1
|
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)
- Narendran Annadurai
- Department of Anesthesiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-4455, USA
| | - Georgette D Kanmogne
- Department of Anesthesiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-4455, USA
| |
Collapse
|
2
|
Clare K, Park K, Pan Y, Lejuez CW, Volkow ND, Du C. Neurovascular effects of cocaine: relevance to addiction. Front Pharmacol 2024; 15:1357422. [PMID: 38455961 PMCID: PMC10917943 DOI: 10.3389/fphar.2024.1357422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 01/22/2024] [Indexed: 03/09/2024] Open
Abstract
Cocaine is a highly addictive drug, and its use is associated with adverse medical consequences such as cerebrovascular accidents that result in debilitating neurological complications. Indeed, brain imaging studies have reported severe reductions in cerebral blood flow (CBF) in cocaine misusers when compared to the brains of healthy non-drug using controls. Such CBF deficits are likely to disrupt neuro-vascular interaction and contribute to changes in brain function. This review aims to provide an overview of cocaine-induced CBF changes and its implication to brain function and to cocaine addiction, including its effects on tissue metabolism and neuronal activity. Finally, we discuss implications for future research, including targeted pharmacological interventions and neuromodulation to limit cocaine use and mitigate the negative impacts.
Collapse
Affiliation(s)
- Kevin Clare
- New York Medical College, Valhalla, NY, United States
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Kicheon Park
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Yingtian Pan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Carl W. Lejuez
- Department of Psychology, Stony Brook University, Stony Brook, NY, United States
| | - Nora D. Volkow
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Congwu Du
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
3
|
Colón Ortiz R, Knerler S, Fridman LB, Mercado A, Price AS, Rosado-Franco JJ, Wilkins H, Flores BR, Orsburn BC, Williams DW. Cocaine regulates antiretroviral therapy CNS access through pregnane-x receptor-mediated drug transporter and metabolizing enzyme modulation at the blood brain barrier. Fluids Barriers CNS 2024; 21:5. [PMID: 38200564 PMCID: PMC10777548 DOI: 10.1186/s12987-023-00507-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Appropriate interactions between antiretroviral therapies (ART) and drug transporters and metabolizing enzymes at the blood brain barrier (BBB) are critical to ensure adequate dosing of the brain to achieve HIV suppression. These proteins are modulated by demographic and lifestyle factors, including substance use. While understudied, illicit substances share drug transport and metabolism pathways with ART, increasing the potential for adverse drug:drug interactions. This is particularly important when considering the brain as it is relatively undertreated compared to peripheral organs and is vulnerable to substance use-mediated damage. METHODS We used an in vitro model of the human BBB to determine the extravasation of three first-line ART drugs, emtricitabine (FTC), tenofovir (TFV), and dolutegravir (DTG), in the presence and absence of cocaine, which served as our illicit substance model. The impact of cocaine on BBB integrity and permeability, drug transporters, metabolizing enzymes, and their master transcriptional regulators were evaluated to determine the mechanisms by which substance use impacted ART central nervous system (CNS) availability. RESULTS We determined that cocaine had a selective impact on ART extravasation, where it increased FTC's ability to cross the BBB while decreasing TFV. DTG concentrations that passed the BBB were below quantifiable limits. Interestingly, the potent neuroinflammatory modulator, lipopolysaccharide, had no effect on ART transport, suggesting a specificity for cocaine. Unexpectedly, cocaine did not breach the BBB, as permeability to albumin and 4 kDa FITC-dextran, as well as tight junction proteins and adhesion molecules remained unchanged. Rather, cocaine selectively decreased the pregnane-x receptor (PXR), but not constitutive androstane receptor (CAR). Consequently, drug transporter expression and activity decreased in endothelial cells of the BBB, including p-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and multidrug resistance-associated protein 4 (MRP4). Further, cytochrome P450 3A4 (CYP3A4) enzymatic activity increased following cocaine treatment that coincided with decreased expression. Finally, cocaine modulated adenylate kinases that are required to facilitate biotransformation of ART prodrugs to their phosphorylated, pharmacologically active counterparts. CONCLUSION Our findings indicate that additional considerations are needed in CNS HIV treatment strategies for people who use cocaine, as it may limit ART efficacy through regulation of drug transport and metabolizing pathways at the BBB.
Collapse
Affiliation(s)
- Rodnie Colón Ortiz
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Stephen Knerler
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Lisa B Fridman
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Alicia Mercado
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Amira-Storm Price
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Jose J Rosado-Franco
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Hannah Wilkins
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Bianca R Flores
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Benjamin C Orsburn
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Dionna W Williams
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Department of Molecular Microbiology & Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA.
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Rollins Research Center, 1510 Clifton Road NE, 30322, Atlanta, Georgia.
| |
Collapse
|
4
|
Fridman LB, Knerler S, Price AS, Ortiz RC, Mercado A, Wilkins H, Flores BR, Orsburn BC, Williams DW. Cocaine Regulates Antiretroviral Therapy CNS Access Through Pregnane-X Receptor-Mediated Drug Transporter and Metabolizing Enzyme Modulation at the Blood Brain Barrier. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.28.551042. [PMID: 37546800 PMCID: PMC10402182 DOI: 10.1101/2023.07.28.551042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Background Appropriate interactions between antiretroviral therapies (ART) and drug transporters and metabolizing enzymes at the blood brain barrier (BBB) are critical to ensure adequate dosing of the brain to achieve HIV suppression. These proteins are modulated by demographic and lifestyle factors, including substance use. While understudied, illicit substances share drug transport and metabolism pathways with ART, increasing the potential for adverse drug:drug interactions. This is particularly important when considering the brain as it is relatively undertreated compared to peripheral organs and is vulnerable to substance use-mediated damage. Methods We used an in vitro model of the human BBB to determine the extravasation of three first-line ART drugs, emtricitabine (FTC), tenofovir (TFV), and dolutegravir (DTG), in the presence and absence of cocaine, which served as our illicit substance model. The impact of cocaine on BBB integrity and permeability, drug transporters, metabolizing enzymes, and their master transcriptional regulators were evaluated to determine the mechanisms by which substance use impacted ART central nervous system (CNS) availability. Results We determined that cocaine had a selective impact on ART extravasation, where it increased FTC's ability to cross the BBB while decreasing TFV. DTG concentrations that passed the BBB were below quantifiable limits. Interestingly, the potent neuroinflammatory modulator, lipopolysaccharide, had no effect on ART transport, suggesting a specificity for cocaine. Unexpectedly, cocaine did not breach the BBB, as permeability to albumin and tight junction proteins and adhesion molecules remained unchanged. Rather, cocaine selectively decreased the pregnane-x receptor (PXR), but not constitutive androstane receptor (CAR). Consequently, drug transporter expression and activity decreased in endothelial cells of the BBB, including p-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and multidrug resistance-associated protein 4 (MRP4). Further, cytochrome P450 3A4 (CYP3A4) enzymatic activity increased following cocaine treatment that coincided with decreased expression. Finally, cocaine modulated adenylate kinases are required to facilitate biotransformation of ART prodrugs to their phosphorylated, pharmacologically active counterparts. Conclusion Our findings indicate that additional considerations are needed in CNS HIV treatment strategies for people who use cocaine, as it may limit ART efficacy through regulation of drug transport and metabolizing pathways at the BBB.
Collapse
Affiliation(s)
- Lisa B. Fridman
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Stephen Knerler
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Amira-Storm Price
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Rodnie Colón Ortiz
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Alicia Mercado
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Hannah Wilkins
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Bianca R. Flores
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Benjamin C. Orsburn
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Dionna W. Williams
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Department of Molecular Microbiology & Immunology, Johns Hopkins School of Public Health, Baltimore, Maryland 21205
| |
Collapse
|
5
|
Cheng D, Luo Z, Fitting S, Stoops W, Heath SL, Ndhlovu LC, Jiang W. The link between chronic cocaine use, B cell perturbations, and blunted immune recovery in HIV-infected individuals on suppressive ART. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:71-79. [PMID: 37027536 PMCID: PMC10070012 DOI: 10.1515/nipt-2022-0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/27/2023] [Indexed: 06/04/2023]
Abstract
Background We recently reveal that anti-CD4 autoantibodies contribute to blunted CD4+ T cell reconstitution in HIV+ individuals on antiretroviral therapy (ART). Cocaine use is common among HIV+ individuals and is associated with accelerated disease progression. However, the mechanisms underlying cocaine-induced immune perturbations remain obscure. Methods We evaluated plasma levels of anti-CD4 IgG and markers of microbial translocation, as well as B-cell gene expression profiles and activation in HIV+ chronic cocaine users and non-users on suppressive ART, as well as uninfected controls. Plasma purified anti-CD4 IgGs were assessed for antibody-dependent cytotoxicity (ADCC). Results HIV+ cocaine users had increased plasma levels of anti-CD4 IgGs, lipopolysaccharide (LPS), and soluble CD14 (sCD14) versus non-users. An inverse correlation was observed in cocaine users, but not non-drug users. Anti-CD4 IgGs from HIV+ cocaine users mediated CD4+ T cell death through ADCC in vitro. B cells from HIV+ cocaine users exhibited activation signaling pathways and activation (cycling and TLR4 expression) related to microbial translocation versus non-users. Conclusions This study improves our understanding of cocaine associated B cell perturbations and immune failure and the new appreciation for autoreactive B cells as novel therapeutic targets.
Collapse
Affiliation(s)
- Da Cheng
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Zhenwu Luo
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Sylvia Fitting
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - William Stoops
- Department of Behavioral Science, Department of Psychiatry, Center on Drug and Alcohol Research, Department of Psychology, University of Kentucky College of Medicine and College of Arts and Sciences, Lexington, KY, USA
| | - Sonya L. Heath
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lishomwa C. Ndhlovu
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC, USA
- Divison of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, USA
| |
Collapse
|
6
|
High-Dose Acetaminophen Alters the Integrity of the Blood-Brain Barrier and Leads to Increased CNS Uptake of Codeine in Rats. Pharmaceutics 2022; 14:pharmaceutics14050949. [PMID: 35631535 PMCID: PMC9144323 DOI: 10.3390/pharmaceutics14050949] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/12/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
The consumption of acetaminophen (APAP) can induce neurological changes in human subjects; however, effects of APAP on blood-brain barrier (BBB) integrity are unknown. BBB changes by APAP can have profound consequences for brain delivery of co-administered drugs. To study APAP effects, female Sprague-Dawley rats (12-16 weeks old) were administered vehicle (i.e., 100% dimethyl sulfoxide (DMSO), intraperitoneally (i.p.)) or APAP (80 mg/kg or 500 mg/kg in DMSO, i.p.; equivalent to a 900 mg or 5600 mg daily dose for a 70 kg human subject). BBB permeability was measured via in situ brain perfusion using [14C]sucrose and [3H]codeine, an opioid analgesic drug that is co-administered with APAP (i.e., Tylenol #3). Localization and protein expression of tight junction proteins (i.e., claudin-5, occludin, ZO-1) were studied in rat brain microvessels using Western blot analysis and confocal microscopy, respectively. Paracellular [14C]sucrose "leak" and brain [3H]codeine accumulation were significantly enhanced in rats treated with 500 mg/kg APAP only. Additionally, claudin-5 localization and protein expression were altered in brain microvessels isolated from rats administered 500 mg/kg APAP. Our novel and translational data show that BBB integrity is altered following a single high APAP dose, results that are relevant to patients abusing or misusing APAP and/or APAP/opioid combination products.
Collapse
|
7
|
Morissette F, Mongeau-Pérusse V, Rizkallah E, Thébault P, Lepage S, Brissette S, Bruneau J, Dubreucq S, Stip E, Cailhier JF, Jutras-Aswad D. Exploring cannabidiol effects on inflammatory markers in individuals with cocaine use disorder: a randomized controlled trial. Neuropsychopharmacology 2021; 46:2101-2111. [PMID: 34331010 PMCID: PMC8505631 DOI: 10.1038/s41386-021-01098-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 01/29/2023]
Abstract
Cocaine use disorder (CUD) is a major public health issue associated with physical, social, and psychological problems. Excessive and repeated cocaine use induces oxidative stress leading to a systemic inflammatory response. Cannabidiol (CBD) has gained substantial interest for its anti-inflammatory properties, safety, and tolerability profile. However, CBD anti-inflammatory properties have yet to be confirmed in humans. This exploratory study is based on a single-site randomized controlled trial that enrolled participants with CUD between 18 and 65 years, randomized (1:1) to daily receive either CBD (800 mg) or placebo for 92 days. The trial was divided into a 10-day detoxification (phase I) followed by a 12-week outpatient follow-up (phase II). Blood samples were collected from 48 participants at baseline, day 8, week 4, and week 12 and were analyzed to determine monocytes and lymphocytes phenotypes, and concentrations of various inflammatory markers such as cytokines. We used generalized estimating equations to detect group differences. Participants treated with CBD had lower levels of interleukin-6 (p = 0.017), vascular endothelial growth factor (p = 0.032), intermediate monocytes CD14+CD16+ (p = 0.024), and natural killer CD56negCD16hi (p = 0.000) compared with participants receiving placebo. CD25+CD4+T cells were higher in the CBD group (p = 0.007). No significant group difference was observed for B lymphocytes. This study suggests that CBD may exert anti-inflammatory effects in individuals with CUD.
Collapse
Affiliation(s)
- Florence Morissette
- grid.14848.310000 0001 2292 3357Faculty of Medicine, Department of Psychiatry and Addictology, Université de Montréal, Montreal, QC Canada ,grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada
| | - Violaine Mongeau-Pérusse
- grid.14848.310000 0001 2292 3357Faculty of Medicine, Department of Psychiatry and Addictology, Université de Montréal, Montreal, QC Canada ,grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada
| | - Elie Rizkallah
- grid.14848.310000 0001 2292 3357Faculty of Medicine, Department of Psychiatry and Addictology, Université de Montréal, Montreal, QC Canada ,grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada
| | - Paméla Thébault
- grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada ,Montreal Cancer Institute, Montreal, QC Canada
| | - Stéphanie Lepage
- grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada ,Montreal Cancer Institute, Montreal, QC Canada
| | - Suzanne Brissette
- grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada ,grid.14848.310000 0001 2292 3357Faculty of Medicine, Department of Family and Emergency Medicine, Université de Montréal, Montreal, QC Canada
| | - Julie Bruneau
- grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada ,grid.14848.310000 0001 2292 3357Faculty of Medicine, Department of Family and Emergency Medicine, Université de Montréal, Montreal, QC Canada
| | - Simon Dubreucq
- grid.14848.310000 0001 2292 3357Faculty of Medicine, Department of Psychiatry and Addictology, Université de Montréal, Montreal, QC Canada ,grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada
| | - Emmanuel Stip
- grid.14848.310000 0001 2292 3357Faculty of Medicine, Department of Psychiatry and Addictology, Université de Montréal, Montreal, QC Canada ,grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada ,grid.43519.3a0000 0001 2193 6666Department of Psychiatry and Behavioral Science, College of Medicine and Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates
| | - Jean-François Cailhier
- grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada ,Montreal Cancer Institute, Montreal, QC Canada ,grid.14848.310000 0001 2292 3357Division of Nephrology, Department of Medicine, Université de Montréal, Montreal, QC Canada
| | - Didier Jutras-Aswad
- Faculty of Medicine, Department of Psychiatry and Addictology, Université de Montréal, Montreal, QC, Canada. .,Research Centre of Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada. .,University Institute on Addictions, Montreal, QC, Canada.
| |
Collapse
|
8
|
The Blood-Brain Barrier: Much More Than a Selective Access to the Brain. Neurotox Res 2021; 39:2154-2174. [PMID: 34677787 DOI: 10.1007/s12640-021-00431-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/30/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022]
Abstract
The blood-brain barrier is a dynamic structure, collectively referred to as the neurovascular unit. It is responsible for the exchange of blood, oxygen, ions, and other molecules between the peripheral circulation and the brain compartment. It is the main entrance to the central nervous system and as such critical for the maintenance of its homeostasis. Dysfunction of the blood-brain barrier is a characteristic of several neurovascular pathologies. Moreover, physiological changes, environmental factors, nutritional habits, and psychological stress can modulate the tightness of the barrier. In this contribution, we summarize our current understanding of structure and function of this important component of the brain. We also describe the neurological deficits associated with its damage. A special emphasis is placed in the effect of the exposure to xenobiotics and pollutants in the permeability of the barrier. Finally, current protective strategies as well as the culture models to study this fascinating structure are discussed.
Collapse
|
9
|
Assessing combinatorial effects of HIV infection and former cocaine dependence on cognitive control processes: A high-density electrical mapping study of response inhibition. Neuropharmacology 2021; 195:108636. [PMID: 34090915 DOI: 10.1016/j.neuropharm.2021.108636] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/19/2021] [Accepted: 05/28/2021] [Indexed: 01/09/2023]
Abstract
Stimulant drug use in HIV + patients is associated with poor personal and public health outcomes, including high-risk sexual behavior and faster progression from HIV to AIDS. Inhibitory control--the ability to withhold a thought, feeling, or action--is a central construct involved in the minimization of risk-taking behaviors. Recent neuroimaging and behavioral evidence indicate normalization of inhibitory control processes in former cocaine users as a function of the duration of drug abstinence, but it is unknown whether this recovery trajectory persists in former users with comorbid HIV. Here, we investigate the neural correlates of inhibitory control in 103 human subjects using high-density EEG recording as participants performed a Go/NoGo response inhibition task. Four groups of participants were recruited, varying on HIV and cocaine-dependence status. Electrophysiological responses to successful inhibitions and behavioral task performance were compared among groups. Results indicate persistent behavioral and neurophysiological impairment in HIV+ patients' response inhibition despite current abstinence from cocaine. Analysis of task performance showed that HIV+ abstinent cocaine-dependent participants demonstrate the lowest performance of all groups across all metrics of task accuracy. Planned comparisons of electrophysiological components revealed a main effect of scalp site and an interaction between HIV-status and scalp site on N2 amplitudes during successful inhibitions. Analysis of the P3 time region showed a main effect of scalp site and an interaction between HIV-status and cocaine dependence. These results suggest synergistic alterations in the neurophysiology of response inhibition and indicate that abstinence-related recovery of inhibitory control may be attenuated in patients with HIV.
Collapse
|
10
|
Hersey M, Bacon AK, Bailey LG, Coggiano MA, Newman AH, Leggio L, Tanda G. Psychostimulant Use Disorder, an Unmet Therapeutic Goal: Can Modafinil Narrow the Gap? Front Neurosci 2021; 15:656475. [PMID: 34121988 PMCID: PMC8187604 DOI: 10.3389/fnins.2021.656475] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
The number of individuals affected by psychostimulant use disorder (PSUD) has increased rapidly over the last few decades resulting in economic, emotional, and physical burdens on our society. Further compounding this issue is the current lack of clinically approved medications to treat this disorder. The dopamine transporter (DAT) is a common target of psychostimulant actions related to their use and dependence, and the recent availability of atypical DAT inhibitors as a potential therapeutic option has garnered popularity in this research field. Modafinil (MOD), which is approved for clinical use for the treatment of narcolepsy and sleep disorders, blocks DAT just like commonly abused psychostimulants. However, preclinical and clinical studies have shown that it lacks the addictive properties (in both behavioral and neurochemical studies) associated with other abused DAT inhibitors. Clinical availability of MOD has facilitated its off-label use for several psychiatric disorders related to alteration of brain dopamine (DA) systems, including PSUD. In this review, we highlight clinical and preclinical research on MOD and its R-enantiomer, R-MOD, as potential medications for PSUD. Given the complexity of PSUD, we have also reported the effects of MOD on psychostimulant-induced appearance of several symptoms that could intensify the severity of the disease (i.e., sleep disorders and impairment of cognitive functions), besides the potential therapeutic effects of MOD on PSUD.
Collapse
Affiliation(s)
- Melinda Hersey
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Amanda K. Bacon
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Lydia G. Bailey
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Mark A. Coggiano
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Amy H. Newman
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Lorenzo Leggio
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
- Clinical Psychoneuroendo- crinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
- National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Bethesda, MD, United States
| | - Gianluigi Tanda
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
11
|
Kumar S, Crenshaw BJ, Williams SD, Bell CR, Matthews QL, Sims B. Cocaine-Specific Effects on Exosome Biogenesis in Microglial Cells. Neurochem Res 2021; 46:1006-1018. [PMID: 33559104 PMCID: PMC7946671 DOI: 10.1007/s11064-021-03231-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/18/2020] [Accepted: 01/04/2021] [Indexed: 12/31/2022]
Abstract
Cocaine is a highly addictive stimulant and a well-known drug, with multiple effects on physiology. Cocaine can have direct effects on all cell types in the brain, including microglia. Microglia can be activated by other conditions, such as infection, inflammation, or injury. However, how cocaine regulates microglia and the influence of cocaine on microglial-derived exosomes remains unknown. Exosomes are nanovesicles that are responsible for intercellular communications, signaling, and trafficking necessary cargo for cell homeostasis. In this study, we hypothesized that cocaine affects exosome biogenesis and composition in BV2 microglial cells. BV2 microglial cells were cultured in exosome-depleted RPMI-1640 media and were treated according to the experimental designs. We observed that cell viability decreased by 11% at 100 µM cocaine treatment but was unaffected at other concentrations. After treatments, the exosomes were isolated from the condition media. Purified exosomes were characterized and quantified using transmission electron microscope (TEM) and nanoparticle tracking analysis (NTA). By NTA, there was a significant decrease in particles/mL after cocaine treatment. There was a 39.5%, 58.1%, 32.3% and 28.1% decrease in particles/mL at 100 nM, 1 μM, 10 μM and 100 μM cocaine, respectively. The characterization of exosomes and exosomal protein was performed by western/dot blot analyses. Tetraspanins CD11b, CD18 and CD63 were relatively unchanged after cocaine treatment. The heat shock proteins (Hsps), Hsp70 and Hsp90, were both significantly increased at 10 μM and 100 μM, but only hsp70 was significantly increased at 10 nM. The Rab proteins were assessed to investigate their role in cocaine-mediated exosomal decrease. Rab11 was significantly decreased at 10 nM, 100 nM, 1 μM, 10 μM and 100 μM by 15%, 28%, 25%, 38% and 22%, respectively. Rab27 was decreased at all concentrations but only significantly decreased at 100 nM, 1 μM and 100 μM cocaine by 21%, 24% and 23%, respectively. Rab35 had no significant changes noted when compared to control. Rab7 increased at all cocaine concentrations but only a significant increase in expression at 100 nM and 10 μM by 1.32-fold and 1.4-fold increase. Cocaine was found to alter exosome biogenesis and composition in BV2 microglial cells. Western and dot blot analyses verified the identities of purified exosomes, and the specific protein compositions of exosomes were found to change in the presence of cocaine. Furthermore, cocaine exposure modulated the expression of exosomal proteins, such as Hsps and Rab GTPases, suggesting the protein composition and formation of microglial-derived exosomes were regulated by cocaine.
Collapse
Affiliation(s)
- Sanjay Kumar
- Department of Pediatrics/Division of Neonatology and Center of Glial Biology in Medicine at the University of Alabama School of Medicine, UAB Women and Infant Center, University of Alabama, 1700 6th Ave South, Birmingham, AL, 35294, USA
| | - Brennetta J Crenshaw
- Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL, 36104, USA
| | - Sparkle D Williams
- Department of Pediatrics/Division of Neonatology and Center of Glial Biology in Medicine at the University of Alabama School of Medicine, UAB Women and Infant Center, University of Alabama, 1700 6th Ave South, Birmingham, AL, 35294, USA
| | - Courtnee' R Bell
- Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL, 36104, USA
| | - Qiana L Matthews
- Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL, 36104, USA
| | - Brian Sims
- Department of Pediatrics/Division of Neonatology and Center of Glial Biology in Medicine at the University of Alabama School of Medicine, UAB Women and Infant Center, University of Alabama, 1700 6th Ave South, Birmingham, AL, 35294, USA.
| |
Collapse
|
12
|
Doke M, Jeganathan V, McLaughlin JP, Samikkannu T. HIV-1 Tat and cocaine impact mitochondrial epigenetics: effects on DNA methylation. Epigenetics 2020; 16:980-999. [PMID: 33100130 PMCID: PMC8451453 DOI: 10.1080/15592294.2020.1834919] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Human immunodeficiency virus (HIV) infection and the psychostimulant drug cocaine are known to induce epigenetic changes in DNA methylation that are linked with the severity of viral replication and disease progression, which impair neuronal functions. Increasing evidence suggests that changes in DNA methylation and hydroxymethylation occur in mitochondrial DNA (mtDNA) and represent mitochondrial genome epigenetic modifications (mitoepigenetic modifications). These modifications likely regulate both mtDNA replication and gene expression. However, mtDNA methylation has not been studied extensively in the contexts of cocaine abuse and HIV-1 infection. In the present study, epigenetic factors changed the levels of the DNA methyltransferases (DNMTs) DNMT1, DNMT3a, and DNMT3b, the Ten-eleven translocation (TET) enzymes 1, 2, and 3, and mitochondrial DNMTs (mtDNMTs) both in vitro and in vivo. These changes resulted in alterations in mtDNA methylation levels at CpG and non-CpG sites in human primary astrocytes as measured using targeted next-generation bisulphite sequencing (TNGBS). Moreover, mitochondrial methylation levels in the MT-RNR1, MT-ND5, MT-ND1, D-loop and MT-CYB regions of mtDNA were lower in the HIV-1 Tat and cocaine treatment groups than in the control group. In summary, the present findings suggest that mitoepigenetic modification in the human brain causes the mitochondrial dysfunction that gives rise to neuro-AIDS.
Collapse
Affiliation(s)
- Mayur Doke
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX, USA
| | - Venkatesh Jeganathan
- Department of Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Thangavel Samikkannu
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX, USA
| |
Collapse
|
13
|
Małkiewicz MA, Małecki A, Toborek M, Szarmach A, Winklewski PJ. Substances of abuse and the blood brain barrier: Interactions with physical exercise. Neurosci Biobehav Rev 2020; 119:204-216. [PMID: 33038347 DOI: 10.1016/j.neubiorev.2020.09.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 05/22/2020] [Accepted: 09/25/2020] [Indexed: 12/22/2022]
Abstract
Substance use disorders pose a common medical, social and financial problem. Among the pathomechanisms of substance use disorders, the disruption and increased permeability of the blood-brain barrier has been recently revealed. Physical exercise appears to be a relatively inexpensive and feasible way to implement behavioral therapy counteracting the blood-brain barrier impairment. Concomitantly, there are also studies supporting a potential protective role of selected substances of abuse in maintaining the blood-brain barrier integrity. In this review, we aim to provide a summary on the modulatory influence of physical exercise, a non-pharmacological intervention, on the blood-brain barrier alterations caused by substances of abuse. Further studies are needed to understand the precise mechanisms that underlie various effects of physical exercise in substance use disorders.
Collapse
Affiliation(s)
- Marta A Małkiewicz
- Applied Cognitive Neuroscience Lab, Department of Human Physiology, Medical University of Gdansk, Gdansk, Poland; Department of Psychiatry, Medical University of Gdansk, Gdansk, Poland.
| | - Andrzej Małecki
- Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| | - Michal Toborek
- Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland; Department of Biochemistry and Molecular Biology, University of Miami, Miami, USA
| | - Arkadiusz Szarmach
- 2-nd Department of Radiology, Medical University of Gdansk, Gdansk, Poland
| | - Paweł J Winklewski
- 2-nd Department of Radiology, Medical University of Gdansk, Gdansk, Poland; Department of Human Physiology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
14
|
Wei Y, Shah R. Substance Use Disorder in the COVID-19 Pandemic: A Systematic Review of Vulnerabilities and Complications. Pharmaceuticals (Basel) 2020; 13:E155. [PMID: 32708495 PMCID: PMC7407364 DOI: 10.3390/ph13070155] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 01/18/2023] Open
Abstract
As the world endures the coronavirus disease 2019 (COVID-19) pandemic, the conditions of 35 million vulnerable individuals struggling with substance use disorders (SUDs) worldwide have not received sufficient attention for their special health and medical needs. Many of these individuals are complicated by underlying health conditions, such as cardiovascular and lung diseases and undermined immune systems. During the pandemic, access to the healthcare systems and support groups is greatly diminished. Current research on COVID-19 has not addressed the unique challenges facing individuals with SUDs, including the heightened vulnerability and susceptibility to the disease. In this systematic review, we will discuss the pathogenesis and pathology of COVID-19, and highlight potential risk factors and complications to these individuals. We will also provide insights and considerations for COVID-19 treatment and prevention in patients with SUDs.
Collapse
Affiliation(s)
- Yufeng Wei
- Department of Chemistry, New Jersey City University, Jersey City, NJ 07305, USA;
| | | |
Collapse
|
15
|
Moretti M, Belli G, Morini L, Monti MC, Osculati AMM, Visonà SD. Drug Abuse-Related Neuroinflammation in Human Postmortem Brains: An Immunohistochemical Approach. J Neuropathol Exp Neurol 2020; 78:1059-1065. [PMID: 31559425 DOI: 10.1093/jnen/nlz084] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/02/2019] [Accepted: 08/09/2019] [Indexed: 12/27/2022] Open
Abstract
The aim of the study was to investigate blood-brain barrier alterations, neuroinflammation, and glial responses in drug abusers. Five immunohistochemical markers (CD3, zonula occludens-1 [ZO-1], intracellular adhesion molecule 1 [ICAM-1], vascular cell adhesion molecule [VCAM-1], and glial fibrillary acidic protein [GFAP]) were assessed on postmortem brain samples collected from drug abusers who died from acute intoxication of cocaine, heroin, or a combination of both, compared with controls. CD3 and ICAM-1 immunopositivity were significantly stronger in drug abusers than in controls. VCAM-1 immunopositivity was similar across drug abuser and control groups. In heroin abusers, significantly lower ZO-1 immunopositivity was observed relative to controls. GFAP positivity did not show significant differences between groups, but its distribution within the brain did differ. Both cocaine and heroin abuse promoted neuroinflammation, increasing expression of ICAM-1 and recruiting CD3+ lymphocytes. Heroin affected the molecular integrity of tight junctions, as reflected by reduced ZO-1 expression. The outcomes of the present study are, overall, consistent with prior available evidence, which is almost exclusively from studies conducted in vitro or in animal models. These findings provide important information about the downstream consequences of neuroinflammation in drug abusers and may help to inform the development of potential therapeutic targets.
Collapse
Affiliation(s)
| | | | - Luca Morini
- Unit of Legal Medicine and Forensic Sciences
| | | | | | - Silvia Damiana Visonà
- Unit of Biostatistics and Clinical Epidemiology, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
16
|
Barr JL, Brailoiu GC, Abood ME, Rawls SM, Unterwald EM, Brailoiu E. Acute cocaine administration alters permeability of blood-brain barrier in freely-moving rats- Evidence using miniaturized fluorescence microscopy. Drug Alcohol Depend 2020; 206:107637. [PMID: 31734036 PMCID: PMC6980767 DOI: 10.1016/j.drugalcdep.2019.107637] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/17/2019] [Accepted: 09/17/2019] [Indexed: 11/17/2022]
Abstract
BACKGROUND Cocaine has a variety of negative effects on the central nervous system, including reports of decreased barrier function of brain microvascular endothelial cells. However, few studies have directly shown the effects of cocaine on blood-brain barrier (BBB) function in vivo. The miniature integrated fluorescence microscope (i.e., miniscope) technology was used to visualize cocaine-induced changes in BBB permeability in awake, freely-moving rats. METHODS The miniscope was implanted in the prefrontal cortex of adult male rats. After recovery and acclimation, rats received an injection of cocaine (5-20 mg/kg ip) 15 minutes following iv infusion of sodium fluorescein, a low molecular weight tracer. Fluorescence intensity was recordedin vivo via the miniscope for 30 minutes or 24 hours post cocaine administration and served as an indicator of BBB permeability. RESULTS Results demonstrate that cocaine increased the sodium fluorescein extravasation in brain microcirculation in a dose-dependent manner 30 minutes, but not 24 hours after administration. CONCLUSION We report for the first time using direct visualization of brain microcirculation with the miniscope technology in awake, freely-moving rats, that acute cocaine administration produced a transient increase in the BBB permeability.
Collapse
Affiliation(s)
- Jeffrey L Barr
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - G Cristina Brailoiu
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mary E Abood
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Scott M Rawls
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Ellen M Unterwald
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| | - Eugen Brailoiu
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
17
|
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: 36] [Impact Index Per Article: 6.0] [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
|
18
|
HENDERSON LJ, JOHNSON TP, SMITH BR, REOMA LB, SANTAMARIA UA, BACHANI M, DEMARINO C, BARCLAY RA, SNOW J, SACKTOR N, MCARTHUR J, LETENDRE S, STEINER J, KASHANCHI F, NATH A. Presence of Tat and transactivation response element in spinal fluid despite antiretroviral therapy. AIDS 2019; 33 Suppl 2:S145-S157. [PMID: 31789815 PMCID: PMC11032747 DOI: 10.1097/qad.0000000000002268] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The aim of this study was to measure the protein concentration and biological activity of HIV-1 Tat in cerebrospinal fluid (CSF) of individuals on suppressive antiretroviral therapy (ART). DESIGN CSF was collected from 68 HIV-positive individuals on ART with plasma viral load less than 40 copies/ml, and from 25 HIV-negative healthy controls. Duration of HIV infection ranged from 4 to more than 30 years. METHODS Tat levels in CSF were evaluated by an ELISA. Tat protein and viral RNA were quantified from exosomes isolated from CSF, followed by western blot or quantitative reverse transcription PCR, respectively. Functional activity of Tat was assessed using an LTR transactivation assay. RESULTS Tat protein was detected in 36.8% of CSF samples from HIV-positive patients. CSF Tat concentration increased in four out of five individuals after initiation of therapy, indicating that Tat was not inhibited by ART. Similarly, exosomes from 34.4% of CSF samples were strongly positive for Tat protein and/or TAR RNA. Exosomal Tat retained transactivation activity in a CEM-LTR reporter assay in 66.7% of samples assayed, which indicates that over half of the Tat present in CSF is functional. Presence of Tat in CSF was highly associated with previous abuse of psychostimulants (cocaine or amphetamines; P = 0.01) and worse performance in the psychomotor speed (P = 0.04) and information processing (P = 0.02) cognitive domains. CONCLUSION Tat and TAR are produced in the central nervous system despite adequate ART and are packaged into CSF exosomes. Tat remains biologically active within this compartment. These studies suggest that Tat may be a quantifiable marker of the viral reservoir and highlight a need for new therapies that directly inhibit Tat.
Collapse
Affiliation(s)
- Lisa J. HENDERSON
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Tory P. JOHNSON
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bryan R. SMITH
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Lauren Bowen REOMA
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Ulisses A. SANTAMARIA
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Muzna BACHANI
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda Maryland
| | - Catherine DEMARINO
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas Virginia
| | - Robert A. BARCLAY
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas Virginia
| | - Joseph SNOW
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Ned SACKTOR
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Justin MCARTHUR
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Scott LETENDRE
- University of California San Diego School of Medicine, Division of Infectious Diseases and Global Public Health, San Diego California
| | - Joseph STEINER
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda Maryland
| | - Fatah KASHANCHI
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas Virginia
| | - Avindra NATH
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
19
|
Moretti M, Freni F, Valentini B, Vignali C, Groppi A, Visonà SD, Osculati AMM, Morini L. Determination of Antidepressants and Antipsychotics in Dried Blood Spots (DBSs) Collected from Post-Mortem Samples and Evaluation of the Stability over a Three-Month Period. Molecules 2019; 24:molecules24203636. [PMID: 31600953 PMCID: PMC6832719 DOI: 10.3390/molecules24203636] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/04/2019] [Accepted: 10/06/2019] [Indexed: 01/17/2023] Open
Abstract
An LC-MS/MS method for the identification and quantification of antidepressants and antipsychotics was developed on dried blood spots (DBSs). Moreover, analyte stability on DBSs within a 3-month period was monitored. Aliquots of 85 µL of blood from autopsy cases were pipetted onto DBS cards, which were dried and stored at room temperature. DBSs were analyzed in triplicate immediately, within the following 3 weeks, and after 3 months. For each analysis, a whole blood stain was extracted in phosphate buffer and purified using Solid Phase Extraction (SPE) cartridges in order to avoid matrix effects and injected in the LC-MS/MS system. Thirty-nine molecules were screened. Limits of detection (LODs) ranged between 0.1 and 3.2 ng/mL (g) and 0.1 and 5.2 ng/mL (g) for antidepressants and antipsychotics, respectively. Limits of quantification (LOQs) varied from 5 to 10.0 ng/mL for both. Sixteen cases among the 60 analyzed resulted positive for 17 different analytes; for 14 of these the method was fully validated. A general good agreement between the concentrations on DBSs and those measured in conventional blood samples (collected concurrently and stored at -20 °C) was observed. The degradation/enhancement percentage for most of the substances was lower than 20% within the 3-month period. Our results, obtained from real post-mortem cases, suggest that DBSs can be used for routine sample storage.
Collapse
Affiliation(s)
- Matteo Moretti
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Forlanini 12, 27100 Pavia, Italy.
| | - Francesca Freni
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Forlanini 12, 27100 Pavia, Italy.
| | - Beatrice Valentini
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Forlanini 12, 27100 Pavia, Italy.
| | - Claudia Vignali
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Forlanini 12, 27100 Pavia, Italy.
| | - Angelo Groppi
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Forlanini 12, 27100 Pavia, Italy.
| | - Silvia Damiana Visonà
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Forlanini 12, 27100 Pavia, Italy.
| | - Antonio Marco Maria Osculati
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Forlanini 12, 27100 Pavia, Italy.
| | - Luca Morini
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Forlanini 12, 27100 Pavia, Italy.
| |
Collapse
|
20
|
Tiani KA, Stover PJ, Field MS. The Role of Brain Barriers in Maintaining Brain Vitamin Levels. Annu Rev Nutr 2019; 39:147-173. [DOI: 10.1146/annurev-nutr-082018-124235] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
It is increasingly recognized that tissue-specific nutrient deficiencies can exist in the absence of whole-body deficiency and that these deficiencies may result from disease or disease-related physiological processes. Brain and central nervous system tissues require adequate nutrient levels to function. Many nutrients are concentrated in the cerebrospinal fluid relative to the serum in healthy individuals, and other nutrients resist depletion in the presence of whole-body nutrient depletion. The endothelial, epithelial, and arachnoid brain barriers work in concert to selectively transport, concentrate, and maintain levels of the specific nutrients required by the brain while also blocking the passage of blood-borne toxins and pathogens to brain and central nervous system tissues. These barriers preserve nutrient levels within the brain and actively concentrate nutrients within the cerebrospinal fluid and brain. The roles of physical and energetic barriers, including the blood–brain and blood–nerve barriers, in maintaining brain nutrient levels in health and disease are discussed.
Collapse
Affiliation(s)
- Kendra A. Tiani
- Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Patrick J. Stover
- College of Agriculture and Life Sciences, Texas A & M University, College Station, Texas 77843-2142, USA
| | - Martha S. Field
- Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
21
|
Sil S, Niu F, Tom E, Liao K, Periyasamy P, Buch S. Cocaine Mediated Neuroinflammation: Role of Dysregulated Autophagy in Pericytes. Mol Neurobiol 2019; 56:3576-3590. [PMID: 30151726 PMCID: PMC6393223 DOI: 10.1007/s12035-018-1325-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/20/2018] [Indexed: 12/14/2022]
Abstract
Cocaine, a known psychostimulant, results in oxidative stress and inflammation. Recent studies from our group have shown that cocaine induces inflammation in glial cells. Our current study was aimed at investigating whether cocaine exposure could also induce inflammation in non-glial cells such as the pericytes with a focus on the endoplasmic reticulum (ER) stress/autophagy axis. Our in vitro findings demonstrated that exposure of pericytes to cocaine resulted in upregulation of the pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) in both the intracellular as well as extracellular compartments, thus underpinning pericytes as yet another source of neuroinflammation. Cocaine exposure of pericytes resulted in increased formation of autophagosomes as demonstrated by a time-dependent increase of autophagy markers, with a concomitant defect in the fusion of the autophagosome with the lysosomes. Pharmacological blocking of the sigma 1 receptor underscored its role in cocaine-mediated activation of pericytes. Furthermore, it was also demonstrated that cocaine-mediated dysregulation of autophagy involved upstream activation of the ER stress pathways, with a subsequent downstream production of pro-inflammatory cytokines in pericytes. These findings were also validated in an in vivo model wherein pericytes in the isolated brain microvessels of cocaine injected mice (7 days) exhibited increased expression of both the autophagy marker-LC3 as well as the pro-inflammatory cytokine, IL-6. This is the first report describing the role of pericytes in cocaine-mediated neuroinflammation. Interventions aimed at blocking either the sigma-1 receptor or the upstream ER stress mediators could likely be envisioned as promising therapeutic targets for abrogating cocaine-mediated inflammation in pericytes.
Collapse
Affiliation(s)
- Susmita Sil
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Fang Niu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Eric Tom
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Ke Liao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA.
| |
Collapse
|
22
|
Ysrayl BB, Balasubramaniam M, Albert I, Villalta F, Pandhare J, Dash C. A Novel Role of Prolidase in Cocaine-Mediated Breach in the Barrier of Brain Microvascular Endothelial Cells. Sci Rep 2019; 9:2567. [PMID: 30796241 PMCID: PMC6385491 DOI: 10.1038/s41598-018-37495-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 11/26/2018] [Indexed: 02/06/2023] Open
Abstract
Cocaine use is associated with breach in the blood brain barrier (BBB) and increased HIV-1 neuro-invasion. We show that the cellular enzyme "Prolidase" plays a key role in cocaine-induced disruption of the BBB. We established a barrier model to mimic the BBB by culturing human brain microvascular endothelial cells (HBMECs) in transwell inserts. In this model, cocaine treatment enhanced permeability of FITC-dextran suggesting a breach in the barrier. Interestingly, cocaine treatment increased the activity of matrix metallo-proteinases that initiate degradation of the BBB-associated collagen. Cocaine exposure also induced prolidase expression and activity in HBMECs. Prolidase catalyzes the final and rate-limiting step of collagen degradation during BBB remodeling. Knock-down of prolidase abrogated cocaine-mediated increased permeability suggesting a direct role of prolidase in BBB breach. To decipher the mechanism by which cocaine regulates prolidase, we probed the inducible nitric oxide synthase (iNOS) mediated phosphorylation of prolidase since mRNA levels of the protein were not altered upon cocaine treatment. We observed increased iNOS expression concurrent with increased prolidase phosphorylation in cocaine treated cells. Subsequently, inhibition of iNOS decreased prolidase phosphorylation and reduced cocaine-mediated permeability. Finally, cocaine treatment increased transmigration of monocytic cells through the HBMEC barrier. Knock-down of prolidase reduced cocaine-mediated monocyte transmigration, establishing a key role of prolidase in cocaine-induced breach in endothelial cell barrier.
Collapse
Affiliation(s)
- Binah Baht Ysrayl
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA
- Center for Molecular and Behavioral Neurosciences, Meharry Medical College, Nashville, Tennessee, USA
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, USA
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, Tennessee, USA
| | - Muthukumar Balasubramaniam
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA
- Center for Molecular and Behavioral Neurosciences, Meharry Medical College, Nashville, Tennessee, USA
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, Tennessee, USA
| | - Ife Albert
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA
| | - Fernando Villalta
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, Tennessee, USA
| | - Jui Pandhare
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA.
- Center for Molecular and Behavioral Neurosciences, Meharry Medical College, Nashville, Tennessee, USA.
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, USA.
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, Tennessee, USA.
| | - Chandravanu Dash
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA.
- Center for Molecular and Behavioral Neurosciences, Meharry Medical College, Nashville, Tennessee, USA.
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, USA.
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, Tennessee, USA.
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA.
| |
Collapse
|
23
|
Vonder Haar C, Ferland JMN, Kaur S, Riparip LK, Rosi S, Winstanley CA. Cocaine self-administration is increased after frontal traumatic brain injury and associated with neuroinflammation. Eur J Neurosci 2018; 50:2134-2145. [PMID: 30118561 DOI: 10.1111/ejn.14123] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 07/30/2018] [Accepted: 08/07/2018] [Indexed: 12/18/2022]
Abstract
Traumatic brain injury (TBI) has been linked to the development of numerous psychiatric diseases, including substance use disorder. However, it can be difficult to ascertain from clinical data whether the TBI is cause or consequence of increased addiction vulnerability. Surprisingly few studies have taken advantage of animal models to investigate the causal nature of this relationship. In terms of a plausible neurobiological mechanism through which TBI could magnify the risk of substance dependence, numerous studies indicate that TBI can cause widespread disruption to monoaminergic signaling in striatal regions, and also increases neuroinflammation. In the current study, male Long-Evans rats received either a mild or severe TBI centered over the frontal cortex via controlled cortical impact, and were subsequently trained to self-administer cocaine over 10 6-hour sessions. At the end of the study, markers of striatal dopaminergic function, and levels of inflammatory cytokine levels in the frontal lobes, were assessed via western blot and multiplex ELISA, respectively. There was significantly higher cocaine intake in a subset of animals with either mild or severe TBI. However, many animals within both TBI groups failed to acquire self-administration. Principal components analysis suggested that both dopaminergic and neuroinflammatory proteins were associated with overall cocaine intake, yet only an inflammatory component was associated with acquisition of self-administration, suggesting neuroinflammation may make a more substantial contribution to the likelihood of drug-taking. Should neuroinflammation play a causal role in mediating TBI-induced addiction risk, anti-inflammatory therapy may reduce the likelihood of substance abuse in TBI populations.
Collapse
Affiliation(s)
- Cole Vonder Haar
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, PO Box 6040, 53 Campus Drive, Morgantown, WV, 26505, USA.,Laboratory of Molecular and Behavioural Neuroscience, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Jacqueline-Marie N Ferland
- Laboratory of Molecular and Behavioural Neuroscience, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Sukhbir Kaur
- Laboratory of Molecular and Behavioural Neuroscience, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Lara-Kirstie Riparip
- Brain and Spinal Injury Center, Departments of Physical Therapy Rehabilitation Science and Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Susanna Rosi
- Brain and Spinal Injury Center, Departments of Physical Therapy Rehabilitation Science and Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Catharine A Winstanley
- Laboratory of Molecular and Behavioural Neuroscience, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| |
Collapse
|
24
|
Wakim KM, Molloy CJ, Bell RP, Ross LA, Foxe JJ. White Matter Changes in HIV+ Women with a History of Cocaine Dependence. Front Neurol 2017; 8:562. [PMID: 29163330 PMCID: PMC5671562 DOI: 10.3389/fneur.2017.00562] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/09/2017] [Indexed: 01/08/2023] Open
Abstract
Cocaine use is associated with the transmission of human immunodeficiency (HIV) virus through risky sexual behavior. In HIV+ individuals, cocaine use is linked with poor health outcomes, including HIV-medication non-adherence and faster disease progression. Both HIV and cocaine dependence are associated with reduced integrity of cerebral white matter (WM), but the effects of HIV during cocaine abstinence have not yet been explored. We used diffusion tensor imaging (DTI) to understand the effect of combined HIV+ serostatus and former cocaine dependence on cerebral WM integrity. DTI data obtained from 15 HIV+ women with a history of cocaine dependence (COC+/HIV+) and 21 healthy females were included in the analysis. Diffusion-based measures [fractional anisotropy (FA), radial diffusivity (RD), mean diffusivity, and axial diffusivity] were examined using tract-based spatial statistics and region-of-interest analyses. In a whole-brain analysis, COC+/HIV+ women showed significantly reduced FA and increased RD in all major WM tracts, except the left corticospinal tract for RD. The tract with greatest percentage of voxels showing significant between-group differences was the forceps minor (FA: 75.6%, RD: 59.7%). These widespread changes in diffusion measures indicate an extensive neuropathological effect of HIV and former cocaine dependence on WM.
Collapse
Affiliation(s)
- Kathryn-Mary Wakim
- The Cognitive Neurophysiology Laboratory, The Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, Rochester, NY, United States
| | - Ciara J Molloy
- The Cognitive Neurophysiology Laboratory, The Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, Rochester, NY, United States
| | - Ryan P Bell
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, United States.,The Sheryl and Daniel R. Tishman Cognitive Neurophysiology Laboratory, Department of Pediatrics, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States.,The Sheryl and Daniel R. Tishman Cognitive Neurophysiology Laboratory, Department of Neuroscience, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States
| | - Lars A Ross
- The Sheryl and Daniel R. Tishman Cognitive Neurophysiology Laboratory, Department of Pediatrics, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States.,The Sheryl and Daniel R. Tishman Cognitive Neurophysiology Laboratory, Department of Neuroscience, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States
| | - John J Foxe
- The Cognitive Neurophysiology Laboratory, The Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, Rochester, NY, United States.,The Sheryl and Daniel R. Tishman Cognitive Neurophysiology Laboratory, Department of Pediatrics, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States.,The Sheryl and Daniel R. Tishman Cognitive Neurophysiology Laboratory, Department of Neuroscience, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States
| |
Collapse
|
25
|
Wang Z, Shang H, Jiang Y. Chemokines and Chemokine Receptors: Accomplices for Human Immunodeficiency Virus Infection and Latency. Front Immunol 2017; 8:1274. [PMID: 29085362 PMCID: PMC5650658 DOI: 10.3389/fimmu.2017.01274] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/25/2017] [Indexed: 12/22/2022] Open
Abstract
Chemokines are small chemotactic cytokines that are involved in the regulation of immune cell migration. Multiple functional properties of chemokines, such as pro-inflammation, immune regulation, and promotion of cell growth, angiogenesis, and apoptosis, have been identified in many pathological and physiological contexts. Human immunodeficiency virus (HIV) infection is characterized by persistent inflammation and immune activation during both acute and chronic phases, and the "cytokine storm" is one of the hallmarks of HIV infection. Along with immune activation after HIV infection, an extensive range of chemokines and other cytokines are elevated, thereby generating the so-called "cytokine storm." In this review, the effects of the upregulated chemokines and chemokine receptors on the processes of HIV infection are discussed. The objective of this review was to focus on the main chemokines and chemokine receptors that have been found to be associated with HIV infection and latency. Elevated chemokines and chemokine receptors have been shown to play important roles in the HIV life cycle, disease progression, and HIV reservoir establishment. Thus, targeting these chemokines and receptors and the other proteins of related signaling pathways might provide novel therapeutic strategies, and the evidence indicates a promising future regarding the development of a functional cure for HIV.
Collapse
Affiliation(s)
- Zhuo Wang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Hong Shang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Yongjun Jiang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
26
|
Zhang L, Tan J, Jiang X, Qian W, Yang T, Sun X, Chen Z, Zhu Q. Neuron-derived CCL2 contributes to microglia activation and neurological decline in hepatic encephalopathy. Biol Res 2017; 50:26. [PMID: 28870240 PMCID: PMC5584513 DOI: 10.1186/s40659-017-0130-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/24/2017] [Indexed: 01/12/2023] Open
Abstract
Background CCL2 was up-regulated in neurons and involved in microglia activation and neurological decline in mice suffering from hepatic encephalopathy (HE). However, no data exist concerning the effect of neuron-derived CCL2 on microglia activation in vitro. Methods The rats were pretreated with CCL2 receptor inhibitors (INCB or C021, 1 mg/kg/day i.p.) for 3 days prior to thioacetamide (TAA) administration (300 mg/kg/day i.p.) for inducing HE model. At 8 h following the last injection (and every 4 h after), the grade of encephalopathy was assessed. Blood and whole brains were collected at coma for measuring CCL2 and Iba1 expression. In vitro, primary neurons were stimulated with TNF-α, and then the medium were collected for addition to microglia cultures with or without INCB or C021 pretreatment. The effect of the medium on microglia proliferation and activation was evaluated after 24 h. Results CCL2 expression and microglia activation were elevated in the cerebral cortex of rats received TAA alone. CCL2 receptors inhibition improved neurological score and reduced cortical microglia activation. In vitro, TNF-α treatment induced CCL2 release by neurons. Medium from TNF-α stimulated neurons caused microglia proliferation and M1 markers expression, including iNOS, COX2, IL-6 and IL-1β, which could be suppressed by INCB or C021 pretreatment. The medium could also facilitate p65 nuclear translocation and IκBα phosphorylation, and NF-κB inhibition reduced the increased IL-6 and IL-1β expression induced by the medium. Conclusion Neuron-derived CCL2 contributed to microglia activation and neurological decline in HE. Blocking CCL2 or inhibiting microglia excessive activation may be potential strategies for HE.
Collapse
Affiliation(s)
- Li Zhang
- Department of Radiology, The Second People's Hospital of Lanzhou, No. 388 Jingyuan Road, Chengguan District, Lanzhou, 730046, China
| | - Jinyun Tan
- Department of Radiology, The Second People's Hospital of Lanzhou, No. 388 Jingyuan Road, Chengguan District, Lanzhou, 730046, China
| | - Xiaoping Jiang
- Department of Radiology, The Second People's Hospital of Lanzhou, No. 388 Jingyuan Road, Chengguan District, Lanzhou, 730046, China.
| | - Weiwei Qian
- Department of Radiology, The Second People's Hospital of Lanzhou, No. 388 Jingyuan Road, Chengguan District, Lanzhou, 730046, China
| | - Ting Yang
- Department of Radiology, The Second People's Hospital of Lanzhou, No. 388 Jingyuan Road, Chengguan District, Lanzhou, 730046, China
| | - Xijun Sun
- Department of Radiology, The Second People's Hospital of Lanzhou, No. 388 Jingyuan Road, Chengguan District, Lanzhou, 730046, China
| | - Zhaohui Chen
- Department of Radiology, The Second People's Hospital of Lanzhou, No. 388 Jingyuan Road, Chengguan District, Lanzhou, 730046, China
| | - Qiwen Zhu
- Department of Radiology, The Second People's Hospital of Lanzhou, No. 388 Jingyuan Road, Chengguan District, Lanzhou, 730046, China
| |
Collapse
|
27
|
Dalvi P, Spikes L, Allen J, Gupta VG, Sharma H, Gillcrist M, Montes de Oca J, O'Brien-Ladner A, Dhillon NK. Effect of Cocaine on Pulmonary Vascular Remodeling and Hemodynamics in Human Immunodeficiency Virus-Transgenic Rats. Am J Respir Cell Mol Biol 2017; 55:201-12. [PMID: 26820592 DOI: 10.1165/rcmb.2015-0264oc] [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] [Indexed: 11/24/2022] Open
Abstract
Human immunodeficiency virus (HIV)-related pulmonary arterial hypertension has been found to be more prevalent in intravenous drug users. Our earlier cell-culture findings reported down-regulation of bone morphogenetic protein receptors (BMPRs) in combination with enhanced proliferation of human pulmonary arterial smooth muscle cells (PASMCs) in the presence of HIV-Trans-activator of transcription (Tat) and cocaine compared with either treatment alone. Here, we report physiologic evidence of significant increases in mean pulmonary arterial pressure in HIV-transgenic (Tg) rats intraperitoneally administered 40 mg/kg body weight cocaine (HIV-cocaine group) once daily for 21 days when compared with HIV-Tg rats given saline (HIV group) or wild-type (WT) Fischer 334 rats treated with (WT-cocaine group) and without cocaine (WT group). In addition, right ventricle systolic pressure was also found to be significantly higher in the HIV-cocaine rats compared with the WT group. Significant down-regulation in protein expression of BMPR-2 and BMPR-1B was observed in total lung extract from HIV-cocaine rats compared with the other three groups. Furthermore, the PASMCs isolated from HIV-cocaine rats demonstrated a higher level of proliferation and lower levels of apoptosis compared with cells isolated from other rat groups. Interestingly, corroborating our earlier cell-culture findings, we observed higher expression of BMPR-2 and BMPR-1B messenger RNA and significantly lower levels of BMPR-2 and BMPR-1B protein in HIV-cocaine PASMCs compared with cells isolated from all other groups. In conclusion, our findings support an additive effect of cocaine and HIV on smooth muscle dysfunction, resulting in enhanced pulmonary vascular remodeling with associated elevation of mean pulmonary arterial pressure and right ventricle systolic pressure in HIV-Tg rats exposed to cocaine.
Collapse
Affiliation(s)
- Pranjali Dalvi
- 1 Department of Medicine, Division of Pulmonary and Critical Care Medicine, and
| | - Leslie Spikes
- 1 Department of Medicine, Division of Pulmonary and Critical Care Medicine, and
| | - Julie Allen
- 1 Department of Medicine, Division of Pulmonary and Critical Care Medicine, and
| | - Vijayalaxmi G Gupta
- 2 Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Himanshu Sharma
- 1 Department of Medicine, Division of Pulmonary and Critical Care Medicine, and
| | - Marion Gillcrist
- 1 Department of Medicine, Division of Pulmonary and Critical Care Medicine, and
| | | | - Amy O'Brien-Ladner
- 1 Department of Medicine, Division of Pulmonary and Critical Care Medicine, and
| | - Navneet K Dhillon
- 1 Department of Medicine, Division of Pulmonary and Critical Care Medicine, and.,2 Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
28
|
Festoff BW, Sajja RK, van Dreden P, Cucullo L. HMGB1 and thrombin mediate the blood-brain barrier dysfunction acting as biomarkers of neuroinflammation and progression to neurodegeneration in Alzheimer's disease. J Neuroinflammation 2016; 13:194. [PMID: 27553758 PMCID: PMC4995775 DOI: 10.1186/s12974-016-0670-z] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/17/2016] [Indexed: 01/11/2023] Open
Abstract
Background The blood-brain barrier (BBB) dysfunction represents an early feature of Alzheimer’s disease (AD) that precedes the hallmarks of amyloid beta (amyloid β) plaque deposition and neuronal neurofibrillary tangle (NFT) formation. A damaged BBB correlates directly with neuroinflammation involving microglial activation and reactive astrogliosis, which is associated with increased expression and/or release of high-mobility group box protein 1 (HMGB1) and thrombin. However, the link between the presence of these molecules, BBB damage, and progression to neurodegeneration in AD is still elusive. Therefore, we aimed to profile and validate non-invasive clinical biomarkers of BBB dysfunction and neuroinflammation to assess the progression to neurodegeneration in mild cognitive impairment (MCI) and AD patients. Methods We determined the serum levels of various proinflammatory damage-associated molecules in aged control subjects and patients with MCI or AD using validated ELISA kits. We then assessed the specific and direct effects of such molecules on BBB integrity in vitro using human primary brain microvascular endothelial cells or a cell line. Results We observed a significant increase in serum HMGB1 and soluble receptor for advanced glycation end products (sRAGE) that correlated well with amyloid beta levels in AD patients (vs. control subjects). Interestingly, serum HMGB1 levels were significantly elevated in MCI patients compared to controls or AD patients. In addition, as a marker of BBB damage, soluble thrombomodulin (sTM) antigen, and activity were significantly (and distinctly) increased in MCI and AD patients. Direct in vitro BBB integrity assessment further revealed a significant and concentration-dependent increase in paracellular permeability to dextrans by HMGB1 or α-thrombin, possibly through disruption of zona occludins-1 bands. Pre-treatment with anti-HMGB1 monoclonal antibody blocked HMGB1 effects and leaving BBB integrity intact. Conclusions Our current studies indicate that thrombin and HMGB1 are causal proximate proinflammatory mediators of BBB dysfunction, while sTM levels may indicate BBB endothelial damage; HMGB1 and sRAGE might serve as clinical biomarkers for progression and/or therapeutic efficacy along the AD spectrum.
Collapse
Affiliation(s)
- Barry W Festoff
- pHLOGISTIX LLC, 4220 Shawnee Mission Parkway, Fairway, KS, 66205, USA.,Department of Neurology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Ravi K Sajja
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Patrick van Dreden
- Clinical Research Department, R&D, Diagnostica Stago, Gennevilliers, France
| | - Luca Cucullo
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA.
| |
Collapse
|
29
|
Sajja RK, Rahman S, Cucullo L. Drugs of abuse and blood-brain barrier endothelial dysfunction: A focus on the role of oxidative stress. J Cereb Blood Flow Metab 2016; 36:539-54. [PMID: 26661236 PMCID: PMC4794105 DOI: 10.1177/0271678x15616978] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 10/13/2015] [Indexed: 02/01/2023]
Abstract
Psychostimulants and nicotine are the most widely abused drugs with a detrimental impact on public health globally. While the long-term neurobehavioral deficits and synaptic perturbations are well documented with chronic use of methamphetamine, cocaine, and nicotine, emerging human and experimental studies also suggest an increasing incidence of neurovascular complications associated with drug abuse. Short- or long-term administration of psychostimulants or nicotine is known to disrupt blood-brain barrier (BBB) integrity/function, thus leading to an increased risk of brain edema and neuroinflammation. Various pathophysiological mechanisms have been proposed to underlie drug abuse-induced BBB dysfunction suggesting a central and unifying role for oxidative stress in BBB endothelium and perivascular cells. This review discusses drug-specific effects of methamphetamine, cocaine, and tobacco smoking on brain microvascular crisis and provides critical assessment of oxidative stress-dependent molecular pathways focal to the global compromise of BBB. Additionally, given the increased risk of human immunodeficiency virus (HIV) encephalitis in drug abusers, we have summarized the synergistic pathological impact of psychostimulants and HIV infection on BBB integrity with an emphasis on unifying role of endothelial oxidative stress. This mechanistic framework would guide further investigations on specific molecular pathways to accelerate therapeutic approaches for the prevention of neurovascular deficits by drugs of abuse.
Collapse
Affiliation(s)
- Ravi K Sajja
- Center for Blood-Brain Barrier Research, Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, SD, USA
| | - Luca Cucullo
- Center for Blood-Brain Barrier Research, Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| |
Collapse
|
30
|
Brailoiu GC, Deliu E, Console-Bram LM, Soboloff J, Abood ME, Unterwald EM, Brailoiu E. Cocaine inhibits store-operated Ca2+ entry in brain microvascular endothelial cells: critical role for sigma-1 receptors. Biochem J 2016; 473:1-5. [PMID: 26467159 PMCID: PMC4679692 DOI: 10.1042/bj20150934] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 10/14/2015] [Indexed: 01/29/2023]
Abstract
Sigma-1 receptor (Sig-1R) is an intracellular chaperone protein with many ligands, located at the endoplasmic reticulum (ER). Binding of cocaine to Sig-1R has previously been found to modulate endothelial functions. In the present study, we show that cocaine dramatically inhibits store-operated Ca(2+) entry (SOCE), a Ca(2+) influx mechanism promoted by depletion of intracellular Ca(2+) stores, in rat brain microvascular endothelial cells (RBMVEC). Using either Sig-1R shRNA or pharmacological inhibition with the unrelated Sig-1R antagonists BD-1063 and NE-100, we show that cocaine-induced SOCE inhibition is dependent on Sig-1R. In addition to revealing new insight into fundamental mechanisms of cocaine-induced changes in endothelial function, these studies indicate an unprecedented role for Sig-1R as a SOCE inhibitor.
Collapse
Affiliation(s)
- G Cristina Brailoiu
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, U.S.A
| | - Elena Deliu
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Linda M Console-Bram
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Jonathan Soboloff
- Fels Institute for Cancer Research and Molecular Biology and Department of Medical Genetics & Molecular Biochemistry, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Mary E Abood
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A. Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Ellen M Unterwald
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A. Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Eugen Brailoiu
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A.
| |
Collapse
|
31
|
Dalvi PN, Gupta VG, Griffin BR, O'Brien-Ladner A, Dhillon NK. Ligand-Independent Activation of Platelet-Derived Growth Factor Receptor β during Human Immunodeficiency Virus-Transactivator of Transcription and Cocaine-Mediated Smooth Muscle Hyperplasia. Am J Respir Cell Mol Biol 2015; 53:336-45. [PMID: 25569182 DOI: 10.1165/rcmb.2014-0369oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Our previous study supports an additive effect of cocaine to human immunodeficiency virus infection in the development of pulmonary arteriopathy through enhancement of proliferation of pulmonary smooth muscle cells (SMCs), while also suggesting involvement of platelet-derived growth factor receptor (PDGFR) activation in the absence of further increase in PDGF-BB ligand. Redox-related signaling pathways have been shown to regulate tyrosine kinase receptors independent of ligand binding, so we hypothesized that simultaneous treatment of SMCs with transactivator of transcription (Tat) and cocaine may be able to indirectly activate PDGFR through modulation of reactive oxygen species (ROS) without the need for PDGF binding. We found that blocking the binding of ligand using suramin or monoclonal IMC-3G3 antibody significantly reduced ligand-induced autophosphorylation of Y1009 without affecting ligand-independent transphosphorylation of Y934 residue on PDGFRβ in human pulmonary arterial SMCs treated with both cocaine and Tat. Combined treatment of human pulmonary arterial SMCs with cocaine and Tat resulted in augmented production of superoxide radicals and hydrogen peroxide when compared with either treatment alone. Inhibition of this ROS generation prevented cocaine- and Tat-mediated Src activation and transphosphorylation of PDGFRβ at Y934 without any changes in phosphorylation of Y1009, in addition to attenuation of smooth muscle hyperplasia. Furthermore, pretreatment with an Src inhibitor, PP2, also suppressed cocaine- and Tat-mediated enhanced Y934 phosphorylation and smooth muscle proliferation. Finally, we report total abrogation of cocaine- and Tat-mediated synergistic increase in cell proliferation on inhibition of both ligand-dependent and ROS/Src-mediated ligand-independent phosphorylation of PDGFRβ.
Collapse
Affiliation(s)
| | - Vijayalaxmi G Gupta
- 2 Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | | | | | - Navneet K Dhillon
- Departments of 1 Internal Medicine and.,2 Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
32
|
Dash S, Balasubramaniam M, Villalta F, Dash C, Pandhare J. Impact of cocaine abuse on HIV pathogenesis. Front Microbiol 2015; 6:1111. [PMID: 26539167 PMCID: PMC4611962 DOI: 10.3389/fmicb.2015.01111] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 09/25/2015] [Indexed: 12/13/2022] Open
Abstract
Over 1.2 million people in the United States are infected with the human immunodeficiency virus type 1 (HIV-1). Tremendous progress has been made over the past three decades on many fronts in the prevention and treatment of HIV-1 disease. However, HIV-1 infection is incurable and antiretroviral drugs continue to remain the only effective treatment option for HIV infected patients. Unfortunately, only three out of ten HIV-1 infected individuals in the US have the virus under control. Thus, majority of HIV-1 infected individuals in the US are either unaware of their infection status or not connected/retained to care or are non-adherent to antiretroviral therapy (ART). This national public health crisis, as well as the ongoing global HIV/AIDS pandemic, is further exacerbated by substance abuse, which serves as a powerful cofactor at every stage of HIV/AIDS including transmission, diagnosis, pathogenesis, and treatment. Clinical studies indicate that substance abuse may increase viral load, accelerate disease progression and worsen AIDS-related mortality even among ART-adherent patients. However, confirming a direct causal link between substance abuse and HIV/AIDS in human patients remains a highly challenging endeavor. In this review we will discuss the recent and past developments in clinical and basic science research on the effects of cocaine abuse on HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Sabyasachi Dash
- Center for AIDS Health Disparities Research, Meharry Medical College , Nashville, TN, USA ; School of Graduate Studies and Research, Meharry Medical College , Nashville, TN, USA
| | - Muthukumar Balasubramaniam
- Center for AIDS Health Disparities Research, Meharry Medical College , Nashville, TN, USA ; Department of Biochemistry and Cancer Biology, Meharry Medical College , Nashville, TN, USA
| | - Fernando Villalta
- Center for AIDS Health Disparities Research, Meharry Medical College , Nashville, TN, USA ; School of Graduate Studies and Research, Meharry Medical College , Nashville, TN, USA ; Department of Microbiology and Immunology, Meharry Medical College , Nashville, TN, USA
| | - Chandravanu Dash
- Center for AIDS Health Disparities Research, Meharry Medical College , Nashville, TN, USA ; School of Graduate Studies and Research, Meharry Medical College , Nashville, TN, USA ; Department of Biochemistry and Cancer Biology, Meharry Medical College , Nashville, TN, USA
| | - Jui Pandhare
- Center for AIDS Health Disparities Research, Meharry Medical College , Nashville, TN, USA ; School of Graduate Studies and Research, Meharry Medical College , Nashville, TN, USA ; Department of Microbiology and Immunology, Meharry Medical College , Nashville, TN, USA
| |
Collapse
|
33
|
Zhang X, Jiang S, Yu J, Kuzontkoski PM, Groopman JE. Cocaine enhances HIV-1 gp120-induced lymphatic endothelial dysfunction in the lung. Physiol Rep 2015; 3:3/8/e12482. [PMID: 26311830 PMCID: PMC4562568 DOI: 10.14814/phy2.12482] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pulmonary complications are common in both AIDS patients and cocaine users. We addressed the cellular and molecular mechanisms by which HIV and cocaine may partner to induce their deleterious effects. Using primary lung lymphatic endothelial cells (L-LECs), we examined how cocaine and HIV-1 gp120, alone and together, modulate signaling and functional properties of L-LECs. We found that brief cocaine exposure activated paxillin and induced cytoskeletal rearrangement, while sustained exposure increased fibronectin (FN) expression, decreased Robo4 expression, and enhanced the permeability of L-LEC monolayers. Moreover, incubating L-LECs with both cocaine and HIV-1 gp120 exacerbated hyperpermeability, significantly enhanced apoptosis, and further impaired in vitro wound healing as compared with cocaine alone. Our studies also suggested that the sigma-1 receptor (Sigma-1R) and the dopamine-4 receptor (D4R) are involved in cocaine-induced pathology in L-LECs. Seeking clinical correlation, we found that FN levels in sera and lung tissue of HIV(+) donors were significantly elevated as compared to HIV(-) donors. Our in vitro data demonstrate that cocaine and HIV-1 gp120 induce dysfunction and damage of lung lymphatics, and suggest that cocaine use may exacerbate pulmonary edema and fibrosis associated with HIV infection. Continued exploration of the interplay between cocaine and HIV should assist the design of therapeutics to ameliorate HIV-induced pulmonary disorders within the drug using population.
Collapse
Affiliation(s)
- Xuefeng Zhang
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center Harvard Medical School, Boston, Massachusetts, USA
| | - Susan Jiang
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center Harvard Medical School, Boston, Massachusetts, USA
| | - Jinlong Yu
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center Harvard Medical School, Boston, Massachusetts, USA Department of Psychiatry, Mclean Hospital Harvard Medical School, Belmont, Massachusetts, USA
| | - Paula M Kuzontkoski
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center Harvard Medical School, Boston, Massachusetts, USA DynaMed, EBSCO Information Services, Ipswich, Massachusetts, USA
| | - Jerome E Groopman
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Gill AJ, Kolson DL. Chronic inflammation and the role for cofactors (hepatitis C, drug abuse, antiretroviral drug toxicity, aging) in HAND persistence. Curr HIV/AIDS Rep 2015; 11:325-35. [PMID: 24929842 DOI: 10.1007/s11904-014-0210-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
HIV-associated neurocognitive disorders (HAND) is a group of syndromes of varying degrees of cognitive impairment affecting up to 50 % of HIV-infected individuals. The neuropathogenesis of HAND is thought to be driven by HIV invasion and productive replication within brain perivascular macrophages and endogenous microglia, and to some degree by restricted infection of astrocytes. The persistence of HAND in individuals experiencing suppression of systemic HIV viral load with antiretroviral therapy (ART) is incompletely explained, and suggested factors include chronic inflammation, persistent HIV replication in brain macrophages, effects of aging on brain vulnerability, and co-morbid conditions including hepatitis C (HCV) co-infection, substance abuse, and CNS toxicity of ART, among other factors. This review discusses several of these conditions: chronic inflammation, co-infection with HCV, drugs of abuse, aging, and antiretroviral drug effects. Effectively managing these co-morbid conditions in individuals with and without HAND is critical for improving neurocognitive outcomes and decreasing HIV-associated morbidity.
Collapse
Affiliation(s)
- Alexander J Gill
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard 280C Clinical Research Building, Philadelphia, PA, 19104, USA,
| | | |
Collapse
|
35
|
Abstract
Antiretroviral therapy extends the lifespan of human immunodeficiency virus (HIV)-infected patients, but many survivors develop premature impairments in cognition. These residual cognitive impairments may involve aberrant deposition of amyloid β-peptides (Aβ). By unknown mechanisms, Aβ accumulates in the lysosomal and autophagic compartments of neurons in the HIV-infected brain. Here we identify the molecular events evoked by the HIV coat protein gp120 that facilitate the intraneuronal accumulation of Aβ. We created a triple transgenic gp120/APP/PS1 mouse that recapitulates intraneuronal deposition of Aβ in a manner reminiscent of the HIV-infected brain. In cultured neurons, we found that the HIV coat protein gp120 increased the transcriptional expression of BACE1 through repression of PPARγ, and increased APP expression by promoting interaction of the translation-activating RBP heterogeneous nuclear ribonucleoprotein C with APP mRNA. APP and BACE1 were colocalized into stabilized membrane microdomains, where the β-cleavage of APP and Aβ formation were enhanced. Aβ-peptides became localized to lysosomes that were engorged with sphingomyelin and calcium. Stimulating calcium efflux from lysosomes with a TRPM1 agonist promoted calcium efflux, luminal acidification, and cleared both sphingomyelin and Aβ from lysosomes. These findings suggest that therapeutics targeted to reduce lysosomal pH in neurodegenerative conditions may protect neurons by facilitating the clearance of accumulated sphingolipids and Aβ-peptides.
Collapse
|
36
|
Zenón F, Segarra AC, Gonzalez M, Meléndez LM. Cocaine potentiates cathepsin B secretion and neuronal apoptosis from HIV-infected macrophages. J Neuroimmune Pharmacol 2014; 9:703-15. [PMID: 25209871 PMCID: PMC4209444 DOI: 10.1007/s11481-014-9563-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 08/17/2014] [Indexed: 12/16/2022]
Abstract
Substance abuse is a risk factor for HIV infection and progression to AIDS. Recent evidence establishes that cocaine use promotes brain perivascular macrophage infiltration and microglia activation. The lysosomal protease cathepsin B is increased in monocytes from patients with HIV dementia and its secretion induces 10-15% of neurotoxicity. Here we asked if cocaine potentiates cathepsin B secretion from HIV-infected monocyte-derived macrophages (MDM) and its effect in neuronal apoptosis. Samples of plasma, CSF, and post-mortem brain tissue from HIV positive patients that used cocaine were tested for cathepsin B and its inhibitors to determine the in vivo relevance of these findings. MDM were inoculated with HIV-1ADA, exposed to cocaine, and the levels of secreted and bioactive cathepsin B and its inhibitors were measured at different time-points. Cathepsin B expression (p < 0.001) and activity (p < 0.05) increased in supernatants from HIV-infected cocaine treated MDM compared with HIV-infected cocaine negative controls. Increased levels of cystatin B expression was also found in supernatants from HIV-cocaine treated MDM (p < 0.05). A significant increase in 30% of apoptotic neurons was obtained that decreased to 5% with the specific cathepsin B inhibitor (CA-074) or with cathepsin B antibody. Cathepsin B was significantly increased in the plasma and post-mortem brain tissue of HIV/cocaine users over non-drug users. Our results demonstrated that cocaine potentiates cathepsin B secretion in HIV-infected MDM and increase neuronal apoptosis. These findings provide new evidence that cocaine synergize with HIV-1 infection in increasing cathepsin B secretion and neurotoxicity.
Collapse
Affiliation(s)
- Frances Zenón
- Department of Microbiology and Medical Zoology, School of Medicine, University of Puerto Rico
- NeuroAIDS Program, Medical Sciences Campus, San Juan PR
| | | | | | - Loyda M. Meléndez
- Department of Microbiology and Medical Zoology, School of Medicine, University of Puerto Rico
- NeuroAIDS Program, Medical Sciences Campus, San Juan PR
| |
Collapse
|
37
|
Sáez CG, Pereira-Flores K, Ebensperger R, Panes O, Massardo T, Hidalgo P, Mezzano D, Pereira J. Atorvastatin reduces the proadhesive and prothrombotic endothelial cell phenotype induced by cocaine and plasma from cocaine consumers in vitro. Arterioscler Thromb Vasc Biol 2014; 34:2439-48. [PMID: 25234816 DOI: 10.1161/atvbaha.114.304535] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Cocaine consumption is a risk factor for vascular ischemic complications. Although endothelial dysfunction and accelerated atherosclerosis have been observed in cocaine consumers, the mechanisms underlying their pathogenesis are not fully understood. This study aimed at identifying the effects of atorvastatin in relation to a proadhesive and prothrombotic phenotype induced by cocaine and plasma from chronic cocaine users on endothelial cells. APPROACH AND RESULTS Human umbilical vein endothelial cells were exposed to either cocaine or platelet-free plasma (PFP) from chronic cocaine consumers in the presence or absence of 10 μmol/L of atorvastatin. Atorvastatin significantly reduced the enhanced platelet adhesion that was induced by cocaine and PFP from chronic cocaine consumers, as well as the release of the von Willebrand factor. Atorvastatin also avoided striking alterations on cell monolayer structure triggered by both stimuli and enhanced NO reduction because of cocaine stimulation through disrupting interactions between endothelial nitric oxide synthase (eNOS) and caveolin-1, thus increasing eNOS bioavailability. Cocaine-increased tissue factor-dependent procoagulant activity and reactive oxygen species generation were not counteracted by atorvastatin. Although monocyte chemoattractant protein-1 levels were not significantly higher than controls either under cocaine or PFP stimulation, atorvastatin completely avoided monocyte chemoattractant protein-1 release in both conditions. Platelets stimulated with cocaine or PFP did not express P-selectin, glycoprotein IIb/IIIa, or CD40L and failed to adhere to resting human umbilical vein endothelial cell. CONCLUSIONS Cocaine and patient plasma equally induced a proadhesive and prothrombotic phenotype in endothelial cells, except for von Willebrand Factor release, which was only induced by PFP from chronic cocaine consumers. Atorvastatin improved endothelial cell function by reducing cocaine-induced and PFP from chronic cocaine consumer-induced effects on platelet adhesion, cell architecture, and NO production.
Collapse
Affiliation(s)
- Claudia G Sáez
- From the Hematology-Oncology Department, Faculty of Medicine (C.G.S., K.P.-F., O.P., P.H., D.M., J.P.) and Pharmacy Department, Chemistry Faculty (R.E.), Pontificia Universidad Católica de Chile, Santiago, Chile; and Nuclear Medicine Section, Medicine Department, University of Chile Clinical Hospital, Santiago, Chile (T.M.).
| | - Karla Pereira-Flores
- From the Hematology-Oncology Department, Faculty of Medicine (C.G.S., K.P.-F., O.P., P.H., D.M., J.P.) and Pharmacy Department, Chemistry Faculty (R.E.), Pontificia Universidad Católica de Chile, Santiago, Chile; and Nuclear Medicine Section, Medicine Department, University of Chile Clinical Hospital, Santiago, Chile (T.M.)
| | - Roberto Ebensperger
- From the Hematology-Oncology Department, Faculty of Medicine (C.G.S., K.P.-F., O.P., P.H., D.M., J.P.) and Pharmacy Department, Chemistry Faculty (R.E.), Pontificia Universidad Católica de Chile, Santiago, Chile; and Nuclear Medicine Section, Medicine Department, University of Chile Clinical Hospital, Santiago, Chile (T.M.)
| | - Olga Panes
- From the Hematology-Oncology Department, Faculty of Medicine (C.G.S., K.P.-F., O.P., P.H., D.M., J.P.) and Pharmacy Department, Chemistry Faculty (R.E.), Pontificia Universidad Católica de Chile, Santiago, Chile; and Nuclear Medicine Section, Medicine Department, University of Chile Clinical Hospital, Santiago, Chile (T.M.)
| | - Teresa Massardo
- From the Hematology-Oncology Department, Faculty of Medicine (C.G.S., K.P.-F., O.P., P.H., D.M., J.P.) and Pharmacy Department, Chemistry Faculty (R.E.), Pontificia Universidad Católica de Chile, Santiago, Chile; and Nuclear Medicine Section, Medicine Department, University of Chile Clinical Hospital, Santiago, Chile (T.M.)
| | - Patricia Hidalgo
- From the Hematology-Oncology Department, Faculty of Medicine (C.G.S., K.P.-F., O.P., P.H., D.M., J.P.) and Pharmacy Department, Chemistry Faculty (R.E.), Pontificia Universidad Católica de Chile, Santiago, Chile; and Nuclear Medicine Section, Medicine Department, University of Chile Clinical Hospital, Santiago, Chile (T.M.)
| | - Diego Mezzano
- From the Hematology-Oncology Department, Faculty of Medicine (C.G.S., K.P.-F., O.P., P.H., D.M., J.P.) and Pharmacy Department, Chemistry Faculty (R.E.), Pontificia Universidad Católica de Chile, Santiago, Chile; and Nuclear Medicine Section, Medicine Department, University of Chile Clinical Hospital, Santiago, Chile (T.M.)
| | - Jaime Pereira
- From the Hematology-Oncology Department, Faculty of Medicine (C.G.S., K.P.-F., O.P., P.H., D.M., J.P.) and Pharmacy Department, Chemistry Faculty (R.E.), Pontificia Universidad Católica de Chile, Santiago, Chile; and Nuclear Medicine Section, Medicine Department, University of Chile Clinical Hospital, Santiago, Chile (T.M.).
| |
Collapse
|
38
|
Paul D, Ge S, Lemire Y, Jellison ER, Serwanski DR, Ruddle NH, Pachter JS. Cell-selective knockout and 3D confocal image analysis reveals separate roles for astrocyte-and endothelial-derived CCL2 in neuroinflammation. J Neuroinflammation 2014; 11:10. [PMID: 24444311 PMCID: PMC3906899 DOI: 10.1186/1742-2094-11-10] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/28/2013] [Indexed: 12/30/2022] Open
Abstract
Background Expression of chemokine CCL2 in the normal central nervous system (CNS) is nearly undetectable, but is significantly upregulated and drives neuroinflammation during experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis which is considered a contributing factor in the human disease. As astrocytes and brain microvascular endothelial cells (BMEC) forming the blood–brain barrier (BBB) are sources of CCL2 in EAE and other neuroinflammatory conditions, it is unclear if one or both CCL2 pools are critical to disease and by what mechanism(s). Methods Mice with selective CCL2 gene knockout (KO) in astrocytes (Astro KO) or endothelial cells (Endo KO) were used to evaluate the respective contributions of these sources to neuroinflammation, i.e., clinical disease progression, BBB damage, and parenchymal leukocyte invasion in a myelin oligodendrocyte glycoprotein peptide (MOG35-55)-induced EAE model. High-resolution 3-dimensional (3D) immunofluorescence confocal microscopy and colloidal gold immuno-electron microscopy were employed to confirm sites of CCL2 expression, and 3D immunofluorescence confocal microscopy utilized to assess inflammatory responses along the CNS microvasculature. Results Cell-selective loss of CCL2 immunoreactivity was demonstrated in the respective KO mice. Compared to wild-type (WT) mice, Astro KO mice showed reduced EAE severity but similar onset, while Endo KO mice displayed near normal severity but significantly delayed onset. Neither of the KO mice showed deficits in T cell proliferation, or IL-17 and IFN-γ production, following MOG35-55 exposure in vitro, or altered MOG-major histocompatibility complex class II tetramer binding. 3D confocal imaging further revealed distinct actions of the two CCL2 pools in the CNS. Astro KOs lacked the CNS leukocyte penetration and disrupted immunostaining of CLN-5 at the BBB seen during early EAE in WT mice, while Endo KOs uniquely displayed leukocytes stalled in the microvascular lumen. Conclusions These results point to astrocyte and endothelial pools of CCL2 each regulating different stages of neuroinflammation in EAE, and carry implications for drug delivery in neuroinflammatory disease.
Collapse
Affiliation(s)
| | - Shujun Ge
- Department of Cell Biology, Blood-brain Barrier Laboratory, 263 Farmington Ave,, Farmington CT 06030, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Gonçalves J, Baptista S, Silva AP. Psychostimulants and brain dysfunction: a review of the relevant neurotoxic effects. Neuropharmacology 2014; 87:135-49. [PMID: 24440369 DOI: 10.1016/j.neuropharm.2014.01.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 12/18/2013] [Accepted: 01/06/2014] [Indexed: 12/21/2022]
Abstract
Psychostimulants abuse is a major public concern because is associated with serious health complications, including devastating consequences on the central nervous system (CNS). The neurotoxic effects of these drugs have been extensively studied. Nevertheless, numerous questions and uncertainties remain in our understanding of these toxic events. Thus, the purpose of the present manuscript is to review cellular and molecular mechanisms that might be responsible for brain dysfunction induced by psychostimulants. Topics reviewed include some classical aspects of neurotoxicity, such as monoaminergic system and mitochondrial dysfunction, oxidative stress, excitotoxicity and hyperthermia. Moreover, recent literature has suggested new phenomena regarding the toxic effects of psychostimulants. Thus, we also reviewed the impact of these drugs on neuroinflammatory response, blood-brain barrier (BBB) function and neurogenesis. Assessing the relative importance of these mechanisms on psychostimulants-induced brain dysfunction presents an exciting challenge for future research efforts. This article is part of the Special Issue entitled 'CNS Stimulants'.
Collapse
Affiliation(s)
- Joana Gonçalves
- Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548 Coimbra Portugal
| | - Sofia Baptista
- Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548 Coimbra Portugal
| | - Ana Paula Silva
- Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548 Coimbra Portugal.
| |
Collapse
|
40
|
Dalvi P, Wang K, Mermis J, Zeng R, Sanderson M, Johnson S, Dai Y, Sharma G, Ladner AO, Dhillon NK. HIV-1/cocaine induced oxidative stress disrupts tight junction protein-1 in human pulmonary microvascular endothelial cells: role of Ras/ERK1/2 pathway. PLoS One 2014; 9:e85246. [PMID: 24409324 PMCID: PMC3883699 DOI: 10.1371/journal.pone.0085246] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 11/25/2013] [Indexed: 01/08/2023] Open
Abstract
Intravenous drug use (IVDU) is the major risk factor in the development of HIV-related pulmonary arterial hypertension (HRPAH); however, the pathogenesis of HRPAH in association with IVDU has yet to be characterized. Endothelial injury is considered to be an initiating factor for pulmonary vascular remodeling in animal models of PAH. Our previous study shows that simultaneous exposure to HIV-Trans-activator of transcription (Tat) and cocaine exacerbates both disruption of tight junction proteins and permeability of human pulmonary artery endothelial cells compared with either treatment alone. We here now demonstrate that this HIV-Tat and cocaine mediated endothelial dysfunction accompanies with increase in hydrogen peroxide and superoxide radicals generation and involves redox sensitive signaling pathway. Pretreatment with antioxidant cocktail attenuated the cocaine and Tat mediated disassembly of Zonula Occludens (ZO)-1 and enhancement of endothelial monolayer permeability. Furthermore, inhibition of NADPH oxidase by apocynin or siRNA-mediated knockdown of gp-91(phox) abolished the Tat/cocaine-induced reactive oxygen species (ROS) production, suggesting the NADPH oxidase mediated generation of oxidative radicals. In addition, ROS dependent activation of Ras and ERK1/2 Kinase was observed to be mediating the TJP-1 disassembly, and endothelial dysfunction in response to cocaine and Tat exposure. In conclusion, our findings demonstrate that Tat/cocaine -mediated production of ROS activate Ras/Raf/ERK1/2 pathway that contributes to disruption of tight junction protein leading to pulmonary endothelial dysfunction associated with pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Pranjali Dalvi
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Kun Wang
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Joel Mermis
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Ruoxi Zeng
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Miles Sanderson
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Sara Johnson
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Yuqiao Dai
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Garima Sharma
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Amy O’Brien Ladner
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Navneet K. Dhillon
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail:
| |
Collapse
|
41
|
Impact of opiate addiction on neuroinflammation in HIV. J Neurovirol 2012; 18:364-73. [PMID: 22797933 DOI: 10.1007/s13365-012-0118-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 06/08/2012] [Accepted: 06/08/2012] [Indexed: 12/27/2022]
Abstract
To investigate the independent and interactive effects of opiate addiction and HIV on neuroinflammation, we measured microglial/macrophage activation and astrogliosis in multiple regions of human brain. Samples of thalamus, frontal gray matter, and frontal white matter were obtained from 46 individuals categorized as: HIV negatives, HIV-negative opiate addicts, HIV positives, HIV-positive opiate addicts, HIV encephalitis (HIVE), and HIVE opiate addicts. Activated brain microglia/macrophages and astrocytosis were quantified by morphometric analysis of immunohistochemical stains for CD68, HLA-D, CD163, and GFAP. The effects of HIV grouping, opiate addiction, and their interaction on expression of the markers were examined in a series of two-way ANOVAs. In opiate addicts, there was generally higher baseline expression of CD68 and HLA-D in HIV negatives, and lower expression in HIV and HIVE, compared to individuals without opiate abuse. Thus, for these markers, and for GFAP in frontal gray, opiates were associated with attenuated HIV effect. In contrast, for CD163, opiates did not significantly alter responses to HIV, and HIV effects were variably absent in individuals without opiate abuse. The divergent impact that opiate addiction displays on these markers may suggest a generally immunosuppressive role in the CNS, with decreased HIV-associated activation of markers CD68 and HLA-D that potentially reflect neurotoxic pathways, and preservation of CD163, thought to be an indicator of neuroprotective scavenger systems. These results suggest a complex impact of opiates on neuroinflammation in baseline and virally stimulated states.
Collapse
|
42
|
Kousik SM, Napier TC, Carvey PM. The effects of psychostimulant drugs on blood brain barrier function and neuroinflammation. Front Pharmacol 2012; 3:121. [PMID: 22754527 PMCID: PMC3386512 DOI: 10.3389/fphar.2012.00121] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 06/06/2012] [Indexed: 12/17/2022] Open
Abstract
The blood brain barrier (BBB) is a highly dynamic interface between the central nervous system (CNS) and periphery. The BBB is comprised of a number of components and is part of the larger neuro(glio)vascular unit. Current literature suggests that psychostimulant drugs of abuse alter the function of the BBB which likely contributes to the neurotoxicities associated with these drugs. In both preclinical and clinical studies, psychostimulants including methamphetamine, MDMA, cocaine, and nicotine, produce BBB dysfunction through alterations in tight junction protein expression and conformation, increased glial activation, increased enzyme activation related to BBB cytoskeleton remodeling, and induction of neuroinflammatory pathways. These detrimental changes lead to increased permeability of the BBB and subsequent vulnerability of the brain to peripheral toxins. In fact, abuse of these psychostimulants, notably methamphetamine and cocaine, has been shown to increase the invasion of peripheral bacteria and viruses into the brain. Much work in this field has focused on the co-morbidity of psychostimulant abuse and human immunodeficiency virus (HIV) infection. As psychostimulants alter BBB permeability, it is likely that this BBB dysfunction results in increased penetration of the HIV virus into the brain thus increasing the risk of and severity of neuro AIDS. This review will provide an overview of the specific changes in components within the BBB associated with psychostimulant abuse as well as the implications of these changes in exacerbating the neuropathology associated with psychostimulant drugs and HIV co-morbidity.
Collapse
Affiliation(s)
- Sharanya M Kousik
- Department of Pharmacology, Rush University Medical Center Chicago, IL, USA
| | | | | |
Collapse
|
43
|
Liu WY, Wang ZB, Zhang LC, Wei X, Li L. Tight junction in blood-brain barrier: an overview of structure, regulation, and regulator substances. CNS Neurosci Ther 2012; 18:609-15. [PMID: 22686334 DOI: 10.1111/j.1755-5949.2012.00340.x] [Citation(s) in RCA: 202] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Blood-brain barrier (BBB) is a dynamic interference that regulates the nutrition and toxic substance in and out of the central nervous system (CNS), and plays a crucial role in maintaining a stable circumstance of the CNS. Tight junctions among adjacent cells form the basic structure of BBB to limiting paracellular permeability. In the present review, the constituents of tight junction proteins are depicted in detail, together with the regulation of tight junction under stimulation and in pathological conditions. Tight junction modulators are also discussed.
Collapse
Affiliation(s)
- Wei-Ye Liu
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | | | | | | | | |
Collapse
|
44
|
Williams DW, Eugenin EA, Calderon TM, Berman JW. Monocyte maturation, HIV susceptibility, and transmigration across the blood brain barrier are critical in HIV neuropathogenesis. J Leukoc Biol 2012; 91:401-15. [PMID: 22227964 DOI: 10.1189/jlb.0811394] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
HIV continues to be a global health crisis with more than 34 million people infected worldwide (UNAIDS: Report on the Global AIDS Epidemic 2010, Geneva, World Health Organization). HIV enters the CNS within 2 weeks of infection and establishes a spectrum of HAND in a large percentage of infected individuals. These neurologic deficits greatly impact the quality of life of those infected with HIV. The establishment of HAND is largely attributed to monocyte transmigration, particularly that of a mature CD14(+)CD16(+) monocyte population, which is more susceptible to HIV infection, across the BBB into the CNS parenchyma in response to chemotactic signals. To enter the CNS, junctional proteins on the monocytes must participate in homo- and heterotypic interactions with those present on BMVECs of the BBB as they transmigrate across the barrier. This transmigration is responsible for bringing virus into the brain and establishing chronic neuroinflammation. While there is baseline trafficking of monocytes into the CNS, the increased chemotactic signals present during HIV infection of the brain promote exuberant monocyte transmigration into the CNS. This review will discuss the mechanisms of monocyte differentiation/maturation, HIV infectivity, and transmigration into the CNS parenchyma that contribute to the establishment of cognitive impairment in HIV-infected individuals. It will focus on markers of monocyte subpopulations, how differentiation/maturation alters HIV infectivity, and the mechanisms that promote their increased transmigration across the BBB into the CNS.
Collapse
Affiliation(s)
- Dionna W Williams
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY, 10461, USA
| | | | | | | |
Collapse
|
45
|
Xu H, Bae M, Tovar-y-Romo LB, Patel N, Bandaru VVR, Pomerantz D, Steiner JP, Haughey NJ. The human immunodeficiency virus coat protein gp120 promotes forward trafficking and surface clustering of NMDA receptors in membrane microdomains. J Neurosci 2011; 31:17074-90. [PMID: 22114277 PMCID: PMC3254245 DOI: 10.1523/jneurosci.4072-11.2011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 09/28/2011] [Accepted: 09/29/2011] [Indexed: 11/21/2022] Open
Abstract
Infection by the human immunodeficiency virus (HIV) can result in debilitating neurological syndromes collectively known as HIV-associated neurocognitive disorders. Although the HIV coat protein gp120 has been identified as a potent neurotoxin that enhances NMDA receptor function, the exact mechanisms for this effect are not known. Here we provide evidence that gp120 activates two separate signaling pathways that converge to enhance NMDA-evoked calcium flux by clustering NMDA receptors in modified membrane microdomains. gp120 enlarged and stabilized the structure of lipid microdomains on dendrites by mechanisms that involved a redox-regulated translocation of a sphingomyelin hydrolase (neutral sphingomyelinase-2) to the plasma membrane. A concurrent pathway was activated that accelerated the forward traffic of NMDA receptors by a PKA-dependent phosphorylation of the NR1 C-terminal serine 897 (masks an ER retention signal), followed by a PKC-dependent phosphorylation of serine 896 (important for surface expression). NMDA receptors were preferentially targeted to synapses and clustered in modified membrane microdomains. In these conditions, NMDA receptors were unable to laterally disperse and did not internalize, even in response to strong agonist induction. Focal NMDA-evoked calcium bursts were enhanced by threefold in these regions. Inhibiting membrane modification or NR1 phosphorylation prevented gp120 from accelerating the surface localization of NMDA receptors. Disrupting the structure of membrane microdomains after gp120 treatments restored the ability of NMDA receptors to disperse and internalize. These findings demonstrate that gp120 contributes to synaptic dysfunction in the setting of HIV infection by interfering with NMDA receptor trafficking.
Collapse
Affiliation(s)
- Hangxiu Xu
- Departments of Neurology, Richard T. Johnson Division of Neuroimmunology and Neurological Infections and
| | - Mihyun Bae
- Departments of Neurology, Richard T. Johnson Division of Neuroimmunology and Neurological Infections and
| | - Luis B. Tovar-y-Romo
- Departments of Neurology, Richard T. Johnson Division of Neuroimmunology and Neurological Infections and
| | - Neha Patel
- Departments of Neurology, Richard T. Johnson Division of Neuroimmunology and Neurological Infections and
| | | | - Daniel Pomerantz
- Departments of Neurology, Richard T. Johnson Division of Neuroimmunology and Neurological Infections and
| | - Joseph P. Steiner
- Departments of Neurology, Richard T. Johnson Division of Neuroimmunology and Neurological Infections and
| | - Norman J. Haughey
- Departments of Neurology, Richard T. Johnson Division of Neuroimmunology and Neurological Infections and
- Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| |
Collapse
|
46
|
Sáez CG, Olivares P, Pallavicini J, Panes O, Moreno N, Massardo T, Mezzano D, Pereira J. Increased number of circulating endothelial cells and plasma markers of endothelial damage in chronic cocaine users. Thromb Res 2011; 128:e18-23. [PMID: 21601240 DOI: 10.1016/j.thromres.2011.04.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 04/08/2011] [Accepted: 04/25/2011] [Indexed: 11/29/2022]
Abstract
BACKGROUND Cocaine use has been related with the development of accelerated atherosclerosis and with an increased risk of cardiac and cerebrovascular events, such as myocardial infarction, sudden cardiac death, and ischemic stroke. The underlying mechanisms leading to these complications are not fully understood, although thrombus formation and altered vascular function are prominent findings. OBJECTIVES Our aim was to evaluate markers of endothelial dysfunction in chronic cocaine consumers before and after drug withdrawal. PATIENTS/METHODS We determined circulating endothelial cells (CECs) and plasma levels of stromal cell-derived factor-1 (SDF-1), monocyte chemotactic protein-1(MCP-1), soluble intracellular adhesion molecule (sICAM), high-sensitivity C reactive protein (hsCRP) and endothelin-1(ET-1), in DSM-IV cocaine addicts at baseline and after one month of cocaine abstinence. RESULTS Cocaine users showed a strikingly higher numbers of CEC (62.35 ± 18.4 vs 8.25 ± 13.8 CEC/mL) and significantly elevated plasma levels for all the markers evaluated as compared to the control group. After cocaine withdrawal, patients improved SDF-1, ET-1, hsCRP and sICAM levels. However, CEC number and MCP-1 plasma levels remained significantly elevated. All the results were adjusted for blood levels of cholesterol and triglycerides and for smoking habit. CONCLUSIONS Our results demonstrated that chronic cocaine consumption alters several functions of the endothelium towards a pro-thrombotic condition and that some of those functions remain abnormal even after short-term drug withdrawal. These observations support the notion that endothelial dysfunction may play a key role in the pathogenesis of ischemic vascular disease observed in cocaine abusers.
Collapse
Affiliation(s)
- Claudia G Sáez
- Department of Hematology-Oncology, School of Medicine, Pontificia Universidad Católica de Chile
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Strazza M, Pirrone V, Wigdahl B, Nonnemacher MR. Breaking down the barrier: the effects of HIV-1 on the blood-brain barrier. Brain Res 2011; 1399:96-115. [PMID: 21641584 DOI: 10.1016/j.brainres.2011.05.015] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 05/06/2011] [Accepted: 05/07/2011] [Indexed: 01/13/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) primarily infects CD4(+) T cells and cells of the monocyte-macrophage lineage, resulting in immunodeficiency in an infected patient. Along with this immune deficiency, HIV-1 has been linked to a number of neurological symptoms in the absence of opportunistic infections or other co-morbidities, suggesting that HIV-1 is able to cross the blood-brain barrier (BBB), enter the central nervous system (CNS), and cause neurocognitive impairment. HIV-1-infected monocyte-macrophages traverse the BBB and enter the CNS throughout the course of HIV-1 disease. Once in the brain, both free virus and virus-infected cells are able to infect neighboring resident microglia and astrocytes and possibly other cell types. HIV-1-infected cells in both the periphery and the CNS give rise to elevated levels of viral proteins, including gp120, Tat, and Nef, and of host inflammatory mediators such as cytokines and chemokines. It has been shown that the viral proteins may act alone or in concert with host cytokines and chemokines, affecting the integrity of the BBB. The pathological end point of these interactions may facilitate a positive feedback loop resulting in increased penetration of HIV into the CNS. It is proposed in this review that the dysregulation of the BBB during and after neuroinvasion is a critical component of the neuropathogenic process and that dysregulation of this protective barrier is caused by a combination of viral and host factors including secreted viral proteins, components of the inflammatory process, the aging process, therapeutics, and drug or alcohol abuse.
Collapse
Affiliation(s)
- Marianne Strazza
- Department of Microbiology and Immunology, and Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | | | | | | |
Collapse
|
48
|
Abstract
Cocaine is one of the most abused street drugs; according to the National Survey on Drug Abuse, 15% of Americans have used cocaine at least once in their lifetime. Cocaine has been shown to alter behavior and mood, causing feelings of euphoria. One of the most damaging effects of cocaine abuse is that it compromises judgment capacity leading to risky sexual behavior, thereby increasing chances of contracting HIV infection.
Collapse
|
49
|
Trocello JM, Rostene W, Melik-Parsadaniantz S, Godefroy D, Roze E, Kitabgi P, Kuziel WA, Chalon S, Caboche J, Apartis E. Implication of CCR2 Chemokine Receptor in Cocaine-Induced Sensitization. J Mol Neurosci 2011; 44:147-51. [DOI: 10.1007/s12031-011-9508-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 03/01/2011] [Indexed: 01/14/2023]
|
50
|
Cocaine-mediated induction of platelet-derived growth factor: implication for increased vascular permeability. Blood 2010; 117:2538-47. [PMID: 21148086 DOI: 10.1182/blood-2010-10-313593] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Neuroinflammation associated with advanced HIV-1 infection is often exacerbated in cocaine-abusing, HIV-infected patients. The underlying mechanisms could, in part, be attributed to the increased impairment of blood brain barrier integrity in the presence of cocaine. Platelet-derived growth factor (PDGF) has been implicated in several pathologic conditions, specifically attributable to its potent mitogenic effects. Its modulation by drug abuse, however, has received very little attention. In the present study, we demonstrated cocaine-mediated induction of PDGF-BB in human brain microvascular endothelial cells through the binding to its cognate σ receptor. Furthermore, this effect was mediated, with subsequent activation of mitogen-activated protein kinases and Egr-1 pathways, culminating ultimately into increased expression of PDGF-BB. Cocaine exposure resulted in increased permeability of the endothelial barrier, and this effect was abrogated in mice exposed to PDGF-BB neutralizing antibody, thus underscoring its role as a vascular permeant. In vivo relevance of these findings was further corroborated in cocaine-treated mice that were administered neutralizing antibody specific for PDGF-BB as well as in Egr-1(-/-) mice. Understanding the regulation of PDGF-BB expression may provide insights into the development of potential therapeutic targets for neuroinflammation associated with HIV infection and drug abuse.
Collapse
|