1
|
Amegashie EA, Sikeola RO, Tagoe EA, Paintsil E, Torpey K, Quaye O. Oxidative Stress in People Living With HIV: Are Diverse Supplement Sources the Solution? Health Sci Rep 2025; 8:e70824. [PMID: 40330761 PMCID: PMC12054717 DOI: 10.1002/hsr2.70824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/18/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Background and Aim Antiretroviral therapy (ART) has reduced human immunodeficiency virus (HIV)/AIDS to a manageable chronic condition even though no cure exists. Despite ART control, latent HIV infection results in failed memory CD4 T-cell responses, immune overactivation, inflammation, oxidative stress, genomic instability, deoxyribonucleic acid (DNA) damage, and premature CD4 T-cell ageing. Overproduction of reactive oxygen species during oxidative stress can cause mitochondrial DNA damage, cancer, neurodegenerative and cardiovascular diseases, and premature aging in people living with HIV (PLWH). This review outlines current knowledge in oxidative stress among PLWH. Methods Google Scholar, Scopus, PubMed, and Science Direct were searched for literature conforming with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines from studies published from January 2013 to December 2023. A total of 75 studies from 22 countries were identified, with 52 studies carried out in human participants, 17 studies in cell lines, and 6 studies in animal models to assess oxidative stress levels. Results An increased oxidative stress with no changes in antioxidant levels was reported in HIV-positive smokers, and those on substance abuse. Long-term ART usage showed high levels of oxidative protein products and low levels of antioxidants when compared to short-term ART usage. The use of supplements such as N-acetylcysteine, selenium, and silibinin in animal models and cell lines showed increased cell viability, reduced reactive oxygen species, and increased antioxidant levels, which are promising therapeutic interventions that should be studied in PLWH to further help improve their disease outcomes. Conclusions Identifying extracts from natural and synthetic products with antioxidant effects will improve the general well-being of PLWH.
Collapse
Affiliation(s)
- Esimebia Adjovi Amegashie
- West African Center for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular BiologyUniversity of GhanaAccraGreater Accra RegionGhana
| | - Ruth Oyawole Sikeola
- West African Center for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular BiologyUniversity of GhanaAccraGreater Accra RegionGhana
| | - Emmanuel Ayitey Tagoe
- Department of Medical Laboratory SciencesSchool of Biomedical and Allied Health Sciences, University of GhanaAccraGreater Accra RegionGhana
| | - Elijah Paintsil
- Department of PediatricsBoston University Chobanian & Avedisian School of MedicineBostonUSA
| | - Kwasi Torpey
- Department of Population, Family and Reproductive HealthSchool of Public Health, University of GhanaAccraGreater Accra RegionGhana
| | - Osbourne Quaye
- West African Center for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular BiologyUniversity of GhanaAccraGreater Accra RegionGhana
| |
Collapse
|
2
|
Guan F, Wang R, Yi Z, Luo P, Liu W, Xie Y, Liu Z, Xia Z, Zhang H, Cheng Q. Tissue macrophages: origin, heterogenity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther 2025; 10:93. [PMID: 40055311 PMCID: PMC11889221 DOI: 10.1038/s41392-025-02124-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/01/2024] [Accepted: 12/15/2024] [Indexed: 05/04/2025] Open
Abstract
Macrophages are immune cells belonging to the mononuclear phagocyte system. They play crucial roles in immune defense, surveillance, and homeostasis. This review systematically discusses the types of hematopoietic progenitors that give rise to macrophages, including primitive hematopoietic progenitors, erythro-myeloid progenitors, and hematopoietic stem cells. These progenitors have distinct genetic backgrounds and developmental processes. Accordingly, macrophages exhibit complex and diverse functions in the body, including phagocytosis and clearance of cellular debris, antigen presentation, and immune response, regulation of inflammation and cytokine production, tissue remodeling and repair, and multi-level regulatory signaling pathways/crosstalk involved in homeostasis and physiology. Besides, tumor-associated macrophages are a key component of the TME, exhibiting both anti-tumor and pro-tumor properties. Furthermore, the functional status of macrophages is closely linked to the development of various diseases, including cancer, autoimmune disorders, cardiovascular disease, neurodegenerative diseases, metabolic conditions, and trauma. Targeting macrophages has emerged as a promising therapeutic strategy in these contexts. Clinical trials of macrophage-based targeted drugs, macrophage-based immunotherapies, and nanoparticle-based therapy were comprehensively summarized. Potential challenges and future directions in targeting macrophages have also been discussed. Overall, our review highlights the significance of this versatile immune cell in human health and disease, which is expected to inform future research and clinical practice.
Collapse
Affiliation(s)
- Fan Guan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruixuan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wanyao Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yao Xie
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
3
|
Tugizov S. HIV-1 Tat-induced disruption of epithelial junctions and epithelial-mesenchymal transition of oral and genital epithelial cells lead to increased invasiveness of neoplastic cells and the spread of herpes simplex virus and cytomegalovirus. Front Immunol 2025; 16:1541532. [PMID: 40018040 PMCID: PMC11866325 DOI: 10.3389/fimmu.2025.1541532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 01/28/2025] [Indexed: 03/01/2025] Open
Abstract
Human immunodeficiency virus (HIV-1) transactivator Tat is a unique multi-functional viral protein secreted by infected cells. Although its primary function is to promote HIV-1 transcription, secreted Tat interacts with neighboring cells and induces numerous disease-associated pathological changes. Despite the substantial reduction of viral load and disease burden, Tat expression and secretion persist in people living with HIV who are undergoing treatment with highly effective combination antiretroviral therapy (cART). Tat interacts with both oral and genital epithelial cells and impairs their mucosal barrier functions, which facilitates the entry of other pathogenic viruses. Tat-mediated interactions with both human papillomavirus (HPV) -infected and HPV-negative neoplastic epithelial cells lead to epithelial-mesenchymal transition and increased invasiveness of malignant cells. Likewise, Tat-induced disruption of oral epithelial cell junctions leads to herpes simplex virus-1 (HSV-1) infection and spread via exposure of its receptor, nectin-1. HIV-1 Tat facilitates infection and spread of human cytomegalovirus (HCMV) by activating mitogen-activated protein kinases (MAPK) and promoting NF-κB signaling, both critical for the replication and production of progeny virions. HIV extracellular Tat also plays a critical role in human herpesvirus 8 (HHV8) -caused Kaposi sarcoma (KS) pathogenesis by synergizing with HHV-8 lytic proteins and promoting the proliferation, angiogenesis, and migration of endothelial cells. Collectively, these findings emphasize the critical impact of HIV-1 Tat on HIV/AIDS pathogenesis during the cART era and highlight the need for further research on the molecular mechanisms underlying Tat-mediated interactions with oral and genital mucosal epithelial cells.
Collapse
Affiliation(s)
- Sharof Tugizov
- Department of Medicine, School of Medicine, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
4
|
Chung NPY, Cheng CY. Testis Is a Sanctuary Site for HIV-1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1469:433-440. [PMID: 40301268 DOI: 10.1007/978-3-031-82990-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
In this review, we summarize some recent findings regarding the likely mechanisms by which HIV-1 uses the testis as a sanctuary site for evading the effects of antiviral drugs from reaching the testis, due to the presence of the blood-testis barrier. This discussion also sheds insights into the possibility of eradicating the small viral pool in the testis behind the blood-testis barrier in future functional studies. These findings also bridge the knowledge gap of eradicating viral particles hiding behind the blood-brain barrier in the brain since it is anticipated that the blood-tissue barriers, namely the blood-testis and the blood-brain barriers, are utilizing similar mechanisms to regulate the dynamic nature of their tight junctions.
Collapse
Affiliation(s)
- Nancy P Y Chung
- Department of Biological Sciences, New York City College of Technology, City University of New York, New York, NY, USA
| | - C Yan Cheng
- Department of Biology, St Francis College, New York, NY, USA
| |
Collapse
|
5
|
Keane AM, Swartz TH. The impacts of tobacco and nicotine on HIV-1 infection, inflammation, and the blood-brain barrier in the central nervous system. Front Pharmacol 2024; 15:1477845. [PMID: 39529883 PMCID: PMC11550980 DOI: 10.3389/fphar.2024.1477845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Human immunodeficiency virus (HIV-1) remains a persistent global health crisis. Even while successfully virologically suppressed, people with HIV (PWH) experience a higher risk for inflammatory disorders such as HIV-associated neurocognitive disorder (HAND). Tobacco use puts PWH at higher risk for neurocognitive symptoms resulting from HIV-associated neuroinflammation. The NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome has been implicated as a driver of HIV-associated inflammation, including HAND. Nicotine, the psychoactive component of tobacco smoke, has also been shown to signal through the NLRP3 inflammasome and modulate inflammatory signaling in the CNS. Here, we explore the impacts of nicotine and tobacco on the complex neurobiology of HAND, including effects on cognition, inflammation, viral latency, and blood-brain barrier integrity. We outline nicotine's role in the establishment of active and latent infection in the brain and posit the NLRP3 inflammasome as a common pathway by which HIV-1 and nicotine promote neuroinflammation in PWH.
Collapse
Affiliation(s)
- Aislinn M. Keane
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Talia H. Swartz
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
6
|
Jones AM, Rademeyer KM, Rosen EP, Contaifer S, Wijesinghe D, Hauser KF, McRae M. Examining the effects of the HIV-1 protein Tat and morphine on antiretroviral accumulation and distribution within the brain. Clin Transl Sci 2024; 17:e70035. [PMID: 39382215 PMCID: PMC11462598 DOI: 10.1111/cts.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024] Open
Abstract
Despite combination antiretroviral therapy effectively suppressing HIV within the periphery, neuro-acquired HIV (neuroHIV) remains a significant problem and approximately half of people living with HIV will experience HIV-associated neurocognitive disorders (HAND). Concurrent opioid use exacerbates neuroHIV by promoting neuroinflammation, neuronal injury and synaptodendritic culling, viral replication, and potentially altering antiretroviral concentrations within the brain. The present study examined the effects of HIV and morphine co-exposure on the accumulation and spatial distribution of antiretroviral drugs across multiple regions within the brain in an HIV-1 Tat transgenic mouse model by using infrared-matrix-assisted laser desorption electrospray ionization mass spectrometry imaging (IR-MALDESI MSI). Morphine exposure uniquely decreased antiretroviral concentrations in anterior cerebral (primary motor and somatosensory) cortices, corpus collosum (anterior forceps), caudoputamen, nucleus accumbens, and posterior regions including the hippocampus, corpus callosum (main body), cerebral cortex (somatosensory and auditory cortices), thalamus, and hypothalamus. Interestingly, male mice experienced greater morphine-associated decreases in antiretroviral concentrations than females. The study also assessed whether changes in antiretroviral concentrations were linked with inflammation in astroglia, assessed through the measurement of astroglial activation using glial fibrillary acidic protein (GFAP) as a marker. Alterations in antiretroviral concentrations co-registering with areas of astroglial activation exhibited sex-specific treatment differences. This study highlights the intricate interplay between HIV, opioids, and antiretroviral drugs within the CNS, elucidating distinct regional and sex variations in responsiveness. Our findings emphasize the identification of vulnerabilities within the neural landscape and underscore the necessity of carefully monitoring opioid use to maintain the efficacy of antiretroviral therapies.
Collapse
Affiliation(s)
- Austin M. Jones
- Department of Pharmacotherapy and Outcomes Science, School of PharmacyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Kara M. Rademeyer
- Department of Pharmacotherapy and Outcomes Science, School of PharmacyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Elias P. Rosen
- Division of Pharmacotherapy and Experimental TherapeuticsUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Silas Contaifer
- Department of Pharmacotherapy and Outcomes Science, School of PharmacyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Dayanjan Wijesinghe
- Department of Pharmacotherapy and Outcomes Science, School of PharmacyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Kurt F. Hauser
- Department of Pharmacology and Toxicology, School of MedicineVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of Anatomy and Neurobiology, School of MedicineVirginia Commonwealth UniversityRichmondVirginiaUSA
- Institute for Drug and Alcohol StudiesVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - MaryPeace McRae
- Department of Pharmacotherapy and Outcomes Science, School of PharmacyVirginia Commonwealth UniversityRichmondVirginiaUSA
| |
Collapse
|
7
|
Cao Q, Zeng W, Nie J, Ye Y, Chen Y. The protective effects of apelin-13 in HIV-1 tat- induced macrophage infiltration and BBB impairment. Tissue Barriers 2024:2392361. [PMID: 39264117 DOI: 10.1080/21688370.2024.2392361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/05/2024] [Accepted: 08/11/2024] [Indexed: 09/13/2024] Open
Abstract
Impairment of the blood - brain barrier (BBB) and subsequent inflammatory responses contribute to the development of human immunodeficiency virus (HIV)-1-associated neurocognitive disorders (HAND). Apelin-13, the most abundant member of the apelin family, acts as the ligand of the angiotensin receptor-like 1 (APJ). However, its pharmacological function in HAND and its underlying mechanism are unknown. In the current study, we report that the presence of HIV-1 Tat reduced the levels of Apelin-13 and APJ in the cortex tissue of mice. Importantly, Apelin-13 preserved BBB integrity against HIV-1 Tat in mice by increasing the expression of the tight junction protein zonula occludens-1 (ZO-1) and occludin. Interestingly, increased macrophage infiltration, indicated by elevated CD68-positive staining was observed in the cortex after stimulation with HIV-1, which was mitigated by the administration of Apelin-13. Correspondingly, Apelin-13 reduced the expression of monocyte chemoattractant protein-1; (MCP-1). An in vitro two-chamber and two-cell trans-well assay demonstrated that HIV-1 Tat challenge significantly promoted macrophage migration, which was notably attenuated by the introduction of Apelin-13. Accordingly, treatment with Apelin-13 restored the HIV-1 Tat-induced reduction of occludin and ZO-1, while preventing the upregulation of MCP-1 in human brain microvascular endothelial cells (HBMVECs). Our results suggest that Apelin-13 may reduce macrophage infiltration into brain tissues and mitigate BBB dysfunction in patients with HAND.
Collapse
Affiliation(s)
- Qi Cao
- First Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Wei Zeng
- Department of Emergency, Chongqing Public Health Medical Center, Chongqing, China
| | - Jingmin Nie
- First Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Yongjun Ye
- Department of General Surgery, Chongqing Public Health Medical Center, Chongqing, China
| | - Yanchao Chen
- Department of General Internal Medicine, Chongqing Public Health Medical Center, Chongqing, China
| |
Collapse
|
8
|
Annadurai N, Kanmogne GD. Structural and Functional Dysregulation of the Brain Endothelium in HIV Infection and Substance Abuse. Cells 2024; 13:1415. [PMID: 39272987 PMCID: PMC11393916 DOI: 10.3390/cells13171415] [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: 06/19/2024] [Revised: 08/12/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Blood-brain barrier (BBB) injury and dysfunction following infection with the human immunodeficiency virus (HIV) enables viral entry into the brain, infection of resident brain cells, neuronal injury and subsequent neurodegeneration leading to HIV-associated neurocognitive disorders (HAND). Although combination antiretroviral therapy has significantly reduced the incidence and prevalence of acquired immunodeficiency syndrome and increased the life expectancy of people living with HIV, the prevalence of HAND remains high. With aging of people living with HIV associated with increased comorbidities, the prevalence of HIV-related central nervous system (CNS) complications is expected to remain high. Considering the principal role of the brain endothelium in HIV infection of the CNS and HAND, the purpose of this manuscript is to review the current literature on the pathobiology of the brain endothelium structural and functional dysregulation in HIV infection, including in the presence of HIV-1 and viral proteins (gp120, Tat, Nef, and Vpr). We summarize evidence from human and animal studies, in vitro studies, and associated mechanisms. We further summarize evidence of synergy or lack thereof between commonly abused substances (cocaine, methamphetamine, alcohol, tobacco, opioids, and cannabinoids) and HIV- or viral protein-induced BBB injury and dysfunction.
Collapse
Affiliation(s)
| | - Georgette D. Kanmogne
- Department of Anesthesiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-4455, USA;
| |
Collapse
|
9
|
Galpayage Dona KNU, Benmassaoud MM, Gipson CD, McLaughlin JP, Ramirez SH, Andrews AM. Something to talk about; crosstalk disruption at the neurovascular unit during HIV infection of the CNS. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2024; 3:97-111. [PMID: 39958876 PMCID: PMC11823645 DOI: 10.1515/nipt-2024-0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/11/2024] [Indexed: 02/18/2025]
Abstract
Although treatable with antiretroviral therapy, HIV infection persists in people living with HIV (PLWH). It is well known that the HIV virus finds refuge in places for which antiretroviral medications do not reach therapeutic levels, mainly the CNS. It is clear that as PLWH age, the likelihood of developing HIV-associated neurological deficits increases. At the biochemical level neurological dysfunction is the manifestation of altered cellular function and ineffective intercellular communication. In this review, we examine how intercellular signaling in the brain is disrupted in the context of HIV. Specifically, the concept of how the blood-brain barrier can be a convergence point for crosstalk, is explored. Crosstalk between the cells of the neurovascular unit (NVU) (endothelium, pericytes, astrocytes, microglia and neurons) is critical for maintaining proper brain function. In fact, the NVU allows for rapid matching of neuronal metabolic needs, regulation of blood-brain barrier (BBB) dynamics for nutrient transport and changes to the level of immunosurveillance. This review invites the reader to conceptually consider the BBB as a router or convergence point for NVU crosstalk, to facilitate a better understanding of the intricate signaling events that underpin the function of the NVU during HIV associated neuropathology.
Collapse
Affiliation(s)
- Kalpani N. Udeni Galpayage Dona
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Mohammed M. Benmassaoud
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Cassandra D. Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Servio H. Ramirez
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Allison M. Andrews
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
10
|
Singh S, Deshetty UM, Ray S, Oladapo A, Horanieh E, Buch S, Periyasamy P. Non-Coding RNAs in HIV Infection, NeuroHIV, and Related Comorbidities. Cells 2024; 13:898. [PMID: 38891030 PMCID: PMC11171711 DOI: 10.3390/cells13110898] [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: 04/17/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
NeuroHIV affects approximately 30-60% of people living with HIV-1 (PLWH) and is characterized by varying degrees of cognitive impairments, presenting a multifaceted challenge, the underlying cause of which is chronic, low-level neuroinflammation. Such smoldering neuroinflammation is likely an outcome of lifelong reliance on antiretrovirals coupled with residual virus replication in the brains of PLWH. Despite advancements in antiretroviral therapeutics, our understanding of the molecular mechanism(s) driving inflammatory processes in the brain remains limited. Recent times have seen the emergence of non-coding RNAs (ncRNAs) as critical regulators of gene expression, underlying the neuroinflammatory processes in HIV infection, NeuroHIV, and their associated comorbidities. This review explores the role of various classes of ncRNAs and their regulatory functions implicated in HIV infection, neuropathogenesis, and related conditions. The dysregulated expression of ncRNAs is known to exacerbate the neuroinflammatory responses, thus contributing to neurocognitive impairments in PLWH. This review also discusses the diagnostic and therapeutic potential of ncRNAs in HIV infection and its comorbidities, suggesting their utility as non-invasive biomarkers and targets for modulating neuroinflammatory pathways. Understanding these regulatory roles could pave the way for novel diagnostic strategies and therapeutic interventions in the context of HIV and its comorbidities.
Collapse
Affiliation(s)
| | | | | | | | | | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (S.S.); (U.M.D.); (S.R.); (A.O.); (E.H.)
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (S.S.); (U.M.D.); (S.R.); (A.O.); (E.H.)
| |
Collapse
|
11
|
Rademeyer KM, R Nass S, Jones AM, Ohene-Nyako M, Hauser KF, McRae M. Fentanyl dysregulates neuroinflammation and disrupts blood-brain barrier integrity in HIV-1 Tat transgenic mice. J Neurovirol 2024; 30:1-21. [PMID: 38280928 PMCID: PMC11232468 DOI: 10.1007/s13365-023-01186-4] [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: 06/17/2023] [Revised: 10/29/2023] [Accepted: 11/16/2023] [Indexed: 01/29/2024]
Abstract
Opioid overdose deaths have dramatically increased by 781% from 1999 to 2021. In the setting of HIV, opioid drug abuse exacerbates neurotoxic effects of HIV in the brain, as opioids enhance viral replication, promote neuronal dysfunction and injury, and dysregulate an already compromised inflammatory response. Despite the rise in fentanyl abuse and the close association between opioid abuse and HIV infection, the interactive comorbidity between fentanyl abuse and HIV has yet to be examined in vivo. The HIV-1 Tat-transgenic mouse model was used to understand the interactive effects between fentanyl and HIV. Tat is an essential protein produced during HIV that drives the transcription of new virions and exerts neurotoxic effects within the brain. The Tat-transgenic mouse model uses a glial fibrillary acidic protein (GFAP)-driven tetracycline promoter which limits Tat production to the brain and this model is well used for examining mechanisms related to neuroHIV. After 7 days of fentanyl exposure, brains were harvested. Tight junction proteins, the vascular cell adhesion molecule, and platelet-derived growth factor receptor-β were measured to examine the integrity of the blood brain barrier. The immune response was assessed using a mouse-specific multiplex chemokine assay. For the first time in vivo, we demonstrate that fentanyl by itself can severely disrupt the blood-brain barrier and dysregulate the immune response. In addition, we reveal associations between inflammatory markers and tight junction proteins at the blood-brain barrier.
Collapse
Affiliation(s)
- Kara M Rademeyer
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, 23298, U.S.A
| | - Sara R Nass
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - Austin M Jones
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, 23298, U.S.A
| | - Michael Ohene-Nyako
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - MaryPeace McRae
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, 22908, U.S.A..
| |
Collapse
|
12
|
Li Z, Wang H, Yin Y. Peripheral inflammation is a potential etiological factor in Alzheimer's disease. Rev Neurosci 2024; 35:99-120. [PMID: 37602685 DOI: 10.1515/revneuro-2023-0049] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023]
Abstract
Peripheral inflammation could constitute a risk factor for AD. This review summarizes the research related to peripheral inflammation that appears to have a relationship with Alzheimer's disease. We find there are significant associations between AD and peripheral infection induced by various pathogens, including herpes simplex virus type 1, cytomegalovirus, Epstein-Barr virus, human immunodeficiency virus, severe acute respiratory syndrome coronavirus 2, Porphyromonas gingivalis, Helicobacter pylori, and Toxoplasma gondii. Chronic inflammatory diseases are also reported to contribute to the pathophysiology of AD. The mechanisms by which peripheral inflammation affects the pathophysiology of AD are complex. Pathogen-derived neurotoxic molecule composition, disrupted BBB, and dysfunctional neurogenesis may all play a role in peripheral inflammation, promoting the development of AD. Anti-pathogenic medications and anti-inflammatory treatments are reported to decrease the risk of AD. Studies that could improve understanding the associations between AD and peripheral inflammation are needed. If our assumption is correct, early intervention against inflammation may be a potential method of preventing and treating AD.
Collapse
Affiliation(s)
- Ziyuan Li
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Yangpu District, Shanghai 200092, China
| | - Hui Wang
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Yangpu District, Shanghai 200092, China
| | - Yafu Yin
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Yangpu District, Shanghai 200092, China
| |
Collapse
|
13
|
Guo P, Lu Q, Hu S, Yang Y, Wang X, Yang X, Wang X. Daucosterol confers protection against T-2 toxin induced blood-brain barrier toxicity through the PGC-1α-mediated defensive response in vitro and in vivo. JOURNAL OF HAZARDOUS MATERIALS 2023; 459:132262. [PMID: 37604032 DOI: 10.1016/j.jhazmat.2023.132262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/28/2023] [Accepted: 08/08/2023] [Indexed: 08/23/2023]
Abstract
T-2 toxin is a common environmental pollutant and contaminant in food and animal feed that represents a great challenge to human and animal' health throughout the world. Using natural compounds to prevent the detrimental effects of T-2 toxin represents an attractive strategy. Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) is a critical regulator in various cellular processes. Recently, PGC-1α activation has been reported to confer protection against neurological injuries. We aimed to identify a potent PGC-1α activator from plants as a chemopreventive compound and to demonstrate the efficacy of the compound in attenuating T-2 toxin-induced blood-brain barrier (BBB) toxicity. We identified daucosterol, which binds directly to the 71-74 (-1100 to -1000 bp) position of the second promoter of human PGC-1α by hydrogen bonding. An in vitro and in vivo T-2 toxin induced BBB injury model revealed that this compound can protect against this injury by increasing transepithelial/transendothelial electrical resistance, reducing sodium fluorescein (NaF) infiltration and increasing the expression of tight junction-related proteins (zonula occludens-1 (ZO-1), occludin (OCLN), claudin-5 (CLDN5)) expression. In conclusion, we identified daucosterol as representing a novel of PGC-1α activators and illustrated the mechanism of specific binding site. Furthermore, we have demonstrated the feasibility of using natural compounds targeting PGC-1α as a therapeutic approach to protect humans from environmental insults that may occur daily such as lipopolysaccharide.
Collapse
Affiliation(s)
- Pu Guo
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
| | - Qirong Lu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
| | - Siyi Hu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yaqin Yang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xinru Wang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xinzhou Yang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, Hubei 430070, China.
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| |
Collapse
|
14
|
Namba MD, Xie Q, Barker JM. Advancing the preclinical study of comorbid neuroHIV and substance use disorders: Current perspectives and future directions. Brain Behav Immun 2023; 113:453-475. [PMID: 37567486 PMCID: PMC10528352 DOI: 10.1016/j.bbi.2023.07.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/23/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
Human immunodeficiency virus (HIV) remains a persistent public health concern throughout the world. Substance use disorders (SUDs) are a common comorbidity that can worsen treatment outcomes for people living with HIV. The relationship between HIV infection and SUD outcomes is likely bidirectional, making clear interrogation of neurobehavioral outcomes challenging in clinical populations. Importantly, the mechanisms through which HIV and addictive drugs disrupt homeostatic immune and CNS function appear to be highly overlapping and synergistic within HIV-susceptible reward and motivation circuitry in the central nervous system. Decades of animal research have revealed invaluable insights into mechanisms underlying the pathophysiology SUDs and HIV, although translational studies examining comorbid SUDs and HIV are very limited due to the technical challenges of modeling HIV infection preclinically. In this review, we discuss preclinical animal models of HIV and highlight key pathophysiological characteristics of each model, with a particular emphasis on rodent models of HIV. We then review the implementation of these models in preclinical SUD research and identify key gaps in knowledge in the field. Finally, we discuss how cutting-edge behavioral neuroscience tools, which have revealed key insights into the neurobehavioral mechanisms of SUDs, can be applied to preclinical animal models of HIV to reveal potential, novel treatment avenues for comorbid HIV and SUDs. Here, we argue that future preclinical SUD research would benefit from incorporating comorbidities such as HIV into animal models and would facilitate the discovery of more refined, subpopulation-specific mechanisms and effective SUD prevention and treatment targets.
Collapse
Affiliation(s)
- Mark D Namba
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Qiaowei Xie
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Jacqueline M Barker
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Lark ARS, Silva LK, Nass SR, Marone MG, Ohene-Nyako M, Ihrig TM, Marks WD, Yarotskyy V, Rory McQuiston A, Knapp PE, Hauser KF. Progressive Degeneration and Adaptive Excitability in Dopamine D1 and D2 Receptor-Expressing Striatal Neurons Exposed to HIV-1 Tat and Morphine. Cell Mol Neurobiol 2023; 43:1105-1127. [PMID: 35695980 PMCID: PMC9976699 DOI: 10.1007/s10571-022-01232-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/10/2022] [Indexed: 11/03/2022]
Abstract
The striatum is especially vulnerable to HIV-1 infection, with medium spiny neurons (MSNs) exhibiting marked synaptodendritic damage that can be exacerbated by opioid use disorder. Despite known structural defects in MSNs co-exposed to HIV-1 Tat and opioids, the pathophysiological sequelae of sustained HIV-1 exposure and acute comorbid effects of opioids on dopamine D1 and D2 receptor-expressing (D1 and D2) MSNs are unknown. To address this question, Drd1-tdTomato- or Drd2-eGFP-expressing reporter and conditional HIV-1 Tat transgenic mice were interbred. MSNs in ex vivo slices from male mice were assessed by whole-cell patch-clamp electrophysiology and filled with biocytin to explore the functional and structural effects of progressive Tat and acute morphine exposure. Although the excitability of both D1 and D2 MSNs increased following 48 h of Tat exposure, D1 MSN firing rates decreased below control (Tat-) levels following 2 weeks and 1 month of Tat exposure but returned to control levels after 2 months. D2 neurons continued to display Tat-dependent increases in excitability at 2 weeks, but also returned to control levels following 1 and 2 months of Tat induction. Acute morphine exposure increased D1 MSN excitability irrespective of the duration of Tat exposure, while D2 MSNs were variably affected. That D1 and D2 MSN excitability would return to control levels was unexpected since both subpopulations displayed significant synaptodendritic degeneration and pathologic phospho-tau-Thr205 accumulation following 2 months of Tat induction. Thus, despite frank morphologic damage, D1 and D2 MSNs uniquely adapt to sustained Tat and acute morphine insults.
Collapse
Affiliation(s)
- Arianna R S Lark
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
| | - Lindsay K Silva
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
- PPD®, Part of Thermo Fisher Scientific, Richmond, VA, 23230-3323, USA
| | - Sara R Nass
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
| | - Michael G Marone
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
| | - Michael Ohene-Nyako
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
| | - Therese M Ihrig
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
| | - William D Marks
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
- Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas, TX, 75235, USA
| | - Viktor Yarotskyy
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
| | - A Rory McQuiston
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, PO Box 980709, Richmond, VA, 23298-0709, USA
| | - Pamela E Knapp
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, PO Box 980709, Richmond, VA, 23298-0709, USA
- Institute for Drug and Alcohol Studies, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA.
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, PO Box 980709, Richmond, VA, 23298-0709, USA.
- Institute for Drug and Alcohol Studies, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| |
Collapse
|
16
|
Naranjo O, Torices S, Clifford PR, Daftari MT, Osborne OM, Fattakhov N, Toborek M. Pericyte infection by HIV-1: a fatal attraction. Retrovirology 2022; 19:27. [PMID: 36476484 PMCID: PMC9730689 DOI: 10.1186/s12977-022-00614-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
While HIV-1 is primarily an infection of CD4 + T cells, there is an emerging interest towards understanding how infection of other cell types can contribute to HIV-associated comorbidities. For HIV-1 to cross from the blood stream into tissues, the virus must come in direct contact with the vascular endothelium, including pericytes that envelope vascular endothelial cells. Pericytes are multifunctional cells that have been recognized for their essential role in angiogenesis, vessel maintenance, and blood flow rate. Most importantly, recent evidence has shown that pericytes can be a target of HIV-1 infection and support an active stage of the viral life cycle, with latency also suggested by in vitro data. Pericyte infection by HIV-1 has been confirmed in the postmortem human brains and in lungs from SIV-infected macaques. Moreover, pericyte dysfunction has been implicated in a variety of pathologies ranging from ischemic stroke to diabetes, which are common comorbidities among people with HIV-1. In this review, we discuss the role of pericytes during HIV-1 infection and their contribution to the progression of HIV-associated comorbidities.
Collapse
Affiliation(s)
- Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Paul R. Clifford
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Manav T. Daftari
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Olivia M. Osborne
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Nikolai Fattakhov
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| |
Collapse
|
17
|
Nühn MM, Gumbs SBH, Buchholtz NVEJ, Jannink LM, Gharu L, de Witte LD, Wensing AMJ, Lewin SR, Nijhuis M, Symons J. Shock and kill within the CNS: A promising HIV eradication approach? J Leukoc Biol 2022; 112:1297-1315. [PMID: 36148896 PMCID: PMC9826147 DOI: 10.1002/jlb.5vmr0122-046rrr] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 01/18/2023] Open
Abstract
The most studied HIV eradication approach is the "shock and kill" strategy, which aims to reactivate the latent reservoir by latency reversing agents (LRAs) and allowing elimination of these cells by immune-mediated clearance or viral cytopathic effects. The CNS is an anatomic compartment in which (persistent) HIV plays an important role in HIV-associated neurocognitive disorder. Restriction of the CNS by the blood-brain barrier is important for maintenance of homeostasis of the CNS microenvironment, which includes CNS-specific cell types, expression of transcription factors, and altered immune surveillance. Within the CNS predominantly myeloid cells such as microglia and perivascular macrophages are thought to be a reservoir of persistent HIV infection. Nevertheless, infection of T cells and astrocytes might also impact HIV infection in the CNS. Genetic adaptation to this microenvironment results in genetically distinct, compartmentalized viral populations with differences in transcription profiles. Because of these differences in transcription profiles, LRAs might have different effects within the CNS as compared with the periphery. Moreover, reactivation of HIV in the brain and elimination of cells within the CNS might be complex and could have detrimental consequences. Finally, independent of activity on latent HIV, LRAs themselves can have adverse neurologic effects. We provide an extensive overview of the current knowledge on compartmentalized (persistent) HIV infection in the CNS and on the "shock and kill" strategy. Subsequently, we reflect on the impact and promise of the "shock and kill" strategy on the elimination of persistent HIV in the CNS.
Collapse
Affiliation(s)
- Marieke M. Nühn
- Translational Virology, Department of Medical MicrobiologyUniversity Medical CenterUtrechtthe Netherlands
| | - Stephanie B. H. Gumbs
- Translational Virology, Department of Medical MicrobiologyUniversity Medical CenterUtrechtthe Netherlands
| | - Ninée V. E. J. Buchholtz
- Translational Virology, Department of Medical MicrobiologyUniversity Medical CenterUtrechtthe Netherlands
| | - Lisanne M. Jannink
- Translational Virology, Department of Medical MicrobiologyUniversity Medical CenterUtrechtthe Netherlands
| | - Lavina Gharu
- Translational Virology, Department of Medical MicrobiologyUniversity Medical CenterUtrechtthe Netherlands
| | - Lot D. de Witte
- Translational Virology, Department of Medical MicrobiologyUniversity Medical CenterUtrechtthe Netherlands,Department of PsychiatryIcahn School of MedicineNew YorkNew YorkUSA
| | - Annemarie M. J. Wensing
- Translational Virology, Department of Medical MicrobiologyUniversity Medical CenterUtrechtthe Netherlands
| | - Sharon R. Lewin
- Department of Infectious DiseasesThe University of Melbourne at the Peter Doherty Institute of Immunity and InfectionMelbourneVICAustralia,Victorian Infectious Diseases ServiceThe Royal Melbourne Hospital at the Peter Doherty Institute of Immunity and InfectionMelbourneVICAustralia,Department of Infectious DiseasesAlfred Hospital and Monash UniversityMelbourneVICAustralia
| | - Monique Nijhuis
- Translational Virology, Department of Medical MicrobiologyUniversity Medical CenterUtrechtthe Netherlands
| | - Jori Symons
- Translational Virology, Department of Medical MicrobiologyUniversity Medical CenterUtrechtthe Netherlands
| |
Collapse
|
18
|
Leibrand CR, Paris JJ, Jones AM, Ohene-Nyako M, Rademeyer KM, Nass SR, Kim WK, Knapp PE, Hauser KF, McRae M. Independent actions by HIV-1 Tat and morphine to increase recruitment of monocyte-derived macrophages into the brain in a region-specific manner. Neurosci Lett 2022; 788:136852. [PMID: 36028004 PMCID: PMC9845733 DOI: 10.1016/j.neulet.2022.136852] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 08/07/2022] [Accepted: 08/18/2022] [Indexed: 01/21/2023]
Abstract
Despite advances in the treatment of human immunodeficiency virus (HIV), approximately one-half of people infected with HIV (PWH) experience neurocognitive impairment. Opioid use disorder (OUD) can exacerbate the cognitive and pathological changes seen in PWH. HIV increases inflammation and immune cell trafficking into the brain; however, less is known about how opioid use disorder affects the recruitment of immune cells. Accordingly, we examined the temporal consequences of HIV-1 Tat and/or morphine on the recruitment of endocytic cells (predominantly perivascular macrophages and microglia) in the dorsal striatum and hippocampus by infusing multi-colored, fluorescently labeled dextrans before and after exposure. To address this question, transgenic mice that conditionally expressed HIV-1 Tat (Tat+), or their control counterparts (Tat-), received three sequential intracerebroventricular (i.c.v.) infusions of Cascade Blue-, Alexa Fluor 488-, and Alexa Fluor 594-labeled dextrans, respectively infused 1 day before, 1-day after, or 13-days after morphine and/or Tat exposure. At the end of the study, the number of cells labeled with each fluorescent dextran were counted. The data demonstrated a significantly higher influx of newly-labeled cells into the perivascular space than into the parenchyma. In the striatum, Tat or morphine exposure increased the number of endocytic cells in the perivascular space, while only morphine increased the recruitment of endocytic cells into the parenchyma. In the hippocampus, morphine (but not Tat) increased the influx of dextran-labeled cells into the perivascular space, but there were too few labeled cells within the hippocampal parenchyma to analyze. Collectively, these data suggest that HIV-1 Tat and morphine act independently to increase the recruitment of endocytic cells into the brain in a region-specific manner.
Collapse
Affiliation(s)
- Crystal R Leibrand
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Jason J Paris
- Department of BioMolecular Sciences, School of Pharmacy, The University of Mississippi, University, MS 38677, United States
| | - Austin M Jones
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Michael Ohene-Nyako
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Kara M Rademeyer
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Sara R Nass
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Woong-Ki Kim
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23507, United States
| | - Pamela E Knapp
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States; Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States; Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - MaryPeace McRae
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, United States.
| |
Collapse
|
19
|
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: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
|
20
|
Sharma AL, Wang H, Zhang Z, Millien G, Tyagi M, Hongpaisan J. HIV Promotes Neurocognitive Impairment by Damaging the Hippocampal Microvessels. Mol Neurobiol 2022; 59:4966-4986. [PMID: 35665894 PMCID: PMC10071835 DOI: 10.1007/s12035-022-02890-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/18/2022] [Indexed: 10/18/2022]
Abstract
Current evidence suggests that mild cerebrovascular changes could induce neurodegeneration and contribute to HIV-associated neurocognitive disease (HAND) in HIV patients. We investigated both the quantitative and qualitative impact of HIV infection on brain microvessels, especially on hippocampal microvessels, which are crucial for optimal O2 supply, and thus for maintaining memory and cognitive abilities. The results obtained using cultured human brain microvascular endothelial cells (HBMEC) were reproduced using a suitable mouse model and autopsied human HIV hippocampus. In HBMEC, we found significantly higher oxidative stress-dependent apoptotic cell loss following 5 h of treatment of GST-Tat (1 µg/ml) compared to GST (1 µg/ml) control. We noticed complete recovery of HBMEC cells after 24 h of GST-Tat treatment, due to temporal degradation or inactivation of GST-Tat. Interestingly, we found a sustained increase in mitochondrial oxidative DNA damage marker 8-OHdG, as well as an increase in hypoxia-inducible factor hypoxia-inducible factor-1α (HIF-1α). In our mouse studies, upon short-term injection of GST-Tat, we found the loss of small microvessels (mostly capillaries) and vascular endothelial growth factor (VEGF), but not large microvessels (arterioles and venules) in the hippocampus. In addition to capillary loss, in the post-mortem HIV-infected human hippocampus, we observed large microvessels with increased wall cells and perivascular tissue degeneration. Together, our data show a crucial role of Tat in inducing HIF-1α-dependent inhibition of mitochondrial transcriptional factor A (TFAM) and dilated perivascular space. Thus, our results further define the underlying molecular mechanism promoting mild cerebrovascular disease, neuropathy, and HAND pathogenesis in HIV patients.
Collapse
Affiliation(s)
- Adhikarimayum Lakhikumar Sharma
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA
| | - Huaixing Wang
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA
| | - Zongxiu Zhang
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA
| | - Guetchyn Millien
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA
| | - Mudit Tyagi
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA.
| | - Jarin Hongpaisan
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA.
| |
Collapse
|
21
|
Expression of Human Immunodeficiency Virus Transactivator of Transcription (HIV-Tat 1-86) Protein Alters Nociceptive Processing that is Sensitive to Anti-Oxidant and Anti-Inflammatory Interventions. J Neuroimmune Pharmacol 2022; 17:152-164. [PMID: 33619645 PMCID: PMC8380260 DOI: 10.1007/s11481-021-09985-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 01/24/2021] [Indexed: 12/29/2022]
Abstract
Despite the success of combined antiretroviral therapy (cART) in reducing viral load, a substantial portion of Human Immunodeficiency Virus (HIV)+ patients report chronic pain. The exact mechanism underlying this co-morbidity even with undetectable viral load remains unknown, but the transactivator of transcription (HIV-Tat) protein is of particular interest. Functional HIV-Tat protein is observed even in cerebrospinal fluid of patients who have an undetectable viral load. It is hypothesized that Tat protein exposure is sufficient to induce neuropathic pain-like manifestations via both activation of microglia and generation of oxidative stress. iTat mice conditionally expressed Tat(1-86) protein in the central nervous system upon daily administration of doxycycline (100 mg/kg/d, i.p., up to 14 days). The effect of HIV-Tat protein exposure on the well-being of the animal was assessed using sucrose-evoked grooming and acute nesting behavior for pain-depressed behaviors, and the development of hyperalgesia assessed with warm-water tail-withdrawal and von Frey assays for thermal hyperalgesia and mechanical allodynia, respectively. Tissue harvested at select time points was used to assess ex vivo alterations in oxidative stress, astrocytosis and microgliosis, and blood-brain barrier integrity with assays utilizing fluorescence-based indicators. Tat protein induced mild thermal hyperalgesia but robust mechanical allodynia starting after 4 days of exposure, reaching a nadir after 7 days. Changes in nociceptive processing were associated with reduced sucrose-evoked grooming behavior without altering acute nesting behavior, and in spinal cord dysregulated free radical generation as measured by DCF fluorescence intensity, altered immunohistochemical expression of the gliotic markers, Iba-1 and GFAP, and increased permeability of the blood-brain barrier to the small molecule fluorescent tracer, sodium fluorescein, in a time-dependent manner. Pretreatment with the anti-inflammatory, indomethacin (1 mg/kg/d, i.p.), the antioxidant, methylsulfonylmethane (100 mg/kg/d i.p.), or the immunomodulatory agent, dimethylfumarate (100 mg/kg/d p.o.) thirty minutes prior to daily injections of doxycycline (100 mg/kg/d i.p.) over 7 days significantly attenuated the development of Tat-induced mechanical allodynia. Collectively, the data suggests that even acute exposure to HIV-1 Tat protein at pathologically relevant levels is sufficient to produce select neurophysiological and behavioral manifestations of chronic pain consistent with that reported by HIV-positive patients.
Collapse
|
22
|
Mori L, Valente ST. Cure and Long-Term Remission Strategies. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2407:391-428. [PMID: 34985678 DOI: 10.1007/978-1-0716-1871-4_26] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The majority of virally suppressed individuals will experience rapid viral rebound upon antiretroviral therapy (ART) interruption, providing a strong rationale for the development of cure strategies. Moreover, despite ART virological control, HIV infection is still associated with chronic immune activation, inflammation, comorbidities, and accelerated aging. These effects are believed to be due, in part, to low-grade persistent transcription and trickling production of viral proteins from the pool of latent proviruses constituting the viral reservoir. In recent years there has been an increasing interest in developing what has been termed a functional cure for HIV. This approach entails the long-term, durable control of viral expression in the absence of therapy, preventing disease progression and transmission, despite the presence of detectable integrated proviruses. One such strategy, the block-and-lock approach for a functional cure, proposes the epigenetic silencing of proviral expression, locking the virus in a profound latent state, from which reactivation is very unlikely. The proof-of-concept for this approach was demonstrated with the use of a specific small molecule targeting HIV transcription. Here we review the principles behind the block-and-lock approach and some of the additional strategies proposed to silence HIV expression.
Collapse
Affiliation(s)
- Luisa Mori
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL, USA
| | - Susana T Valente
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL, USA.
| |
Collapse
|
23
|
Wu S, Frank I, Derby N, Martinelli E, Cheng CY. HIV-1 Establishes a Sanctuary Site in the Testis by Permeating the BTB Through Changes in Cytoskeletal Organization. Endocrinology 2021; 162:6338140. [PMID: 34343260 PMCID: PMC8407494 DOI: 10.1210/endocr/bqab156] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Indexed: 11/19/2022]
Abstract
Studies suggest that HIV-1 invades the testis through initial permeation of the blood-testis barrier (BTB). The selectivity of the BTB to antiretroviral drugs makes this site a sanctuary for the virus. Little is known about how HIV-1 crosses the BTB and invades the testis. Herein, we used 2 approaches to examine the underlying mechanism(s) by which HIV-1 permeates the BTB and gains entry into the seminiferous epithelium. First, we examined if recombinant Tat protein was capable of perturbing the BTB and making the barrier leaky, using the primary rat Sertoli cell in vitro model that mimics the BTB in vivo. Second, we used HIV-1-infected Sup-T1 cells to investigate the activity of HIV-1 infection on cocultured Sertoli cells. Using both approaches, we found that the Sertoli cell tight junction permeability barrier was considerably perturbed and that HIV-1 effectively permeates the BTB by inducing actin-, microtubule-, vimentin-, and septin-based cytoskeletal changes in Sertoli cells. These studies suggest that HIV-1 directly perturbs BTB function, potentially through the activity of the Tat protein.
Collapse
Affiliation(s)
- Siwen Wu
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| | - Ines Frank
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| | - Nina Derby
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Elena Martinelli
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - C Yan Cheng
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| |
Collapse
|
24
|
Possible mechanisms of HIV neuro-infection in alcohol use: Interplay of oxidative stress, inflammation, and energy interruption. Alcohol 2021; 94:25-41. [PMID: 33864851 DOI: 10.1016/j.alcohol.2021.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/05/2021] [Accepted: 04/01/2021] [Indexed: 11/21/2022]
Abstract
Alcohol use and HIV-1 infection have a pervasive impact on brain function, which extends to the requirement, distribution, and utilization of energy within the central nervous system. This effect on neuroenergetics may explain, in part, the exacerbation of HIV-1 disease under the influence of alcohol, particularly the persistence of HIV-associated neurological complications. The objective of this review article is to highlight the possible mechanisms of HIV/AIDS progression in alcohol users from the perspective of oxidative stress, neuroinflammation, and interruption of energy metabolism. These include the hallmark of sustained immune cell activation and high metabolic energy demand by HIV-1-infected cells in the central nervous system, with at-risk alcohol use. Here, we discussed the point that the increase in energy supply requirement by HIV-1-infected neuroimmune cells as well as the deterrence of nutrient uptake across the blood-brain barrier significantly depletes the energy source and neuro-environment homeostasis in the CNS. We also described the mechanistic idea that comorbidity of HIV-1 infection and alcohol use can cause a metabolic shift and redistribution of energy usage toward HIV-1-infected neuroimmune cells, as shown in neuropathological evidence. Under such an imbalanced neuro-environment, meaningless energy waste is expected in infected cells, along with unnecessary malnutrition in non-infected neuronal cells, which is likely to accelerate HIV neuro-infection progression in alcohol use. Thus, it will be important to consider the factor of nutrients/energy imbalance in formulating treatment strategies to help impede the progression of HIV-1 disease and associated neurological disorders in alcohol use.
Collapse
|
25
|
Abstract
Long-term effective use of antiretroviral therapy (ART) among people with HIV (PWH) has significantly reduced the burden of disease, yet a cure for HIV has not been universally achieved, likely due to the persistence of an HIV reservoir. The central nervous system (CNS) is an understudied HIV sanctuary. Importantly, due to viral persistence in the brain, cognitive disturbances persist to various degrees at high rates in PWH despite suppressive ART. Given the complexity and accessibility of the CNS compartment and that it is a physiologically and anatomically unique immune site, human studies to reveal molecular mechanisms of viral entry, reservoir establishment, and the cellular and structural interactions leading to viral persistence and brain injury to advance a cure and either prevent or limit cognitive impairments in PWH remain challenging. Recent advances in human brain organoids show that they can mimic the intercellular dynamics of the human brain and may recapitulate many of the events involved in HIV infection of the brain (neuroHIV). Human brain organoids can be produced, spontaneously or with addition of growth factors and at immature or mature states, and have become stronger models to study neurovirulent viral infections of the CNS. While organoids provide opportunities to study neuroHIV, obstacles such as the need to incorporate microglia need to be overcome to fully utilize this model. Here, we review the current achievements in brain organoid biology and their relevance to neuroHIV research efforts.
Collapse
|
26
|
Li J, Huang J, He Y, Wang W, Leung CK, Zhang D, Zhang R, Wang S, Li Y, Liu L, Zeng X, Li Z. The protective effect of gastrodin against the synergistic effect of HIV-Tat protein and METH on the blood-brain barrier via glucose transporter 1 and glucose transporter 3. Toxicol Res (Camb) 2021; 10:91-101. [PMID: 33613977 DOI: 10.1093/toxres/tfaa102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/04/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022] Open
Abstract
Many individuals infected with human immunodeficiency virus (HIV) are also afflicted with HIV-associated neurocognitive disorders (HANDs). Methamphetamine (METH) abuse puts HIV-1 patients at risk for HANDs because METH and HIV-1 proteins, such as trans-activator of transcription (Tat), can synergistically damage the blood-brain barrier (BBB). However, the underlying mechanism of METH- and HIV-Tat-induced BBB damage remains unclear. In this study, male adult tree shrews and human brain capillary endothelial cells were treated with HIV-Tat, METH, and gastrodin. We used western blotting to examine the expressions of glucose transporters (GLUT1 and GLUT3), tight junctions, and junctional adhesion molecule A (JAMA) and to evaluate the damage and detect Evans blue (EB) and fluorescein sodium in the brain to assess BBB permeability to study the effect of METH and the HIV-1 Tat protein on BBB function in vitro and in vivo. The results showed that the group treated with Tat and METH experienced a significant change at the ultrastructural level of the tree shrew cerebral cortex, decreased protein levels of occluding, claudin-5, Zonula occludens 1 (ZO1), and JAMA in vitro and in vivo, and increased levels of EB and fluorescein sodium in the tree shrew cerebral cortex. The protein levels of GLUT1 and GLUT3 was downregulated in patients with Tat- and METH-induced BBB damage. Pretreatment with gastrodin significantly increased the levels of EB and fluorescein sodium in the tree shrew cerebral cortex and increased the expressions of occluding, ZO1, JAMA, and GLUT1 and GLUT. These results indicate that gastrodin may offer a potential therapeutic option for patients with HANDs.
Collapse
Affiliation(s)
- Juan Li
- School of Basic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China.,Center of Tree Shrew Germplasm Resources, Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Yunnan Innovation Team of Standardization and Application Research in Tree Shrew, Chinese Academy of Medical Science and Peking Union Medical College, 935 Jiaoling road,Yunnan 650531, China
| | - Jian Huang
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China.,School of Forensic Medicine, Southern Medical University, 1838 Guangzhou Dadao Bei, Baiyun District, Guangzhou 510515, Guangdong, China
| | - Yongwang He
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| | - Wenguang Wang
- Center of Tree Shrew Germplasm Resources, Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Yunnan Innovation Team of Standardization and Application Research in Tree Shrew, Chinese Academy of Medical Science and Peking Union Medical College, 935 Jiaoling road,Yunnan 650531, China
| | - Chi-Kwan Leung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.,CUHK-SDU Joint Laboratory of Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Dongxian Zhang
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| | - Ruilin Zhang
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| | - Shangwen Wang
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| | - Yuanyuan Li
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| | - Liu Liu
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| | - Xiaofeng Zeng
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| | - Zhen Li
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| |
Collapse
|
27
|
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: 25] [Impact Index Per Article: 5.0] [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
|
28
|
Bhattacharya A, Jha V, Singhal K, Fatima M, Singh D, Chaturvedi G, Dholakia D, Kutum R, Pandey R, Bakken TE, Seth P, Pillai B, Mukerji M. Multiple Alu Exonization in 3'UTR of a Primate-Specific Isoform of CYP20A1 Creates a Potential miRNA Sponge. Genome Biol Evol 2020; 13:5958120. [PMID: 33434274 PMCID: PMC7802813 DOI: 10.1093/gbe/evaa233] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2020] [Indexed: 12/13/2022] Open
Abstract
Alu repeats contribute to phylogenetic novelties in conserved regulatory networks in primates. Our study highlights how exonized Alus could nucleate large-scale mRNA-miRNA interactions. Using a functional genomics approach, we characterize a transcript isoform of an orphan gene, CYP20A1 (CYP20A1_Alu-LT) that has exonization of 23 Alus in its 3'UTR. CYP20A1_Alu-LT, confirmed by 3'RACE, is an outlier in length (9 kb 3'UTR) and widely expressed. Using publically available data sets, we demonstrate its expression in higher primates and presence in single nucleus RNA-seq of 15,928 human cortical neurons. miRanda predicts ∼4,700 miRNA recognition elements (MREs) for ∼1,000 miRNAs, primarily originated within these 3'UTR-Alus. CYP20A1_Alu-LT could be a potential multi-miRNA sponge as it harbors ≥10 MREs for 140 miRNAs and has cytosolic localization. We further tested whether expression of CYP20A1_Alu-LT correlates with mRNAs harboring similar MRE targets. RNA-seq with conjoint miRNA-seq analysis was done in primary human neurons where we observed CYP20A1_Alu-LT to be downregulated during heat shock response and upregulated in HIV1-Tat treatment. In total, 380 genes were positively correlated with its expression (significantly downregulated in heat shock and upregulated in Tat) and they harbored MREs for nine expressed miRNAs which were also enriched in CYP20A1_Alu-LT. MREs were significantly enriched in these 380 genes compared with random sets of differentially expressed genes (P = 8.134e-12). Gene ontology suggested involvement of these genes in neuronal development and hemostasis pathways thus proposing a novel component of Alu-miRNA-mediated transcriptional modulation that could govern specific physiological outcomes in higher primates.
Collapse
Affiliation(s)
- Aniket Bhattacharya
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Vineet Jha
- Persistent LABS, Persistent Systems Ltd., Pune, Maharashtra, India
| | - Khushboo Singhal
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Mahar Fatima
- Department of Molecular and Cellular Neuroscience, Neurovirology Section, National Brain Research Centre (NBRC), Manesar, Haryana, India
| | - Dayanidhi Singh
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Gaura Chaturvedi
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Dhwani Dholakia
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rintu Kutum
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rajesh Pandey
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | | | - Pankaj Seth
- Department of Molecular and Cellular Neuroscience, Neurovirology Section, National Brain Research Centre (NBRC), Manesar, Haryana, India
| | - Beena Pillai
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Mitali Mukerji
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
29
|
The Paradox of HIV Blood-Brain Barrier Penetrance and Antiretroviral Drug Delivery Deficiencies. Trends Neurosci 2020; 43:695-708. [PMID: 32682564 PMCID: PMC7483662 DOI: 10.1016/j.tins.2020.06.007] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 06/04/2020] [Accepted: 06/21/2020] [Indexed: 12/13/2022]
Abstract
HIV attacks the body's immune cells, frequently compromises the integrity of the blood-brain barrier (BBB), and infects the CNS in the early stages of infection. Dysfunction of the BBB further potentiates viral replication within the CNS, which can lead to HIV-associated neuropathology. Antiretroviral therapy (ART) significantly improves HIV patient outcomes and reduces mortality rates. However, there has been limited progress in targeting latent viral reservoirs within the CNS, which may eventually lead to rebound viremia. While ART drugs are shown to be effective in attenuating HIV replication in the periphery, the protection of the brain by the BBB offers an isolated sanctuary to harbor HIV and maintains chronic and persistent replication within the CNS. In this review, we elucidate the pathology of the BBB, its ability to potentiate viral replication, as well as current therapies and insufficiencies in treating HIV-infected individuals.
Collapse
|
30
|
Zhan X, Asmara H, Cheng N, Sahu G, Sanchez E, Zhang FX, Zamponi GW, Rho JM, Turner RW. FMRP(1-297)-tat restores ion channel and synaptic function in a model of Fragile X syndrome. Nat Commun 2020; 11:2755. [PMID: 32488011 PMCID: PMC7265297 DOI: 10.1038/s41467-020-16250-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 04/22/2020] [Indexed: 01/21/2023] Open
Abstract
Fragile X Syndrome results from a loss of Fragile X Mental Retardation Protein (FMRP). We now show that FMRP is a member of a Cav3-Kv4 ion channel complex that is known to regulate A-type potassium current in cerebellar granule cells to produce mossy fiber LTP. Mossy fiber LTP is absent in Fmr1 knockout (KO) mice but is restored by FMRP(1-297)-tat peptide. This peptide further rapidly permeates the blood-brain barrier to enter cells across the cerebellar-cortical axis that restores the balance of protein translation for at least 24 h and transiently reduces elevated levels of activity of adult Fmr1 KO mice in the Open Field Test. These data reveal that FMRP(1-297)-tat can improve function from the levels of protein translation to synaptic efficacy and behaviour in a model of Fragile X syndrome, identifying a potential therapeutic strategy for this genetic disorder.
Collapse
Affiliation(s)
- Xiaoqin Zhan
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Hadhimulya Asmara
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Ning Cheng
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Giriraj Sahu
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Eduardo Sanchez
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Fang-Xiong Zhang
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Gerald W Zamponi
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Jong M Rho
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Ray W Turner
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
31
|
Nass SR, Hahn YK, McLane VD, Varshneya NB, Damaj MI, Knapp PE, Hauser KF. Chronic HIV-1 Tat exposure alters anterior cingulate cortico-basal ganglia-thalamocortical synaptic circuitry, associated behavioral control, and immune regulation in male mice. Brain Behav Immun Health 2020; 5:100077. [PMID: 33083793 PMCID: PMC7571616 DOI: 10.1016/j.bbih.2020.100077] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 04/25/2020] [Indexed: 12/28/2022] Open
Abstract
HIV-1 selectively disrupts neuronal integrity within specific brain regions, reflecting differences in viral tropism and/or the regional differences in the vulnerability of distinct neuronal subpopulations within the CNS. Deficits in prefrontal cortex (PFC)-mediated executive function and the resultant loss of behavioral control are a particularly debilitating consequence of neuroHIV. To explore how HIV-1 disrupts executive function, we investigated the effects of 48 h, 2 and/or 8 weeks of HIV-1 Tat exposure on behavioral control, synaptic connectivity, and neuroimmune function in the anterior cingulate cortex (ACC) and associated cortico-basal ganglia (BG)-thalamocortical circuitry in adult, Tat transgenic male mice. HIV-1 Tat exposure increased novelty-exploration in response to novel food, flavor, and environmental stimuli, suggesting that Tat triggers increased novelty-exploration in situations of competing motivation (e.g., drive to feed or explore vs. fear of novel, brightly lit open areas). Furthermore, Tat induced adaptability in response to an environmental stressor and pre-attentive filtering deficits. The behavioral insufficiencies coincided with decreases in the inhibitory pre- and post-synaptic proteins, synaptotagmin 2 and gephyrin, respectively, in the ACC, and alterations in specific pro- and anti-inflammatory cytokines out of 23 assayed. The interaction of Tat exposure and the resultant time-dependent, selective alterations in CCL4, CXCL1, IL-12p40, and IL-17A levels in the PFC predicted significant decreases in adaptability. Tat decreased dendritic spine density and cortical VGLUT1 inputs, while increasing IL-1β, IL-6, CCL5, and CCL11 in the striatum. Alternatively, IL-1α, CCL5, and IL-13 were decreased in the mediodorsal thalamus despite the absence of synaptic changes. Thus, HIV-1 Tat appears to uniquely and systematically disrupt immune regulation and the inhibitory and excitatory synaptic balance throughout the ACC-BG-thalamocortical circuitry resulting in a loss of behavioral control.
Collapse
Affiliation(s)
- Sara R. Nass
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
| | - Yun K. Hahn
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980709, Richmond, VA, 23298-0709, USA
| | - Virginia D. McLane
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
| | - Neil B. Varshneya
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
| | - M. Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
| | - Pamela E. Knapp
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980709, Richmond, VA, 23298-0709, USA
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980059, Richmond, VA, 23298-0059, USA
| | - Kurt F. Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980709, Richmond, VA, 23298-0709, USA
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980059, Richmond, VA, 23298-0059, USA
| |
Collapse
|
32
|
Chiu CF, Chu LW, Liao IC, Simanjuntak Y, Lin YL, Juan CC, Ping YH. The Mechanism of the Zika Virus Crossing the Placental Barrier and the Blood-Brain Barrier. Front Microbiol 2020; 11:214. [PMID: 32153526 PMCID: PMC7044130 DOI: 10.3389/fmicb.2020.00214] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/30/2020] [Indexed: 12/17/2022] Open
Abstract
Zika virus (ZIKV) infection causes severe neurological symptoms in adults and fetal microcephaly and the virus is detected in the brain of microcephaly and meningoencephalitis patient. However, the mechanism of ZIKV crossing the physiological barrier to the central nervous systems (CNS) remains elusive. The placental barrier and the blood brain barrier (BBB) protect the fetus from pathogens and ensure healthy brain development during pregnancy. In this study, we used human placenta trophoblasts cells (JEG-3) and human brain-derived endothelial cells (hCMEC/D3) as in vitro models of the physiological barriers. Results showed that ZIKV could infect JEG-3 cells effectively and reduce the amounts of ZO-1 and occludin between adjacent cells by the proteasomal degradation pathway, suggesting that the permeability of the barrier differentially changed in response to ZIKV infection, allowing the virus particle to cross the host barrier. In contrast, ZIKV could infect hCMEC/D3 cells without disrupting the BBB barrier permeability and tight junction protein expression. Although no disruption to the BBB was observed during ZIKV infection, ZIKV particles were released on the basal side of the BBB model and infected underlying cells. In addition, we observed that fluorescence-labeled ZIKV particles could cross the in vitro placenta barrier and BBB model by transcytosis and the action of transcytosis could be blocked by either low temperature or pharmacological inhibitors of endocytosis. In summary, the ZIKV uses a cell-type specific paracellular pathway to cross the placenta monolayer barrier by disrupting cellular tight junction. In addition, the ZIKV can also cross both the placenta barrier and the BBB by transcytosis. Our study provided new insights into on the mechanism of the cellular barrier penetration of ZIKV particles.
Collapse
Affiliation(s)
- Chi-Fen Chiu
- Department and Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Li-Wei Chu
- Department and Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - I-Chen Liao
- Department and Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Yogy Simanjuntak
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Ling Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chi-Chang Juan
- Department and Institute of Physiology, National Yang-Ming University, Taipei, Taiwan
| | - Yueh-Hsin Ping
- Department and Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
- Institute of Biophotonics, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
33
|
Chilunda V, Calderon TM, Martinez-Aguado P, Berman JW. The impact of substance abuse on HIV-mediated neuropathogenesis in the current ART era. Brain Res 2019; 1724:146426. [PMID: 31473221 PMCID: PMC6889827 DOI: 10.1016/j.brainres.2019.146426] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/16/2019] [Accepted: 08/28/2019] [Indexed: 12/21/2022]
Abstract
Approximately 37 million people worldwide are infected with human immunodeficiency virus (HIV). One highly significant complication of HIV infection is the development of HIV-associated neurocognitive disorders (HAND) in 15-55% of people living with HIV (PLWH), that persists even in the antiretroviral therapy (ART) era. The entry of HIV into the central nervous system (CNS) occurs within 4-8 days after peripheral infection. This establishes viral reservoirs that may persist even in the presence of ART. Once in the CNS, HIV infects resident macrophages, microglia, and at low levels, astrocytes. In response to chronic infection and cell activation within the CNS, viral proteins, inflammatory mediators, and host and viral neurotoxic factors produced over extended periods of time result in neuronal injury and loss, cognitive deficits and HAND. Substance abuse is a common comorbidity in PLWH and has been shown to increase neuroinflammation and cognitive disorders. Additionally, it has been associated with poor ART adherence, and increased viral load in the cerebrospinal fluid (CSF), that may also contribute to increased neuroinflammation and neuronal injury. Studies have examined mechanisms that contribute to neuroinflammation and neuronal damage in PLWH, and how substances of abuse exacerbate these effects. This review will focus on how substances of abuse, with an emphasis on methamphetamine (meth), cocaine, and opioids, impact blood brain barrier (BBB) integrity and transmigration of HIV-infected and uninfected monocytes across the BBB, as well as their effects on monocytes/macrophages, microglia, and astrocytes within the CNS. We will also address how these substances of abuse may contribute to HIV-mediated neuropathogenesis in the context of suppressive ART. Additionally, we will review the effects of extracellular dopamine, a neurotransmitter that is increased in the CNS by substances of abuse, on HIV neuropathogenesis and how this may contribute to neuroinflammation, neuronal insult, and HAND in PLWH with active substance use. Lastly, we will discuss some potential therapies to limit CNS inflammation and damage in HIV-infected substance abusers.
Collapse
Affiliation(s)
- Vanessa Chilunda
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, USA
| | - Tina M Calderon
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, USA
| | - Pablo Martinez-Aguado
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, USA
| | - Joan W Berman
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, USA; Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, USA.
| |
Collapse
|
34
|
Yang T, Guo R, Zhang F. Brain perivascular macrophages: Recent advances and implications in health and diseases. CNS Neurosci Ther 2019; 25:1318-1328. [PMID: 31749316 PMCID: PMC7154594 DOI: 10.1111/cns.13263] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/20/2022] Open
Abstract
Brain perivascular macrophages (PVMs) belong to a distinct population of brain‐resident myeloid cells located within the perivascular space surrounding arterioles and venules. Their characterization depends on the combination of anatomical localization, phagocytic capacity, and molecular markers. Under physiological status, they provide structural and functional support for maintaining brain homeostasis, including facilitation of blood‐brain barrier integrity and lymphatic drainage, and exertion of immune functions such as phagocytosis and antigen presentation. Increasing evidence also implicates their specific roles in diseased brain, ranging from cerebrovascular diseases, Aβ pathologies, infections, and autoimmunity. Collectively, PVMs are key components of the brain‐resident immune system, actively participate in a broad‐spectrum of processes in normal and diseased status. Details of the processes are largely underexplored. Targeting PVMs would lead to new insights and be a promising strategy for a broad array of human diseases.
Collapse
Affiliation(s)
- Tuo Yang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ruiming Guo
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Feng Zhang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
35
|
Bonney S, Seitz S, Ryan CA, Jones KL, Clarke P, Tyler KL, Siegenthaler JA. Gamma Interferon Alters Junctional Integrity via Rho Kinase, Resulting in Blood-Brain Barrier Leakage in Experimental Viral Encephalitis. mBio 2019; 10:e01675-19. [PMID: 31387911 PMCID: PMC6686045 DOI: 10.1128/mbio.01675-19] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 07/12/2019] [Indexed: 12/20/2022] Open
Abstract
Blood-brain barrier (BBB) breakdown is a hallmark of many diseases of the central nervous system (CNS). Loss of BBB integrity in CNS diseases such as viral encephalitis results in the loss of nutrient/oxygen delivery, rapid infiltration of immune cells, and brain swelling that can exacerbate neuronal injury. Despite this, the cellular and molecular mechanisms that underlie BBB breakdown in viral encephalitis are incompletely understood. We undertook a comprehensive analysis of the cellular and molecular signaling events that induce BBB breakdown in an experimental model of virus-induced encephalitis in which neonatal mice are infected with reovirus (serotype 3 strain Abney). We show that BBB leakage during reovirus infection correlates with morphological changes in the vasculature, reductions in pericytes (BBB supporting cells), and disorganization of vascular junctions. Pathway analysis on RNA sequencing from brain endothelial cells identified the activation of interferon (IFN) signaling within the brain vasculature following reovirus infection. Our in vitro and in vivo studies show that type II IFN mediated by IFN-γ, a well known antiviral signal, is a major contributor to BBB leakage during reovirus infection. We show that IFN-γ reduces barrier properties in cultured brain endothelial cells through Rho kinase (ROCK)-mediated cytoskeletal contractions, resulting in junctional disorganization and cell-cell separations. In vivo neutralization of IFN-γ during reovirus infection significantly improved BBB integrity, pericyte coverage, attenuated vascular ROCK activity, and junctional disorganization. Our work supports a model in which IFN-γ acts directly on the brain endothelium to induce BBB breakdown through a mechanism involving ROCK-induced junctional disorganization.IMPORTANCE In an experimental viral encephalitis mouse model in which mice are infected with reovirus, we show that IFN-γ induces blood-brain barrier leakage. We show that IFN-γ promotes Rho kinase activity, resulting in actin cytoskeletal contractions in the brain endothelium that lead to vascular junctional disorganization and cell-cell separations. These studies now provide insight into a previously unknown mechanism for how blood-brain barrier breakdown occurs in viral encephalitis and implicates IFN-γ-Rho kinase activity as major contributor to this phenomenon. By identifying this mechanism of blood-brain barrier breakdown, we now provide potential therapeutic targets in treating patients with viral causes of encephalitis with the hope of limiting damage to the central nervous system.
Collapse
Affiliation(s)
- Stephanie Bonney
- Department of Pediatrics, Section of Developmental Biology, University of Colorado, School of Medicine, Aurora, Colorado, USA
- Cell Biology, Stem Cells, and Development Graduate Program, University of Colorado, School of Medicine, Aurora, Colorado, USA
| | - Scott Seitz
- Microbiology Graduate Program, University of Colorado, School of Medicine, Aurora, Colorado, USA
- Department of Neurology, University of Colorado, School of Medicine, Aurora, Colorado, USA
| | - Caitlin A Ryan
- Department of Pediatrics, Section of Developmental Biology, University of Colorado, School of Medicine, Aurora, Colorado, USA
| | - Kenneth L Jones
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant, University of Colorado, School of Medicine, Aurora, Colorado, USA
| | - Penny Clarke
- Department of Neurology, University of Colorado, School of Medicine, Aurora, Colorado, USA
| | - Kenneth L Tyler
- Department of Neurology, University of Colorado, School of Medicine, Aurora, Colorado, USA
| | - Julie A Siegenthaler
- Department of Pediatrics, Section of Developmental Biology, University of Colorado, School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
36
|
Leibrand CR, Paris JJ, Jones AM, Masuda QN, Halquist MS, Kim WK, Knapp PE, Kashuba ADM, Hauser KF, McRae M. HIV-1 Tat and opioids act independently to limit antiretroviral brain concentrations and reduce blood-brain barrier integrity. J Neurovirol 2019; 25:560-577. [PMID: 31102185 PMCID: PMC6750988 DOI: 10.1007/s13365-019-00757-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 03/28/2019] [Accepted: 04/25/2019] [Indexed: 12/11/2022]
Abstract
Poor antiretroviral penetration may contribute to human immunodeficiency virus (HIV) persistence within the brain and to neurocognitive deficits in opiate abusers. To investigate this problem, HIV-1 Tat protein and morphine effects on blood-brain barrier (BBB) permeability and drug brain penetration were explored using a conditional HIV-1 Tat transgenic mouse model. Tat and morphine effects on the leakage of fluorescently labeled dextrans (10-, 40-, and 70-kDa) into the brain were assessed. To evaluate effects on antiretroviral brain penetration, Tat+ and Tat- mice received three antiretroviral drugs (dolutegravir, abacavir, and lamivudine) with or without concurrent morphine exposure. Antiretroviral and morphine brain and plasma concentrations were determined by LC-MS/MS. Morphine exposure, and, to a lesser extent, Tat, significantly increased tracer leakage from the vasculature into the brain. Despite enhanced BBB breakdown evidenced by increased tracer leakiness, morphine exposure led to significantly lower abacavir concentrations within the striatum and significantly less dolutegravir within the hippocampus and striatum (normalized to plasma). P-glycoprotein, an efflux transporter for which these drugs are substrates, expression and function were significantly increased in the brains of morphine-exposed mice compared to mice not exposed to morphine. These findings were consistent with lower antiretroviral concentrations in brain tissues examined. Lamivudine concentrations were unaffected by Tat or morphine exposure. Collectively, our investigations indicate that Tat and morphine differentially alter BBB integrity. Morphine decreased brain concentrations of specific antiretroviral drugs, perhaps via increased expression of the drug efflux transporter, P-glycoprotein.
Collapse
Affiliation(s)
- Crystal R Leibrand
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Jason J Paris
- Department of BioMolecular Sciences, School of Pharmacy, The University of Mississippi, University, MS, 38677, USA
| | - Austin M Jones
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Quamrun N Masuda
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Matthew S Halquist
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Woong-Ki Kim
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Pamela E Knapp
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Angela D M Kashuba
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7569, USA
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - MaryPeace McRae
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| |
Collapse
|
37
|
Smith LK, Kuhn TB, Chen J, Bamburg JR. HIV Associated Neurodegenerative Disorders: A New Perspective on the Role of Lipid Rafts in Gp120-Mediated Neurotoxicity. Curr HIV Res 2019; 16:258-269. [PMID: 30280668 PMCID: PMC6398609 DOI: 10.2174/1570162x16666181003144740] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/17/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023]
Abstract
The implementation of combination antiretroviral therapy (cART) as the primary means of treatment for HIV infection has achieved a dramatic decline in deaths attributed to AIDS and the reduced incidence of severe forms of HIV-associated neurocognitive disorders (HAND) in infected individuals. Despite these advances, milder forms of HAND persist and prevalence of these forms of neurocognitive impairment are rising with the aging population of HIV infected individuals. HIV enters the CNS early in the pathophysiology establishing persistent infection in resident macrophages and glial cells. These infected cells, in turn, secrete neurotoxic viral proteins, inflammatory cytokines, and small metabolites thought to contribute to neurodegenerative processes. The viral envelope protein gp120 has been identified as a potent neurotoxin affecting neurodegeneration via indirect and direct mechanisms involving interactions with chemokine co-receptors CCR5 and CXCR4. This short review focuses on gp120 neurotropism and associated mechanisms of neurotoxicity linked to chemokine receptors CCR5 and CXCR4 with a new perspective on plasma membrane lipid rafts as an active participant in gp120-mediated neurodegeneration underlying HIV induced CNS pathology.
Collapse
Affiliation(s)
- Lisa K Smith
- Department of Chemistry and Biochemistry, University of Alaska Fairbanks, Fairbanks, AK, United States
| | - Thomas B Kuhn
- Department of Chemistry and Biochemistry, University of Alaska Fairbanks, Fairbanks, AK, United States
| | - Jack Chen
- Department of Biology and Wildlife, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK, United States
| | - James R Bamburg
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
38
|
Age-Related Decrease in Tyrosine Hydroxylase Immunoreactivity in the Substantia Nigra and Region-Specific Changes in Microglia Morphology in HIV-1 Tg Rats. Neurotox Res 2019; 36:563-582. [PMID: 31286433 DOI: 10.1007/s12640-019-00077-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/28/2019] [Accepted: 06/14/2019] [Indexed: 12/12/2022]
Abstract
Animal models have been used to study cellular processes related to human immunodeficiency virus-1 (HIV-1)-associated neurocognitive disorders (HAND). The HIV-1 transgenic (Tg) rat expresses HIV viral genes except the gag-pol replication genes and exhibits neuropathological features similar to HIV patients receiving combined antiretroviral therapy (cART). Using this rat, alterations in dopaminergic function have been demonstrated; however, the data for neuroinflammation and glial reactivity is conflicting. Differences in behavior, tyrosine hydroxylase (TH) immunoreactivity, neuroinflammation, and glia reactivity were assessed in HIV-1 Tg male rats. At 6 and 12 weeks of age, rotarod performance was diminished, motor activity was not altered, and active avoidance latency performance and memory were diminished in HIV-1 Tg rats. TH+ immunoreactivity in the substantia nigra (SN) was decreased at 8 months but not at 2-5 months. At 5 months, astrocyte and microglia morphology was not altered in the cortex, hippocampus, or SN. In the striatum, astrocytes were unaltered, microglia displayed slightly thickened proximal processes, mRNA levels for Iba1 and Cd11b were elevated, and interleukin (Il)1α,Cxcr3, and cell adhesion molecule, Icam, decreased. In the hippocampus, mRNA levels for Tnfa and Cd11b were slightly elevated. No changes were observed in the cortex or SN. The data support an age-related effect of HIV proteins upon the nigrostriatal dopaminergic system and suggest an early response of microglia in the terminal synaptic region with little evidence of an associated neuroinflammatory response across brain regions.
Collapse
|
39
|
Hammond JW, Qiu WQ, Marker DF, Chamberlain JM, Greaves-Tunnell W, Bellizzi MJ, Lu SM, Gelbard HA. HIV Tat causes synapse loss in a mouse model of HIV-associated neurocognitive disorder that is independent of the classical complement cascade component C1q. Glia 2018; 66:2563-2574. [PMID: 30325063 DOI: 10.1002/glia.23511] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/08/2018] [Accepted: 08/08/2018] [Indexed: 12/14/2022]
Abstract
Microglial activation, increased proinflammatory cytokine production, and a reduction in synaptic density are key pathological features associated with HIV-associated neurocognitive disorders (HAND). Even with combination antiretroviral therapy (cART), more than 50% of HIV-positive individuals experience some type of cognitive impairment. Although viral replication is inhibited by cART, HIV proteins such as Tat are still produced within the nervous system that are neurotoxic, involved in synapse elimination, and provoke enduring neuroinflammation. As complement deposition on synapses followed by microglial engulfment has been shown during normal development and disease to be a mechanism for pruning synapses, we have tested whether complement is required for the loss of synapses that occurs after a cortical Tat injection mouse model of HAND. In Tat-injected animals evaluated 7 or 28 days after injection, levels of early complement pathway components, C1q and C3, are significantly elevated and associated with microgliosis and a loss of synapses. However, C1qa knockout mice have the same level of Tat-induced synapse loss as wild-type (WT) mice, showing that the C1q-initiated classical complement cascade is not driving synapse removal during HIV1 Tat-induced neuroinflammation.
Collapse
Affiliation(s)
- Jennetta W Hammond
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| | - Wen Q Qiu
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| | - Daniel F Marker
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| | - Jeffrey M Chamberlain
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| | - Will Greaves-Tunnell
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| | - Matthew J Bellizzi
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| | - Shao-Ming Lu
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| | - Harris A Gelbard
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
40
|
Abstract
Human immunodeficiency virus (HIV)-associated neurocognitive disorders (HAND) remain a common end-organ manifestation of viral infection. Subclinical and mild symptoms lead to neurocognitive and behavioral abnormalities. These are associated, in part, with viral penetrance and persistence in the central nervous system. Infections of peripheral blood monocytes, macrophages, and microglia are the primary drivers of neuroinflammation and neuronal impairments. While current antiretroviral therapy (ART) has reduced the incidence of HIV-associated dementia, milder forms of HAND continue. Depression, comorbid conditions such as infectious liver disease, drugs of abuse, antiretroviral drugs themselves, age-related neurodegenerative diseases, gastrointestinal maladies, and concurrent social and economic issues can make accurate diagnosis of HAND challenging. Increased life expectancy as a result of ART clearly creates this variety of comorbid conditions that often blur the link between the virus and disease. With the discovery of novel biomarkers, neuropsychologic testing, and imaging techniques to better diagnose HAND, the emergence of brain-penetrant ART, adjunctive therapies, longer life expectancy, and better understanding of disease pathogenesis, disease elimination is perhaps a realistic possibility. This review focuses on HIV-associated disease pathobiology with an eye towards changing trends in the face of widespread availability of ART.
Collapse
|
41
|
Nookala AR, Schwartz DC, Chaudhari NS, Glazyrin A, Stephens EB, Berman NEJ, Kumar A. Methamphetamine augment HIV-1 Tat mediated memory deficits by altering the expression of synaptic proteins and neurotrophic factors. Brain Behav Immun 2018; 71:37-51. [PMID: 29729322 PMCID: PMC6003882 DOI: 10.1016/j.bbi.2018.04.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 01/06/2023] Open
Abstract
Methamphetamine (METH) abuse is common among individuals infected with HIV-1 and has been shown to affect HIV replication and pathogenesis. These HIV-1 infected individuals also exhibit greater neuronal injury and higher cognitive decline. HIV-1 proteins, specifically gp120 and HIV-1 Tat, have been earlier shown to affect neurocognition. HIV-1 Tat, a viral protein released early during HIV-1 replication, contributes to HIV-associated neurotoxicity through various mechanisms including production of pro-inflammatory cytokines, reactive oxygen species and dysregulation of neuroplasticity. However, the combined effect of METH and HIV-1 Tat on neurocognition and its potential effect on neuroplasticity mechanisms remains largely unknown. Therefore, the present study was undertaken to investigate the combined effect of METH and HIV-1 Tat on behavior and on the expression of neuroplasticity markers by utilizing Doxycycline (DOX)-inducible HIV-1 Tat (1-86) transgenic mice. Expression of Tat in various brain regions of these mice was confirmed by RT-PCR. The mice were administered with an escalating dose of METH (0.1 mg/kg to 6 mg/kg, i.p) over a 7-day period, followed by 6 mg/kg, i.p METH twice a day for four weeks. After three weeks of METH administration, Y maze and Morris water maze assays were performed to determine the effect of Tat and METH on working and spatial memory, respectively. Compared with controls, working memory was significantly decreased in Tat mice that were administered METH. Moreover, significant deficits in spatial memory were also observed in Tat-Tg mice that were administered METH. A significant reduction in the protein expressions of synapsin 1, synaptophysin, Arg3.1, PSD-95, and BDNF in different brain regions were also observed. Expression levels of Calmodulin kinase II (CaMKII), a marker of synaptodendritic integrity, were also significantly decreased in HIV-1 Tat mice that were treated with METH. Together, this data suggests that METH enhances HIV-1 Tat-induced memory deficits by reducing the expression of pre- and postsynaptic proteins and neuroplasticity markers, thus providing novel insights into the molecular mechanisms behind neurocognitive impairments in HIV-infected amphetamine users.
Collapse
Affiliation(s)
- Anantha Ram Nookala
- Division of Pharmacology and Toxicology, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO, 64108, USA
| | - Daniel C. Schwartz
- Division of Pharmacology and Toxicology, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO, 64108, USA
| | - Nitish S. Chaudhari
- Division of Pharmacology and Toxicology, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO, 64108, USA
| | - Alexy Glazyrin
- Department of Pathology, School of Medicine, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, 64108, USA
| | - Edward B. Stephens
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Nancy E. J. Berman
- Department of Anatomy and Cell biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Anil Kumar
- Division of Pharmacology and Toxicology, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO, 64108, USA.
| |
Collapse
|
42
|
Gonek M, McLane VD, Stevens DL, Lippold K, Akbarali HI, Knapp PE, Dewey WL, Hauser KF, Paris JJ. CCR5 mediates HIV-1 Tat-induced neuroinflammation and influences morphine tolerance, dependence, and reward. Brain Behav Immun 2018; 69:124-138. [PMID: 29146238 PMCID: PMC5857418 DOI: 10.1016/j.bbi.2017.11.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/02/2017] [Accepted: 11/07/2017] [Indexed: 12/16/2022] Open
Abstract
The HIV-1 regulatory protein, trans-activator of transcription (Tat), interacts with opioids to potentiate neuroinflammation and neurodegeneration within the CNS. These effects may involve the C-C chemokine receptor type 5 (CCR5); however, the behavioral contribution of CCR5 on Tat/opioid interactions is not known. Using a transgenic murine model that expresses HIV-1 Tat protein in a GFAP-regulated, doxycycline-inducible manner, we assessed morphine tolerance, dependence, and reward. To assess the influence of CCR5 on these effects, mice were pretreated with oral vehicle or the CCR5 antagonist, maraviroc, prior to morphine administration. We found that HIV-1 Tat expression significantly attenuated the antinociceptive potency of acute morphine (2-64 mg/kg, i.p.) in non-tolerant mice. Consistent with this, Tat attenuated withdrawal symptoms among morphine-tolerant mice. Pretreatment with maraviroc blocked the effects of Tat, reinstating morphine potency in non-tolerant mice and restoring withdrawal symptomology in morphine-tolerant mice. Twenty-four hours following morphine administration, HIV-1 Tat significantly potentiated (∼3.5-fold) morphine-conditioned place preference and maraviroc further potentiated these effects (∼5.7-fold). Maraviroc exerted no measurable behavioral effects on its own. Protein array analyses revealed only minor changes to cytokine profiles when morphine was administered acutely or repeatedly; however, 24 h post morphine administration, the expression of several cytokines was greatly increased, including endogenous CCR5 chemokine ligands (CCL3, CCL4, and CCL5), as well as CCL2. Tat further elevated levels of several cytokines and maraviroc pretreatment attenuated these effects. These data demonstrate that CCR5 mediates key aspects of HIV-1 Tat-induced alterations in the antinociceptive potency and rewarding properties of opioids.
Collapse
Affiliation(s)
- Maciej Gonek
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA
| | - Virginia D. McLane
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA
| | - David L. Stevens
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA
| | - Kumiko Lippold
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA
| | - Hamid I. Akbarali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA
| | - Pamela E. Knapp
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA,Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980709, Richmond, VA 23298-0709, USA,Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980059, Richmond, VA 23298-0059, USA
| | - William L. Dewey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA,Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980059, Richmond, VA 23298-0059, USA
| | - Kurt F. Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA,Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980709, Richmond, VA 23298-0709, USA,Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980059, Richmond, VA 23298-0059, USA
| | - Jason J. Paris
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA,Department of BioMolecular Sciences, University of Mississippi, School of Pharmacy, P.O. Box 1848, University, MS 38677-1848, USA,Research Institute of Pharmaceutical Sciences, University of Mississippi, School of Pharmacy, P.O. Box 1848, University, MS 38677-1848, USA,Address for Correspondence: Jason J. Paris, Ph.D. Assistant Professor of Pharmacology, The University of Mississippi, School of Pharmacy, P.O. Box 1848, 315 Faser Hall, University, MS 38677-1848, U.S.A. Phone: +1-662-915-3096,
| |
Collapse
|
43
|
McRae M, LaFratta LM, Nguyen BM, Paris JJ, Hauser KF, Conway DE. Characterization of cell-cell junction changes associated with the formation of a strong endothelial barrier. Tissue Barriers 2018; 6:e1405774. [PMID: 29388870 DOI: 10.1080/21688370.2017.1405774] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A principal function of endothelial cells is the formation of a barrier between the blood and tissues. This barrier arises from the physical connections at cell-cell junctions, which includes cytoskeletal tight junction and adherens junction proteins. Methods that alter barrier function must therefore affect these cell-cell connections. The blood brain barrier (BBB) represents perhaps the most selective endothelial barrier, which arises from endothelial cell interactions with astrocytes and pericytes. Even in non-central nervous system (CNS) endothelial cells, barrier properties can be enhanced, mimicking the BBB, through induction of intercellular junctions, by either direct co-culture with astrocytes, supplementation with astrocyte conditioned medium (ACM) and/or pharmacologic enhancement of cAMP. To understand how cell-cell junctions change during endothelial barrier enhancement, we examined the effects of ACM and/or cAMP donors added to standard media on human umbilical vein endothelial cells (HUVEC). HUVEC cultured with cAMP-elevating agents had the most enhanced barrier function as measured by Electric Cell-substrate Impedance Sensing (ECIS®), a real-time, label-free, impedance based method of studying cell barrier properties. However, subtle differences in actin and cell-cell junction proteins were seen across all four culture conditions. cAMP-elevating agents also triggered the redistribution of ZO-1 and VE-cadherin to cell-cell junctions, and intensified the actin microfilament network at the cell cortex. Using a VE-cadherin FRET-force sensor, we observed a decrease in VE-cadherin force in HUVEC cultured with ACM with cAMP donors. Our data indicate cAMP elevation induces both junctional strengthening and reduced VE-cadherin forces. Additionally, treatment with an inhibitor of formin, which reduced actin stress fibers, enhanced barrier function. These data suggest that barrier function is modulated both through the trafficking of proteins to cell-cell junctions, and through the modulation and a relaxation of mechanical force through adherens junctions as intercellular junctional complexes become established.
Collapse
Affiliation(s)
- MaryPeace McRae
- a Department of Pharmacotherapy and Outcomes Science , School of Pharmacy, Virginia Commonwealth University , Richmond , VA , USA
| | - Lindsay M LaFratta
- b Department of Biomedical Engineering , Virginia Commonwealth University , Richmond , VA , USA
| | - Benjamin M Nguyen
- b Department of Biomedical Engineering , Virginia Commonwealth University , Richmond , VA , USA
| | - Jason J Paris
- c Department of BioMolecular Sciences , School of Pharmacy, University of Mississippi, University , MS , USA
| | - Kurt F Hauser
- d Department of Pharmacology , Virginia Commonwealth University , Richmond , VA , USA
| | - Daniel E Conway
- b Department of Biomedical Engineering , Virginia Commonwealth University , Richmond , VA , USA
| |
Collapse
|
44
|
Effects of HIV-1 Tat and Methamphetamine on Blood-Brain Barrier Integrity and Function In Vitro. Antimicrob Agents Chemother 2017; 61:AAC.01307-17. [PMID: 28893794 DOI: 10.1128/aac.01307-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/05/2017] [Indexed: 12/28/2022] Open
Abstract
Human immunodeficiency (HIV) infection results in neurocognitive deficits in about one half of infected individuals. Despite systemic effectiveness, restricted antiretroviral penetration across the blood-brain barrier (BBB) is a major limitation in fighting central nervous system (CNS)-localized infection. Drug abuse exacerbates HIV-induced cognitive and pathological CNS changes. This study's purpose was to investigate the effects of the HIV-1 protein Tat and methamphetamine on factors affecting drug penetration across an in vitro BBB model. Factors affecting paracellular and transcellular flux in the presence of Tat and methamphetamine were examined. Transendothelial electrical resistance, ZO-1 expression, and lucifer yellow (a paracellular tracer) flux were aspects of paracellular processes that were examined. Additionally, effects on P-glycoprotein (P-gp) and multidrug resistance protein 1 (MRP-1) mRNA (via quantitative PCR [qPCR]) and protein (via immunoblotting) expression were measured; Pgp and MRP-1 are drug efflux proteins. Transporter function was examined after exposure of Tat with or without methamphetamine using the P-gp substrate rhodamine 123 and also using the dual P-gp/MRP-1 substrate and protease inhibitor atazanavir. Tat and methamphetamine elicit complex changes affecting transcellular and paracellular transport processes. Neither Tat nor methamphetamine significantly altered P-gp expression. However, Tat plus methamphetamine exposure significantly increased rhodamine 123 accumulation within brain endothelial cells, suggesting that treatment inhibited or impaired P-gp function. Intracellular accumulation of atazanavir was not significantly altered after Tat or methamphetamine exposure. Atazanavir accumulation was, however, significantly increased by simultaneous inhibition of P-gp and MRP. Collectively, our investigations indicate that Tat and methamphetamine alter aspects of BBB integrity without affecting net flux of paracellular compounds. Tat and methamphetamine may also affect several aspects of transcellular transport.
Collapse
|