHIV-associated brain inflammation, as well as characterize its pharmacokinetic properties and brain penetration. In primary cultures of glial cells and in the in vivo mouse model, EcoHIV exposure resulted in a significant elevation of inflammatory markers such as TNFα, IL-1β, CCL3, and C3 which were attenuated with INT131 treatment. Pharmacokinetic analyses revealed that INT131 penetrates into the brain with a brain to blood partition ratio Kp value of 8.5%. Overall, this is the first report to demonstrate that INT131 could be a potential candidate for the treatment of HIV-1-associated brain inflammation.
Collapse
Affiliation(s)
- Amila Omeragic
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Michael F Saikali
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Sydney Currier
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - David J Volsky
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Reina Bendayan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
42
|
Abstract
: The persistence of HIV in the central nervous system is somewhat controversial particularly in the context of HIV viral suppression from combined antiretroviral therapy. Further, its significance in relation to HIV pathogenesis in the context of HIV-associated neurocognitive disorders, systemic HIV pathogenesis, and eradication in general, but especially from the brain, are even more contentious. This review will discuss each of these aspects in detail, highlighting new data, particularly from recent conference presentations.
Collapse
|
43
|
Rivera J, Isidro RA, Loucil-Alicea RY, Cruz ML, Appleyard CB, Isidro AA, Chompre G, Colon-Rivera K, Noel RJ. Infusion of HIV-1 Nef-expressing astrocytes into the rat hippocampus induces enteropathy and interstitial pneumonitis and increases blood-brain-barrier permeability. PLoS One 2019; 14:e0225760. [PMID: 31774879 PMCID: PMC6881014 DOI: 10.1371/journal.pone.0225760] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 11/12/2019] [Indexed: 12/16/2022] Open
Abstract
Even though HIV-1 replication can be suppressed by combination antiretroviral therapy (cART) inflammatory processes still occur, contributing to comorbidities. Comorbidities are attributed to variety of factors, including HIV-1 mediated inflammation. Several HIV-1 proteins mediate central nervous system (CNS) inflammation, including Nef. Nef is an early HIV-1 protein, toxic to neurons and glia and is sufficient to cause learning impairment similar to some deficits observed in HIV-1 associated neurocognitive disorders. To determine whether hippocampal Nef expression by astrocytes contributes to comorbidities, specifically peripheral inflammation, we infused Sprague Dawley rats with GFP- (control) or Nef-transfected astrocytes into the right hippocampus. Brain, lung, and ileum were collected postmortem for the measurement of inflammatory markers. Increased blood-brain-barrier permeability and serum IL-1β levels were detected in the Nef-treated rats. The lungs of Nef-treated rats demonstrated leukocyte infiltration, macrophage upregulation, and enhanced vascular permeability. Ileal tissue showed reactive follicular lymphoid hyperplasia, increased permeability and macrophage infiltration. The intracerebroventricular application of IL-1 receptor antagonist reduced infiltration of immune cells into ileum and lung, indicating the important role of IL-1β in mediating the spread of inflammation from the brain to other tissues. This suggests that localized expression of a single viral protein, HIV-1 Nef, can contribute to a broader inflammatory response by upregulation of IL-1β. Further, these results suggest that Nef contributes to the chronic inflammation seen in HIV patients, even in those whose viremia is controlled by cART.
Collapse
Affiliation(s)
- Jocelyn Rivera
- HIV-1 Immunopathogenesis Laboratory, The Wistar Institute, Philadelphia, PA, United States of America
| | - Raymond A. Isidro
- Department of Basic Sciences, Ponce Health Sciences University, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Raisa Y. Loucil-Alicea
- Department of Basic Sciences, Ponce Health Sciences University, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Myrella L. Cruz
- Department of Basic Sciences, Ponce Health Sciences University, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Caroline B. Appleyard
- Department of Basic Sciences, Ponce Health Sciences University, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Angel A. Isidro
- Department of Basic Sciences, Ponce Health Sciences University, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Gladys Chompre
- Department of Biology, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico, United States of America
| | - Krystal Colon-Rivera
- HIV-1 Immunopathogenesis Laboratory, The Wistar Institute, Philadelphia, PA, United States of America
| | - Richard J. Noel
- Department of Basic Sciences, Ponce Health Sciences University, Ponce Research Institute, Ponce, Puerto Rico, United States of America
- * E-mail:
| |
Collapse
|
44
|
Wallet C, De Rovere M, Van Assche J, Daouad F, De Wit S, Gautier V, Mallon PWG, Marcello A, Van Lint C, Rohr O, Schwartz C. Microglial Cells: The Main HIV-1 Reservoir in the Brain. Front Cell Infect Microbiol 2019; 9:362. [PMID: 31709195 PMCID: PMC6821723 DOI: 10.3389/fcimb.2019.00362] [Citation(s) in RCA: 221] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Despite efficient combination of the antiretroviral therapy (cART), which significantly decreased mortality and morbidity of HIV-1 infection, a definitive HIV cure has not been achieved. Hidden HIV-1 in cellular and anatomic reservoirs is the major hurdle toward a functional cure. Microglial cells, the Central Nervous system (CNS) resident macrophages, are one of the major cellular reservoirs of latent HIV-1. These cells are believed to be involved in the emergence of drugs resistance and reseeding peripheral tissues. Moreover, these long-life reservoirs are also involved in the development of HIV-1-associated neurocognitive diseases (HAND). Clearing these infected cells from the brain is therefore crucial to achieve a cure. However, many characteristics of microglial cells and the CNS hinder the eradication of these brain reservoirs. Better understandings of the specific molecular mechanisms of HIV-1 latency in microglial cells should help to design new molecules and new strategies preventing HAND and achieving HIV cure. Moreover, new strategies are needed to circumvent the limitations associated to anatomical sanctuaries with barriers such as the blood brain barrier (BBB) that reduce the access of drugs.
Collapse
Affiliation(s)
- Clementine Wallet
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| | - Marco De Rovere
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| | - Jeanne Van Assche
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| | - Fadoua Daouad
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| | - Stéphane De Wit
- Division of Infectious Diseases, Saint-Pierre University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Virginie Gautier
- UCD Centre for Experimental Pathogen Host Research (CEPHR), School of Medicine, University College Dublin, Dublin, Ireland
| | - Patrick W G Mallon
- UCD Centre for Experimental Pathogen Host Research (CEPHR), School of Medicine, University College Dublin, Dublin, Ireland
| | - Alessandro Marcello
- Laboratory of Molecular Virology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Carine Van Lint
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Olivier Rohr
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| | - Christian Schwartz
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| |
Collapse
|
45
|
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: 7.4] [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
|
46
|
Henrich TJ, Hsue PY, VanBrocklin H. Seeing Is Believing: Nuclear Imaging of HIV Persistence. Front Immunol 2019; 10:2077. [PMID: 31572355 PMCID: PMC6751256 DOI: 10.3389/fimmu.2019.02077] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/16/2019] [Indexed: 12/19/2022] Open
Abstract
A major obstacle to HIV eradication is the presence of infected cells that persist despite suppressive antiretroviral therapy (ART). HIV largely resides outside of the peripheral circulation, and thus, numerous anatomical and lymphoid compartments that have the capacity to harbor HIV are inaccessible to routine sampling. As a result, there is a limited understanding of the tissue burden of HIV infection or anatomical distribution of HIV transcriptional and translational activity. Novel, non-invasive, in vivo methods are urgently needed to address this fundamental gap in knowledge. In this review, we discuss past and current nuclear imaging approaches that have been applied to HIV infection with an emphasis on current strategies to implement positron emission tomography (PET)-based imaging to directly visualize and characterize whole-body HIV burden. These imaging approaches have various limitations, such as the potential for limited PET sensitivity and specificity in the setting of ART suppression or low viral burden. However, recent advances in high-sensitivity, total-body PET imaging platforms and development of new radiotracer technologies that may enhance anatomical penetration of target-specific tracer molecules are discussed. Potential strategies to image non-viral markers of HIV tissue burden or focal immune perturbation are also addressed. Overall, emerging nuclear imaging techniques and platforms may play an important role in the development of novel therapeutic and HIV reservoir eradication strategies.
Collapse
Affiliation(s)
- Timothy J Henrich
- Division of Experimental Medicine, Department of Medicine, University of San Francisco, San Francisco, CA, United States
| | - Priscilla Y Hsue
- Division of Cardiology, Department of Medicine, University of San Francisco, San Francisco, CA, United States
| | - Henry VanBrocklin
- Radiopharmaceutical Research Program, Center for Molecular and Functional Imaging, University of San Francisco, San Francisco, CA, United States
| |
Collapse
|
47
|
Patel SH, Ismaiel OA, Mylott WR, Yuan M, McClay JL, Paris JJ, Hauser KF, McRae M. Cell-type specific differences in antiretroviral penetration and the effects of HIV-1 Tat and morphine among primary human brain endothelial cells, astrocytes, pericytes, and microglia. Neurosci Lett 2019; 712:134475. [PMID: 31491466 DOI: 10.1016/j.neulet.2019.134475] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/26/2019] [Accepted: 08/31/2019] [Indexed: 12/18/2022]
Abstract
The inability to achieve adequate intracellular antiretroviral concentrations may contribute to HIV persistence within the brain and to neurocognitive deficits in opioid abusers. To investigate, intracellular antiretroviral concentrations were measured in primary human astrocytes, microglia, pericytes, and brain microvascular endothelial cells (BMECs), and in an immortalized brain endothelial cell line (hCMEC/D3). HIV-1 Tat and morphine effects on intracellular antiretroviral concentrations also were evaluated. After pretreatment for 24 h with vehicle, HIV-1 Tat, morphine, or combined Tat and morphine, cells were incubated for 1 h with equal concentrations of a mixture of tenofovir, emtricitabine, and dolutegravir at one of two concentrations (5 μM or 10 μM). Intracellular drug accumulation was measured using LC-MS/MS. Drug penetration differed depending on the drug, the extracellular concentration used for dosing, and cell type. Significant findings included: 1) Dolutegravir (at 5 μM or 10 μM) accumulated more in HBMECs than other cell types. 2) At 5 μM, intracellular emtricitabine levels were higher in microglia than other cell types; while at 10 μM, emtricitabine accumulation was greatest in HBMECs. 3) Tenofovir (5 or 10 μM extracellular dosing) displayed greater accumulation inside HBMECs than in other cell types. 4) After Tat and/or morphine pretreatment, the relative accumulation of antiretroviral drugs was greater in morphine-exposed HBMECs compared to other treatments. The opposite effect was observed in astrocytes in which morphine exposure decreased drug accumulation. In summary, the intracellular accumulation of antiretroviral drugs differed depending on the particular drug involved, the concentration of the applied antiretroviral drug, and the cell type targeted. Moreover, morphine, and to a lesser extent Tat, exposure also had differential effects on antiretroviral accumulation. These data highlight the complexity of optimizing brain-targeted HIV therapeutics, especially in the setting of chronic opioid use or misuse.
Collapse
Affiliation(s)
- Sulay H Patel
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Omnia A Ismaiel
- PPD Laboratories, Richmond, VA, USA; Department of Analytical Chemistry, Faculty of Pharmacy, Zagazig University, Egypt
| | | | | | - Joseph L McClay
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Jason J Paris
- Department of BioMolecular Sciences, School of Pharmacy, The University of Mississippi, University, MS, USA
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - MaryPeace McRae
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
48
|
Balcom EF, Roda WC, Cohen EA, Li MY, Power C. HIV-1 persistence in the central nervous system: viral and host determinants during antiretroviral therapy. Curr Opin Virol 2019; 38:54-62. [PMID: 31390580 DOI: 10.1016/j.coviro.2019.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/03/2019] [Accepted: 06/05/2019] [Indexed: 02/07/2023]
Abstract
Despite remarkable therapeutic advances in the past two decades, the elimination of human immunodeficiency virus type 1 (HIV-1) from latent reservoirs constitutes a major barrier to eradication and preventing neurological disease associated with HIV/AIDS. Invasion of the central nervous system (CNS) by HIV-1 occurs early in infection, leading to viral infection and productive persistence in brain macrophage-like cells (BMCs) including resident microglia and infiltrating macrophages. HIV-1 persistence in the brain and chronic neuroinflammation occur despite effective treatment with antiretroviral therapy (ART). This review examines the evidence from clinical studies, in vivo and in vitro models for HIV-1 CNS persistence, as well as therapeutic considerations in targeting latent CNS reservoirs.
Collapse
Affiliation(s)
- E F Balcom
- Department of Medicine (Neurology), University of Alberta, Edmonton, AB, Canada
| | - W C Roda
- Department of Mathematical & Statistical Sciences, University of Alberta, Edmonton, AB, Canada
| | - E A Cohen
- Departments of Microbiology and Immunology, University of Montreal, Montreal Clinical Research Institute, Montreal, QC, Canada
| | - M Y Li
- Department of Mathematical & Statistical Sciences, University of Alberta, Edmonton, AB, Canada
| | - C Power
- Department of Medicine (Neurology), University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
49
|
Patel SH, Ismaiel OA, Mylott WR, Yuan M, Hauser KF, McRae M. Simultaneous determination of intracellular concentrations of tenofovir, emtricitabine, and dolutegravir in human brain microvascular endothelial cells using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Anal Chim Acta 2019; 1056:79-87. [PMID: 30797464 PMCID: PMC6486649 DOI: 10.1016/j.aca.2019.01.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/27/2018] [Accepted: 01/04/2019] [Indexed: 12/17/2022]
Abstract
Combination antiretroviral therapy (cART) regimens are recommended for HIV patients to better achieve and maintain plasma viral suppression. Despite adequate plasma viral suppression, HIV persists inside the brain, which is, in part thought to result from poor brain penetration of antiretroviral drugs. In this study, a simple and ultra-sensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for simultaneous determination of tenofovir, emtricitabine, and dolutegravir in cell lysates of an immortalized human brain microvascular endothelial cell line (hCMEC/D3) was developed and validated. Analytes were separated on a reverse phase C18 column using water and 0.1% formic acid in acetonitrile as mobile phases. The analytes were detected using positive electrospray ionization mode with multiple reaction monitoring (MRM). The assay was linear in the concentration range of 0.1-100 ng mL-1 for all analytes. Intra- and inter-assay precision and accuracy were within ±13.33% and ±10.53%, respectively. This approach described herein was used to determine the intracellular accumulation of tenofovir, emtricitabine, dolutegravir simultaneously in hCMEC/D3 cells samples.
Collapse
Affiliation(s)
- Sulay H Patel
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, P.O Box 980533, 410 N 12th Street, Richmond, VA, 23298-0533, USA
| | - Omnia A Ismaiel
- PPD Laboratories, Richmond, VA, USA; Department of Analytical Chemistry, Faculty of Pharmacy, Zagazig University, Egypt
| | | | | | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, P.O. Box 980613, 1217 East Marshall Street, Richmond, VA, 23298, USA
| | - MaryPeace McRae
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, P.O Box 980533, 410 N 12th Street, Richmond, VA, 23298-0533, USA.
| |
Collapse
|
50
|
Wallet C, De Rovere M, Van Assche J, Daouad F, De Wit S, Gautier V, Mallon PWG, Marcello A, Van Lint C, Rohr O, Schwartz C. Microglial Cells: The Main HIV-1 Reservoir in the Brain. Front Cell Infect Microbiol 2019. [PMID: 31709195 DOI: 10.3389/fcimb.2019.00362/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2023] Open
Abstract
Despite efficient combination of the antiretroviral therapy (cART), which significantly decreased mortality and morbidity of HIV-1 infection, a definitive HIV cure has not been achieved. Hidden HIV-1 in cellular and anatomic reservoirs is the major hurdle toward a functional cure. Microglial cells, the Central Nervous system (CNS) resident macrophages, are one of the major cellular reservoirs of latent HIV-1. These cells are believed to be involved in the emergence of drugs resistance and reseeding peripheral tissues. Moreover, these long-life reservoirs are also involved in the development of HIV-1-associated neurocognitive diseases (HAND). Clearing these infected cells from the brain is therefore crucial to achieve a cure. However, many characteristics of microglial cells and the CNS hinder the eradication of these brain reservoirs. Better understandings of the specific molecular mechanisms of HIV-1 latency in microglial cells should help to design new molecules and new strategies preventing HAND and achieving HIV cure. Moreover, new strategies are needed to circumvent the limitations associated to anatomical sanctuaries with barriers such as the blood brain barrier (BBB) that reduce the access of drugs.
Collapse
Affiliation(s)
- Clementine Wallet
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| | - Marco De Rovere
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| | - Jeanne Van Assche
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| | - Fadoua Daouad
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| | - Stéphane De Wit
- Division of Infectious Diseases, Saint-Pierre University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Virginie Gautier
- UCD Centre for Experimental Pathogen Host Research (CEPHR), School of Medicine, University College Dublin, Dublin, Ireland
| | - Patrick W G Mallon
- UCD Centre for Experimental Pathogen Host Research (CEPHR), School of Medicine, University College Dublin, Dublin, Ireland
| | - Alessandro Marcello
- Laboratory of Molecular Virology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Carine Van Lint
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Olivier Rohr
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| | - Christian Schwartz
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| |
Collapse
|