1
|
Petro NM, Rempe MP, Schantell M, Ku V, Srinivas AN, O’Neill J, Kubat ME, Bares SH, May-Weeks PE, Wilson TW. Spontaneous cortical activity is altered in persons with HIV and related to domain-specific cognitive function. Brain Commun 2024; 6:fcae228. [PMID: 39035415 PMCID: PMC11258575 DOI: 10.1093/braincomms/fcae228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 05/23/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
Whilst the average lifespan of persons with HIV now approximates that of the general population, these individuals are at a much higher risk of developing cognitive impairment with ∼35-70% experiencing at least subtle cognitive deficits. Previous works suggest that HIV impacts both low-level primary sensory regions and higher-level association cortices. Notably, multiple neuroHIV studies have reported elevated levels of spontaneous cortical activity during the pre-stimulus baseline period of task-based experiments, but only a few have examined such activity during resting-state conditions. In the current study, we examined such spontaneous cortical activity using magnetoencephalography in 79 persons with HIV and 83 demographically matched seronegative controls and related this neural activity to performance on neuropsychological assessments of cognitive function. Consistent with previous works, persons with HIV exhibited stronger spontaneous gamma activity, particularly in inferior parietal, prefrontal and superior temporal cortices. In addition, serostatus moderated the relationship between spontaneous beta activity and attention, motor and processing speed scores, with controls but not persons with HIV showing stronger beta activity with better performance. The current results suggest that HIV predominantly impacts spontaneous activity in association cortices, consistent with alterations in higher-order brain function, and may be attributable to deficient GABAergic signalling, given its known role in the generation of gamma and beta oscillations. Overall, these effects align with previous studies showing aberrant spontaneous activity in persons with HIV and provide a critical new linkage to domain-specific cognitive dysfunction.
Collapse
Affiliation(s)
- Nathan M Petro
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE 68010, USA
| | - Maggie P Rempe
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE 68010, USA
- College of Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA
| | - Mikki Schantell
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE 68010, USA
- College of Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA
| | - Vivian Ku
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE 68010, USA
| | - Advika N Srinivas
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE 68010, USA
| | - Jennifer O’Neill
- Department of Internal Medicine, Division of Infectious Diseases, UNMC, Omaha, NE 68198, USA
| | - Maureen E Kubat
- Department of Internal Medicine, Division of Infectious Diseases, UNMC, Omaha, NE 68198, USA
| | - Sara H Bares
- Department of Internal Medicine, Division of Infectious Diseases, UNMC, Omaha, NE 68198, USA
| | | | - Tony W Wilson
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE 68010, USA
- Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE 68178, USA
| |
Collapse
|
2
|
Pla-Tenorio J, Roig AM, García-Cesaní PA, Santiago LA, Sepulveda-Orengo MT, Noel RJ. Astrocytes: Role in pathogenesis and effect of commonly misused drugs in the HIV infected brain. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 5:100108. [PMID: 38020814 PMCID: PMC10663134 DOI: 10.1016/j.crneur.2023.100108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 06/05/2023] [Accepted: 08/18/2023] [Indexed: 12/01/2023] Open
Abstract
The roles of astrocytes as reservoirs and producers of a subset of viral proteins in the HIV infected brain have been studied extensively as a key to understanding HIV-associated neurocognitive disorders (HAND). However, their comprehensive role in the context of intersecting substance use and neurocircuitry of the reward pathway and HAND has yet to be fully explained. Use of methamphetamines, cocaine, or opioids in the context of HIV infection have been shown to lead to a faster progression of HAND. Glutamatergic, dopaminergic, and GABAergic systems are implicated in the development of HAND-induced cognitive impairments. A thorough review of scientific literature exploring the variety of mechanisms in which these drugs exert their effects on the HIV brain and astrocytes has revealed marked areas of convergence in overexcitation leading to increased drug-seeking behavior, inflammation, apoptosis, and irreversible neurotoxicity. The present review investigates astrocytes, the neural pathways, and mechanisms of drug disruption that ultimately play a larger holistic role in terms of HIV progression and drug use. There are opportunities for future research, therapeutic intervention, and preventive strategies to diminish HAND in the subset population of patients with HIV and substance use disorder.
Collapse
Affiliation(s)
- Jessalyn Pla-Tenorio
- Ponce Health Sciences University, School of Medicine, Department of Basic Sciences, 395 Industrial Reparada, Zona 2, Ponce, PR, 00716, Puerto Rico
| | - Angela M. Roig
- Seattle Children's Hospital, MS OC.7.830, 4800 Sand Point Way NE, Seattle, WA, 98105-0371, United States
| | - Paulina A. García-Cesaní
- Bella Vista Hospital, Family Medicine Residency, Carr. 349 Km 2.7, Cerro Las Mesas, Mayaguez, PR, 00681, Puerto Rico
| | - Luis A. Santiago
- Ponce Health Sciences University, School of Medicine, Department of Basic Sciences, 395 Industrial Reparada, Zona 2, Ponce, PR, 00716, Puerto Rico
| | - Marian T. Sepulveda-Orengo
- Ponce Health Sciences University, School of Medicine, Department of Basic Sciences, 395 Industrial Reparada, Zona 2, Ponce, PR, 00716, Puerto Rico
| | - Richard J. Noel
- Ponce Health Sciences University, School of Medicine, Department of Basic Sciences, 395 Industrial Reparada, Zona 2, Ponce, PR, 00716, Puerto Rico
| |
Collapse
|
3
|
Rodriguez MT, McLaurin KA, Shtutman M, Kubinak JL, Mactutus CF, Booze RM. Therapeutically targeting the consequences of HIV-1-associated gastrointestinal dysbiosis: Implications for neurocognitive and affective alterations. Pharmacol Biochem Behav 2023; 229:173592. [PMID: 37390973 PMCID: PMC10494709 DOI: 10.1016/j.pbb.2023.173592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Approximately 50 % of the individuals living with human immunodeficiency virus type 1 (HIV-1) are plagued by debilitating neurocognitive impairments (NCI) and/or affective alterations. Sizeable alterations in the composition of the gut microbiome, or gastrointestinal dysbiosis, may underlie, at least in part, the NCI, apathy, and/or depression observed in this population. Herein, two interrelated aims will be critically addressed, including: 1) the evidence for, and functional implications of, gastrointestinal microbiome dysbiosis in HIV-1 seropositive individuals; and 2) the potential for therapeutically targeting the consequences of this dysbiosis for the treatment of HIV-1-associated NCI and affective alterations. First, gastrointestinal microbiome dysbiosis in HIV-1 seropositive individuals is characterized by decreased alpha (α) diversity, a decreased relative abundance of bacterial species belonging to the Bacteroidetes phylum, and geographic-specific alterations in Bacillota (formerly Firmicutes) spp. Fundamentally, changes in the relative abundance of Bacteroidetes and Bacillota spp. may underlie, at least in part, the deficits in γ-aminobutyric acid and serotonin neurotransmission, as well as prominent synaptodendritic dysfunction, observed in this population. Second, there is compelling evidence for the therapeutic utility of targeting synaptodendritic dysfunction as a method to enhance neurocognitive function and improve motivational dysregulation in HIV-1. Further research is needed to determine whether the therapeutics enhancing synaptic efficacy exert their effects by altering the gut microbiome. Taken together, understanding gastrointestinal microbiome dysbiosis resulting from chronic HIV-1 viral protein exposure may afford insight into the mechanisms underlying HIV-1-associated neurocognitive and/or affective alterations; mechanisms which can be subsequently targeted via novel therapeutics.
Collapse
Affiliation(s)
- Mason T Rodriguez
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, 1512 Pendleton Street, University of South Carolina, Columbia, SC 29208, United States of America
| | - Kristen A McLaurin
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, 1512 Pendleton Street, University of South Carolina, Columbia, SC 29208, United States of America
| | - Michael Shtutman
- Drug Discovery and Biomedical Sciences, College of Pharmacy, 715 Sumter Street, University of South Carolina, Columbia, SC 29208, United States of America
| | - Jason L Kubinak
- Pathology, Microbiology & Immunology, School of Medicine Columbia, 6311 Garners Ferry Road, Building 2, Columbia, SC 29209, United States of America
| | - Charles F Mactutus
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, 1512 Pendleton Street, University of South Carolina, Columbia, SC 29208, United States of America
| | - Rosemarie M Booze
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, 1512 Pendleton Street, University of South Carolina, Columbia, SC 29208, United States of America.
| |
Collapse
|
4
|
Lark ARS, Silva LK, Nass SR, Marone MG, Ohene-Nyako M, Ihrig TM, Marks WD, Yarotskyy V, Rory McQuiston A, Knapp PE, Hauser KF. Progressive Degeneration and Adaptive Excitability in Dopamine D1 and D2 Receptor-Expressing Striatal Neurons Exposed to HIV-1 Tat and Morphine. Cell Mol Neurobiol 2023; 43:1105-1127. [PMID: 35695980 PMCID: PMC9976699 DOI: 10.1007/s10571-022-01232-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/10/2022] [Indexed: 11/03/2022]
Abstract
The striatum is especially vulnerable to HIV-1 infection, with medium spiny neurons (MSNs) exhibiting marked synaptodendritic damage that can be exacerbated by opioid use disorder. Despite known structural defects in MSNs co-exposed to HIV-1 Tat and opioids, the pathophysiological sequelae of sustained HIV-1 exposure and acute comorbid effects of opioids on dopamine D1 and D2 receptor-expressing (D1 and D2) MSNs are unknown. To address this question, Drd1-tdTomato- or Drd2-eGFP-expressing reporter and conditional HIV-1 Tat transgenic mice were interbred. MSNs in ex vivo slices from male mice were assessed by whole-cell patch-clamp electrophysiology and filled with biocytin to explore the functional and structural effects of progressive Tat and acute morphine exposure. Although the excitability of both D1 and D2 MSNs increased following 48 h of Tat exposure, D1 MSN firing rates decreased below control (Tat-) levels following 2 weeks and 1 month of Tat exposure but returned to control levels after 2 months. D2 neurons continued to display Tat-dependent increases in excitability at 2 weeks, but also returned to control levels following 1 and 2 months of Tat induction. Acute morphine exposure increased D1 MSN excitability irrespective of the duration of Tat exposure, while D2 MSNs were variably affected. That D1 and D2 MSN excitability would return to control levels was unexpected since both subpopulations displayed significant synaptodendritic degeneration and pathologic phospho-tau-Thr205 accumulation following 2 months of Tat induction. Thus, despite frank morphologic damage, D1 and D2 MSNs uniquely adapt to sustained Tat and acute morphine insults.
Collapse
Affiliation(s)
- Arianna R S Lark
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
| | - Lindsay K Silva
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
- PPD®, Part of Thermo Fisher Scientific, Richmond, VA, 23230-3323, USA
| | - Sara R Nass
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
| | - Michael G Marone
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
| | - Michael Ohene-Nyako
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
| | - Therese M Ihrig
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
| | - William D Marks
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
- Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas, TX, 75235, USA
| | - Viktor Yarotskyy
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
| | - A Rory McQuiston
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, PO Box 980709, Richmond, VA, 23298-0709, USA
| | - Pamela E Knapp
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, PO Box 980709, Richmond, VA, 23298-0709, USA
- Institute for Drug and Alcohol Studies, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Molecular Medicine Research Building, Room 4040, 1220 East Broad Street, PO Box 980613, Richmond, VA, 23298-0613, USA.
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, PO Box 980709, Richmond, VA, 23298-0709, USA.
- Institute for Drug and Alcohol Studies, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| |
Collapse
|
5
|
Khodr CE, Chen L, Al-Harthi L, Hu XT. HIV-Induced Hyperactivity of Striatal Neurons Is Associated with Dysfunction of Voltage-Gated Calcium and Potassium Channels at Middle Age. MEMBRANES 2022; 12:737. [PMID: 36005652 PMCID: PMC9415409 DOI: 10.3390/membranes12080737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022]
Abstract
Despite combination antiretroviral therapy, HIV-associated neurocognitive disorders (HAND) occur in ~50% of people living with HIV (PLWH), which are associated with dysfunction of the corticostriatal pathway. The mechanism by which HIV alters the neuronal activity in the striatum is unknown. The goal of this study is to reveal the dysfunction of striatal neurons in the context of neuroHIV during aging. Using patch-clamping electrophysiology, we evaluated the functional activity of medium spiny neurons (MSNs), including firing, Ca2+ spikes mediated by voltage-gated Ca2+ channels (VGCCs), and K+ channel-mediated membrane excitability, in brain slices containing the dorsal striatum (a.k.a. the caudate-putamen) from 12-month-old (12mo) HIV-1 transgenic (HIV-1 Tg) rats. We also assessed the protein expression of voltage-gated Cav1.2/Cav1.3 L-type Ca2+ channels (L-channels), NMDA receptors (NMDAR, NR2B subunit), and GABAA receptors (GABAARs, β2,3 subunit) in the striatum. We found that MSNs had significantly increased firing in 12mo HIV-1 Tg rats compared to age-matched non-Tg control rats. Unexpectedly, Ca2+ spikes were significantly reduced, while Kv channel activity was increased, in MSNs of HIV-1 Tg rats compared to non-Tg ones. The reduced Ca2+ spikes were associated with an abnormally increased expression of a shorter, less functional Cav1.2 L-channel form, while there was no significant change in the expression of NR2Bs or GABAARs. Collectively, the present study initially reveals neuroHIV-induced dysfunction of striatal MSNs in 12mo-old (middle) rats, which is uncoupled from VGCC upregulation and reduced Kv activity (that we previously identified in younger HIV-1 Tg rats). Notably, such striatal dysfunction is also associated with HIV-induced hyperactivity/neurotoxicity of glutamatergic pyramidal neurons in the medial prefrontal cortex (mPFC) that send excitatory input to the striatum (demonstrated in our previous studies). Whether such MSN dysfunction is mediated by alterations in the functional activity instead of the expression of NR2b/GABAAR (or other subtypes) requires further investigation.
Collapse
Affiliation(s)
| | | | | | - Xiu-Ti Hu
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Cohn Research Building, Rm.610, 1735 W. Harrison Street, Chicago, IL 60612, USA; (C.E.K.); (L.C.); (L.A.-H.)
| |
Collapse
|
6
|
Aberrant Synaptic Pruning in CNS Diseases: A Critical Player in HIV-Associated Neurological Dysfunction? Cells 2022; 11:cells11121943. [PMID: 35741071 PMCID: PMC9222069 DOI: 10.3390/cells11121943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/28/2022] [Accepted: 06/14/2022] [Indexed: 02/04/2023] Open
Abstract
Even in the era of effective antiretroviral therapies, people living with Human Immunodeficiency Virus (HIV) are burdened with debilitating neurological dysfunction, such as HIV-associated neurocognitive disorders (HAND) and HIV-associated pain, for which there are no FDA approved treatments. Disruption to the neural circuits of cognition and pain in the form of synaptic degeneration is implicated in developing these dysfunctions. Glia-mediated synaptic pruning is a mechanism of structural plasticity in the healthy central nervous system (CNS), but recently, it has been discovered that dysregulated glia-mediated synaptic pruning is the cause of synaptic degeneration, leading to maladaptive plasticity and cognitive deficits in multiple diseases of the CNS. Considering the essential contribution of activated glial cells during the development of HAND and HIV-associated pain, it is possible that glia-mediated synaptic pruning is the causative mechanism of synaptic degeneration induced by HIV. This review will analyze the known examples of synaptic pruning during disease in order to better understand how this mechanism could contribute to the progression of HAND and HIV-associated pain.
Collapse
|
7
|
Abstract
Infection with SARS-CoV-2, the causative agent of the COVID-19 pandemic, originated in China and quickly spread across the globe. Despite tremendous economic and healthcare devastation, research on this virus has contributed to a better understanding of numerous molecular pathways, including those involving γ-aminobutyric acid (GABA), that will positively impact medical science, including neuropsychiatry, in the post-pandemic era. SARS-CoV-2 primarily enters the host cells through the renin–angiotensin system’s component named angiotensin-converting enzyme-2 (ACE-2). Among its many functions, this protein upregulates GABA, protecting not only the central nervous system but also the endothelia, the pancreas, and the gut microbiota. SARS-CoV-2 binding to ACE-2 usurps the neuronal and non-neuronal GABAergic systems, contributing to the high comorbidity of neuropsychiatric illness with gut dysbiosis and endothelial and metabolic dysfunctions. In this perspective article, we take a closer look at the pathology emerging from the viral hijacking of non-neuronal GABA and summarize potential interventions for restoring these systems.
Collapse
|
8
|
Xu C, Yadav-Samudrala BJ, Xu C, Nath B, Mistry T, Jiang W, Niphakis MJ, Cravatt BF, Mukhopadhyay S, Lichtman AH, Ignatowska-Jankowska BM, Fitting S. Inhibitory Neurotransmission Is Sex-Dependently Affected by Tat Expression in Transgenic Mice and Suppressed by the Fatty Acid Amide Hydrolase Enzyme Inhibitor PF3845 via Cannabinoid Type-1 Receptor Mechanisms. Cells 2022; 11:857. [PMID: 35269478 PMCID: PMC8909692 DOI: 10.3390/cells11050857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/14/2022] [Accepted: 02/23/2022] [Indexed: 11/21/2022] Open
Abstract
(1) Background. The endocannabinoid (eCB) system, which regulates physiological and cognitive processes, presents a promising therapeutic target for treating HIV-associated neurocognitive disorders (HAND). Here we examine whether upregulating eCB tone has potential protective effects against HIV-1 Tat (a key HIV transactivator of transcription) protein-induced alterations in synaptic activity. (2) Methods. Whole-cell patch-clamp recordings were performed to assess inhibitory GABAergic neurotransmission in prefrontal cortex slices of Tat transgenic male and female mice, in the presence and absence of the fatty acid amide hydrolase (FAAH) enzyme inhibitor PF3845. Western blot and mass spectrometry analyses assessed alterations of cannabinoid receptor and enzyme protein expression as well as endogenous ligands, respectively, to determine the impact of Tat exposure on the eCB system. (3) Results. GABAergic activity was significantly altered upon Tat exposure based on sex, whereas the effectiveness of PF3845 to suppress GABAergic activity in Tat transgenic mice was not altered by Tat or sex and involved CB1R-related mechanisms that depended on calcium signaling. Additionally, our data indicated sex-dependent changes for AEA and related non-eCB lipids based on Tat induction. (4) Conclusion. Results highlight sex- and/or Tat-dependent alterations of GABAergic activity and eCB signaling in the prefrontal cortex of Tat transgenic mice and further increase our understanding about the role of FAAH inhibition in neuroHIV.
Collapse
Affiliation(s)
- Changqing Xu
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (C.X.); (B.J.Y.-S.); (C.X.)
| | - Barkha J. Yadav-Samudrala
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (C.X.); (B.J.Y.-S.); (C.X.)
| | - Callie Xu
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (C.X.); (B.J.Y.-S.); (C.X.)
| | - Bhupendra Nath
- Department of Chemistry & Biochemistry, North Carolina Central University, Durham, NC 27707, USA; (B.N.); (T.M.); (S.M.)
| | - Twisha Mistry
- Department of Chemistry & Biochemistry, North Carolina Central University, Durham, NC 27707, USA; (B.N.); (T.M.); (S.M.)
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA;
- Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Micah J. Niphakis
- Department of Chemical Physiology, Scripps Research Institute, La Jolla, CA 92037, USA; (M.J.N.); (B.F.C.)
| | - Benjamin F. Cravatt
- Department of Chemical Physiology, Scripps Research Institute, La Jolla, CA 92037, USA; (M.J.N.); (B.F.C.)
| | - Somnath Mukhopadhyay
- Department of Chemistry & Biochemistry, North Carolina Central University, Durham, NC 27707, USA; (B.N.); (T.M.); (S.M.)
| | - Aron H. Lichtman
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | | | - Sylvia Fitting
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (C.X.); (B.J.Y.-S.); (C.X.)
| |
Collapse
|
9
|
Sil S, Periyasamy P, Thangaraj A, Niu F, Chemparathy DT, Buch S. Advances in the Experimental Models of HIV-Associated Neurological Disorders. Curr HIV/AIDS Rep 2021; 18:459-474. [PMID: 34427869 DOI: 10.1007/s11904-021-00570-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Involvement of the central nervous system (CNS) in HIV-1 infection is commonly associated with neurological disorders and cognitive impairment, commonly referred to as HIV-associated neurocognitive disorders (HAND). Severe and progressive neurocognitive impairment is rarely observed in the post-cART era; however, asymptomatic and mild neurocognitive disorders still exist, despite viral suppression. Additionally, comorbid conditions can also contribute to the pathogenesis of HAND. RECENT FINDINGS In this review, we summarize the characterization of HAND, factors contributing, and the functional impairments in both preclinical and clinical models. Specifically, we also discuss recent advances in the animal models of HAND and in in vitro cultures and the potential role of drugs of abuse in this model system of HAND. Potential peripheral biomarkers associated with HAND are also discussed. Overall, this review identifies some of the recent advances in the field of HAND in cell culture studies, animal models, clinical findings, and the limitations of each model system, which can play a key role in developing novel therapeutics in the field.
Collapse
Affiliation(s)
- Susmita Sil
- 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.
| | - Annadurai Thangaraj
- 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
| | - Divya T Chemparathy
- 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
|
10
|
Huang MX, Huang CW, Harrington DL, Nichols S, Robb-Swan A, Angeles-Quinto A, Le L, Rimmele C, Drake A, Song T, Huang JW, Clifford R, Ji Z, Cheng CK, Lerman I, Yurgil KA, Lee RR, Baker DG. Marked Increases in Resting-State MEG Gamma-Band Activity in Combat-Related Mild Traumatic Brain Injury. Cereb Cortex 2021; 30:283-295. [PMID: 31041986 DOI: 10.1093/cercor/bhz087] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 01/08/2023] Open
Abstract
Combat-related mild traumatic brain injury (mTBI) is a leading cause of sustained impairments in military service members and veterans. Recent animal studies show that GABA-ergic parvalbumin-positive interneurons are susceptible to brain injury, with damage causing abnormal increases in spontaneous gamma-band (30-80 Hz) activity. We investigated spontaneous gamma activity in individuals with mTBI using high-resolution resting-state magnetoencephalography source imaging. Participants included 25 symptomatic individuals with chronic combat-related blast mTBI and 35 healthy controls with similar combat experiences. Compared with controls, gamma activity was markedly elevated in mTBI participants throughout frontal, parietal, temporal, and occipital cortices, whereas gamma activity was reduced in ventromedial prefrontal cortex. Across groups, greater gamma activity correlated with poorer performances on tests of executive functioning and visuospatial processing. Many neurocognitive associations, however, were partly driven by the higher incidence of mTBI participants with both higher gamma activity and poorer cognition, suggesting that expansive upregulation of gamma has negative repercussions for cognition particularly in mTBI. This is the first human study to demonstrate abnormal resting-state gamma activity in mTBI. These novel findings suggest the possibility that abnormal gamma activities may be a proxy for GABA-ergic interneuron dysfunction and a promising neuroimaging marker of insidious mild head injuries.
Collapse
Affiliation(s)
- Ming-Xiong Huang
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,Department of Radiology, University of California, San Diego, CA, USA
| | - Charles W Huang
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Deborah L Harrington
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,Department of Radiology, University of California, San Diego, CA, USA
| | - Sharon Nichols
- Department of Neuroscience, University of California, San Diego, CA, USA
| | - Ashley Robb-Swan
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,Department of Radiology, University of California, San Diego, CA, USA
| | - Annemarie Angeles-Quinto
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,Department of Radiology, University of California, San Diego, CA, USA
| | - Lu Le
- ASPIRE Center, VASDHS Residential Rehabilitation Treatment Program, San Diego, CA, USA
| | - Carl Rimmele
- ASPIRE Center, VASDHS Residential Rehabilitation Treatment Program, San Diego, CA, USA
| | - Angela Drake
- Cedar Sinai Medical Group Chronic Pain Program, Beverly Hills, CA, USA
| | - Tao Song
- Department of Radiology, University of California, San Diego, CA, USA
| | - Jeffrey W Huang
- Department of Computer Science, Columbia University, New York, NY, USA
| | - Royce Clifford
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego, CA, USA.,VA Center of Excellence for Stress and Mental Health, San Diego, CA, USA
| | - Zhengwei Ji
- Department of Radiology, University of California, San Diego, CA, USA
| | - Chung-Kuan Cheng
- Department of Computer Science and Engineering, University of California, San Diego, CA, USA
| | - Imanuel Lerman
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA
| | - Kate A Yurgil
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,VA Center of Excellence for Stress and Mental Health, San Diego, CA, USA.,Department of Psychological Sciences, Loyola University, New Orleans, LA, USA
| | - Roland R Lee
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,Department of Radiology, University of California, San Diego, CA, USA
| | - Dewleen G Baker
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego, CA, USA.,VA Center of Excellence for Stress and Mental Health, San Diego, CA, USA
| |
Collapse
|
11
|
Irollo E, Luchetta J, Ho C, Nash B, Meucci O. Mechanisms of neuronal dysfunction in HIV-associated neurocognitive disorders. Cell Mol Life Sci 2021; 78:4283-4303. [PMID: 33585975 PMCID: PMC8164580 DOI: 10.1007/s00018-021-03785-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/14/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022]
Abstract
HIV-associated neurocognitive disorder (HAND) is characterized by cognitive and behavioral deficits in people living with HIV. HAND is still common in patients that take antiretroviral therapies, although they tend to present with less severe symptoms. The continued prevalence of HAND in treated patients is a major therapeutic challenge, as even minor cognitive impairment decreases patient’s quality of life. Therefore, modern HAND research aims to broaden our understanding of the mechanisms that drive cognitive impairment in people with HIV and identify promising molecular pathways and targets that could be exploited therapeutically. Recent studies suggest that HAND in treated patients is at least partially induced by subtle synaptodendritic damage and disruption of neuronal networks in brain areas that mediate learning, memory, and executive functions. Although the causes of subtle neuronal dysfunction are varied, reversing synaptodendritic damage in animal models restores cognitive function and thus highlights a promising therapeutic approach. In this review, we examine evidence of synaptodendritic damage and disrupted neuronal connectivity in HAND from clinical neuroimaging and neuropathology studies and discuss studies in HAND models that define structural and functional impairment of neurotransmission. Then, we report molecular pathways, mechanisms, and comorbidities involved in this neuronal dysfunction, discuss new approaches to reverse neuronal damage, and highlight current gaps in knowledge. Continued research on the manifestation and mechanisms of synaptic injury and network dysfunction in HAND patients and experimental models will be critical if we are to develop safe and effective therapies that reverse subtle neuropathology and cognitive impairment.
Collapse
Affiliation(s)
- Elena Irollo
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Jared Luchetta
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Chunta Ho
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Bradley Nash
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA. .,Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA. .,Center for Neuroimmunology and CNS Therapeutics, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
12
|
Barbour AJ, Nass SR, Hahn YK, Hauser KF, Knapp PE. Restoration of KCC2 Membrane Localization in Striatal Dopamine D2 Receptor-Expressing Medium Spiny Neurons Rescues Locomotor Deficits in HIV Tat-Transgenic Mice. ASN Neuro 2021; 13:17590914211022089. [PMID: 34445881 PMCID: PMC8404672 DOI: 10.1177/17590914211022089] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 04/23/2021] [Accepted: 05/06/2021] [Indexed: 11/17/2022] Open
Abstract
People infected with HIV (PWH) are highly susceptible to striatal and hippocampal damage. Motor and memory impairments are common among these patients, likely as behavioral manifestations of damage to these brain regions. GABAergic dysfunction from HIV infection and viral proteins such as transactivator of transcription (Tat) have been well documented. We recently demonstrated that the neuron specific Cl- extruder, K+ Cl- cotransporter 2 (KCC2), is diminished after exposure to HIV proteins, including Tat, resulting in disrupted GABAAR-mediated hyperpolarization and inhibition. Here, we utilized doxycycline (DOX)-inducible, GFAP-driven HIV-1 Tat transgenic mice to further explore this phenomenon. After two weeks of Tat expression, we found no changes in hippocampal KCC2 levels, but a significant decrease in the striatum that was associated with hyperlocomotion in the open field assay. We were able to restore KCC2 activity and baseline locomotion with the KCC2 enhancer, CLP290. Additionally, we found that CLP290, whose mechanism of action has yet to be described, acts to restore phosphorylation of serine 940 resulting in increased KCC2 membrane localization. We also examined neuronal subpopulation contributions to the noted effects and found significant differences. Dopamine D2 receptor-expressing medium spiny neurons (MSNs) were selectively vulnerable to Tat-induced KCC2 loss, with no changes observed in dopamine D1 receptor-expressing MSNs. These results suggest that disinhibition/diminished hyperpolarization of dopamine D2 receptor-expressing MSNs can manifest as increased locomotion in this context. They further suggest that KCC2 activity might be a therapeutic target to alleviate motor disturbances related to HIV.
Collapse
Affiliation(s)
- Aaron J. Barbour
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
| | - Sara R. Nass
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
| | - Yun K. Hahn
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
| | - Kurt F. Hauser
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
| | - Pamela E. Knapp
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
| |
Collapse
|
13
|
Benzodiazepine Use and HIV-Associated Neurocognitive Impairment: Which Comes First? J Acquir Immune Defic Syndr 2020; 83:e26-e27. [PMID: 31914007 DOI: 10.1097/qai.0000000000002278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14
|
Barbour AJ, Hauser KF, McQuiston AR, Knapp PE. HIV and opiates dysregulate K +- Cl - cotransporter 2 (KCC2) to cause GABAergic dysfunction in primary human neurons and Tat-transgenic mice. Neurobiol Dis 2020; 141:104878. [PMID: 32344154 PMCID: PMC7685173 DOI: 10.1016/j.nbd.2020.104878] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/19/2020] [Accepted: 04/22/2020] [Indexed: 12/31/2022] Open
Abstract
Approximately half of people infected with HIV (PWH) exhibit HIV-associated neuropathology (neuroHIV), even when receiving combined antiretroviral therapy. Opiate use is widespread in PWH and exacerbates neuroHIV. While neurons themselves are not infected, they incur sublethal damage and GABAergic disruption is selectively vulnerable to viral and inflammatory factors released by infected/affected glia. Here, we demonstrate diminished K+-Cl- cotransporter 2 (KCC2) levels in primary human neurons after exposure to HIV-1 or HIV-1 proteins ± morphine. Resulting disruption of GABAAR-mediated hyperpolarization/inhibition was shown using genetically-encoded voltage (Archon1) and calcium (GCaMP6f) indicators. The HIV proteins Tat (acting through NMDA receptors) and R5-gp120 (acting via CCR5) but not X4-tropic gp120 (acting via CXCR4), and morphine (acting through μ-opioid receptors) all induced KCC2 loss. We demonstrate that modifying KCC2 levels or function, or antagonizing NMDAR, CCR5 or MOR rescues KCC2 and GABAAR-mediated hyperpolarization/inhibition in HIV, Tat, or gp120 ± morphine-exposed neurons. Using an inducible, Tat-transgenic mouse neuroHIV model, we found that chronic exposure to Tat also reduces KCC2. Our results identify KCC2 as a novel therapeutic target for ameliorating the pathobiology of neuroHIV, including PWH exposed to opiates.
Collapse
Affiliation(s)
- Aaron J Barbour
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Kurt F Hauser
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - A Rory McQuiston
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Pamela E Knapp
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA.
| |
Collapse
|
15
|
Benzodiazepine Use Is Associated With an Increased Risk of Neurocognitive Impairment in People Living With HIV. J Acquir Immune Defic Syndr 2020; 82:475-482. [PMID: 31714426 DOI: 10.1097/qai.0000000000002183] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Despite potential for dependence and adverse neurological effects, long-term benzodiazepine (BZD) use is common among people living with HIV (PLWH). As PLWH are at risk for central nervous system dysfunction, we retrospectively examined the association between BZD use and HIV-associated neurocognitive impairment (NCI). METHODS Three hundred six PLWH underwent comprehensive neurobehavioral evaluations. Current BZD use (BZD+) was determined through self-report. Using propensity scores, 153 BZD- individuals were matched to 153 BZD+ participants on demographics and medical comorbidities. Multiple regression models examined NCI and demographically adjusted neurocognitive T-scores as a function of BZD status, adjusting for estimated premorbid ability, current affective symptoms, and nadir CD4 count. Secondary analyses explored neurocognitive correlates of positive BZD urine toxicology screens (TOX+) and specific BZD agents. RESULTS Median duration of BZD use was 24 months. Current BZD use related to higher likelihood of NCI (odds ratio = 2.13, P = 0.003) and poorer global (d = -0.28, P = 0.020), processing speed (d = -0.23, P = 0.047), and motor T-scores (d = -0.32, P = 0.008). Compared with BZD-/TOX-, BZD+/TOX+ exhibited additional decrements in executive function (d = -0.48, P = 0.013), working memory (d = -0.49, P = 0.011), and delayed recall (d = -0.41, P = 0.032). For individual agents, diazepam, lorazepam, and alprazolam were most strongly associated with NCI (odds ratios >2.31). DISCUSSION BZD use may elevate risk for NCI in PLWH, potentially through diffuse neurocognitive slowing and acute compromise of recall and higher-order capacities. These effects are robust to psychosocial and HIV-specific factors and occur in comparison with a tightly matched BZD- group. Prospective and interventional studies should evaluate causal associations between NCI and BZD use and explore treatment alternatives to BZDs in PLWH.
Collapse
|
16
|
Huang M, Lewine JD, Lee RR. Magnetoencephalography for Mild Traumatic Brain Injury and Posttraumatic Stress Disorder. Neuroimaging Clin N Am 2020; 30:175-192. [DOI: 10.1016/j.nic.2020.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
17
|
Nass SR, Hahn YK, McLane VD, Varshneya NB, Damaj MI, Knapp PE, Hauser KF. Chronic HIV-1 Tat exposure alters anterior cingulate cortico-basal ganglia-thalamocortical synaptic circuitry, associated behavioral control, and immune regulation in male mice. Brain Behav Immun Health 2020; 5:100077. [PMID: 33083793 PMCID: PMC7571616 DOI: 10.1016/j.bbih.2020.100077] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 04/25/2020] [Indexed: 12/28/2022] Open
Abstract
HIV-1 selectively disrupts neuronal integrity within specific brain regions, reflecting differences in viral tropism and/or the regional differences in the vulnerability of distinct neuronal subpopulations within the CNS. Deficits in prefrontal cortex (PFC)-mediated executive function and the resultant loss of behavioral control are a particularly debilitating consequence of neuroHIV. To explore how HIV-1 disrupts executive function, we investigated the effects of 48 h, 2 and/or 8 weeks of HIV-1 Tat exposure on behavioral control, synaptic connectivity, and neuroimmune function in the anterior cingulate cortex (ACC) and associated cortico-basal ganglia (BG)-thalamocortical circuitry in adult, Tat transgenic male mice. HIV-1 Tat exposure increased novelty-exploration in response to novel food, flavor, and environmental stimuli, suggesting that Tat triggers increased novelty-exploration in situations of competing motivation (e.g., drive to feed or explore vs. fear of novel, brightly lit open areas). Furthermore, Tat induced adaptability in response to an environmental stressor and pre-attentive filtering deficits. The behavioral insufficiencies coincided with decreases in the inhibitory pre- and post-synaptic proteins, synaptotagmin 2 and gephyrin, respectively, in the ACC, and alterations in specific pro- and anti-inflammatory cytokines out of 23 assayed. The interaction of Tat exposure and the resultant time-dependent, selective alterations in CCL4, CXCL1, IL-12p40, and IL-17A levels in the PFC predicted significant decreases in adaptability. Tat decreased dendritic spine density and cortical VGLUT1 inputs, while increasing IL-1β, IL-6, CCL5, and CCL11 in the striatum. Alternatively, IL-1α, CCL5, and IL-13 were decreased in the mediodorsal thalamus despite the absence of synaptic changes. Thus, HIV-1 Tat appears to uniquely and systematically disrupt immune regulation and the inhibitory and excitatory synaptic balance throughout the ACC-BG-thalamocortical circuitry resulting in a loss of behavioral control.
Collapse
Affiliation(s)
- Sara R. Nass
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
| | - Yun K. Hahn
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980709, Richmond, VA, 23298-0709, USA
| | - Virginia D. McLane
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
| | - Neil B. Varshneya
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
| | - M. Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
| | - Pamela E. Knapp
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980709, Richmond, VA, 23298-0709, USA
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980059, Richmond, VA, 23298-0059, USA
| | - Kurt F. Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980709, Richmond, VA, 23298-0709, USA
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980059, Richmond, VA, 23298-0059, USA
| |
Collapse
|
18
|
Çomakli S, Özdemir S, Değirmençay Ş. Canine distemper virus induces downregulation of GABA A,GABA B, and GAT1 expression in brain tissue of dogs. Arch Virol 2020; 165:1321-1331. [PMID: 32253618 DOI: 10.1007/s00705-020-04617-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/11/2020] [Indexed: 02/08/2023]
Abstract
The aim of the study was to determine the expression profiles of GABAA, GABAB, and GAT1 using RT-PCR and the immunoreactivity of GAT1 via immunohistochemical and immunofluorescence assays in CDV-infected brain tissue of dogs. For this purpose, dogs with CDV and dogs without CDV were selected. The mRNA transcript levels of GABAA, GABAB, and GAT1 were significantly downregulated in brain tissue in the CDV-infected group as compared with that in non-CDV-infected brain tissue in the control group (p < 0.01, p < 0.001). In addition, the immunoreactivity of GAT1 in CDV-infected brain tissue was significantly lower than in the uninfected group (p < 0.05). We conclude that one of the main causes of myoclonus in CDV infections may be the blockage of postsynaptic inhibition in neurons or a lack of metabolism of GABA. In addition, a GABA neurotransmission imbalance could play a role in demyelination in CDV infections.
Collapse
Affiliation(s)
- Selim Çomakli
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - Selçuk Özdemir
- Department of Genetic, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Şükrü Değirmençay
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| |
Collapse
|
19
|
Nash B, Festa L, Lin C, Meucci O. Opioid and chemokine regulation of cortical synaptodendritic damage in HIV-associated neurocognitive disorders. Brain Res 2019; 1723:146409. [PMID: 31465771 PMCID: PMC6766413 DOI: 10.1016/j.brainres.2019.146409] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/20/2019] [Accepted: 08/25/2019] [Indexed: 01/17/2023]
Abstract
Human immunodeficiency virus (HIV)-associated neurocognitive disorders (HAND) persist despite effective antiretroviral therapies (ART). Evidence suggests that modern HAND is driven by subtle synaptodendritic damage in select brain regions, as ART-treated patients do not display overt neuronal death in postmortem brain studies. HAND symptoms are also aggravated by drug abuse, particularly with injection opioids. Opioid use produces region-specific synaptodendritic damage in similar brain regions, suggesting a convergent mechanism that may enhance HAND progression in opioid-using patients. Importantly, studies indicate that synaptodendritic damage and cognitive impairment in HAND may be reversible. Activation of the homeostatic chemokine receptor CXCR4 by its natural ligand CXCL12 positively regulates neuronal survival and dendritic spine density in cortical neurons, reducing functional deficits. However, the molecular mechanisms that underlie CXCR4, as well as opioid-mediated regulation of dendritic spines are not completely defined. Here, we will consolidate studies that describe the region-specific synaptodendritic damage in the cerebral cortex of patients and animal models of HAND, describe the pathways by which opioids may contribute to cortical synaptodendritic damage, and discuss the prospects of using the CXCR4 signaling pathway to identify new approaches to reverse dendritic spine deficits. Additionally, we will discuss novel research questions that have emerged from recent studies of CXCR4 and µ-opioid actions in the cortex. Understanding the pathways that underlie synaptodendritic damage and rescue are necessary for developing novel, effective therapeutics for this growing patient population.
Collapse
Affiliation(s)
- Bradley Nash
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA.
| | - Lindsay Festa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA.
| | - Chihyang Lin
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA.
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA; Department of Microbiology and Immunology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA.
| |
Collapse
|
20
|
Dopaminergic impact of cART and anti-depressants on HIV neuropathogenesis in older adults. Brain Res 2019; 1723:146398. [PMID: 31442412 DOI: 10.1016/j.brainres.2019.146398] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 08/09/2019] [Accepted: 08/19/2019] [Indexed: 01/21/2023]
Abstract
The success of combination antiretroviral therapy (cART) has transformed HIV infection into a chronic condition, resulting in an increase in the number of older, cART-treated adults living with HIV. This has increased the incidence of age-related, non-AIDS comorbidities in this population. One of the most common comorbidities is depression, which is also associated with cognitive impairment and a number of neuropathologies. In older people living with HIV, treating these overlapping disorders is complex, often creating pill burden or adverse drug-drug interactions that can exacerbate these neurologic disorders. Depression, NeuroHIV and many of the neuropsychiatric therapeutics used to treat them impact the dopaminergic system, suggesting that dopaminergic dysfunction may be a common factor in the development of these disorders. Further, changes in dopamine can influence the development of inflammation and the regulation of immune function, which are also implicated in the progression of NeuroHIV and depression. Little is known about the optimal clinical management of drug-drug interactions between cART drugs and antidepressants, particularly in regard to dopamine in older people living with HIV. This review will discuss those interactions, first examining the etiology of NeuroHIV and depression in older adults, then discussing the interrelated effects of dopamine and inflammation on these disorders, and finally reviewing the activity and interactions of cART drugs and antidepressants on each of these factors. Developing better strategies to manage these comorbidities is critical to the health of the aging, HIV-infected population, as the older population may be particularly vulnerable to drug-drug interactions affecting dopamine.
Collapse
|
21
|
Nash B, Tarn K, Irollo E, Luchetta J, Festa L, Halcrow P, Datta G, Geiger JD, Meucci O. Morphine-Induced Modulation of Endolysosomal Iron Mediates Upregulation of Ferritin Heavy Chain in Cortical Neurons. eNeuro 2019; 6:ENEURO.0237-19.2019. [PMID: 31300544 PMCID: PMC6675873 DOI: 10.1523/eneuro.0237-19.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 07/01/2019] [Indexed: 01/01/2023] Open
Abstract
HIV-associated neurocognitive disorders (HAND) remain prevalent and are aggravated by µ-opioid use. We have previously shown that morphine and other µ-opioids may contribute to HAND by inhibiting the homeostatic and neuroprotective chemokine receptor CXCR4 in cortical neurons, and this novel mechanism depends on upregulation of the protein ferritin heavy chain (FHC). Here, we examined the cellular events and potential mechanisms involved in morphine-mediated FHC upregulation using rat cortical neurons of either sex in vitro and in vivo. Morphine dose dependently increased FHC protein levels in primary neurons through µ-opioid receptor (µOR) and Gαi-protein signaling. Cytoplasmic FHC levels were significantly elevated, but nuclear FHC levels and FHC gene expression were unchanged. Morphine-treated rats also displayed increased FHC levels in layer 2/3 neurons of the prefrontal cortex. Importantly, both in vitro and in vivo FHC upregulation was accompanied by loss of mature dendritic spines, which was also dependent on µOR and Gαi-protein signaling. Moreover, morphine upregulated ferritin light chain (FLC), a component of the ferritin iron storage complex, suggesting that morphine altered neuronal iron metabolism. Indeed, prior to FHC upregulation, morphine increased cytoplasmic labile iron levels as a function of decreased endolysosomal iron. In line with this, chelation of endolysosomal iron (but not extracellular iron) blocked morphine-induced FHC upregulation and dendritic spine reduction, whereas iron overloading mimicked the effect of morphine on FHC and dendritic spines. Overall, these data demonstrate that iron mediates morphine-induced FHC upregulation and consequent dendritic spine deficits and implicate endolysosomal iron efflux to the cytoplasm in these effects.
Collapse
Affiliation(s)
- Bradley Nash
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Kevin Tarn
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Elena Irollo
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Jared Luchetta
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Lindsay Festa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Peter Halcrow
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Gaurav Datta
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102
| |
Collapse
|
22
|
Servaes S, Glorie D, Stroobants S, Staelens S. Neuroreceptor kinetics in rats repeatedly exposed to quinpirole as a model for OCD. PLoS One 2019; 14:e0213313. [PMID: 30845202 PMCID: PMC6405120 DOI: 10.1371/journal.pone.0213313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 02/19/2019] [Indexed: 11/24/2022] Open
Abstract
Background Obsessive-compulsive disorder (OCD) is a chronic, incapacitating, early onset psychiatric disorder that is characterized by obsessions and compulsions originating from a disturbance in the cortico-striato-thalamico-cortical circuit. We implemented the preclinical quinpirole (QP) rat model for compulsive checking in OCD to analyse the behaviour and visualize the D2R, mGluR5 and GLT1 density in order to contribute to the understanding of the neuroreceptor kinetics. Methods Animals (n = 14) were exposed to either saline (1 mL/kg) or QP (dopamine D2-agonist, 0.5 mg/kg) twice-weekly during 7 weeks. After each injection animals were placed on an open field test. After model setup, animals were placed in a behavioural cage equipped with tracking software and hardware in order to analyse the behaviour. Subsequently, sagittal slides were made of the CP in the right hemisphere and a staining was done with the D2R, mGluR5 and GLT-1 antibody to visualize the corresponding receptor. Results The QP animals displayed a strong increase in travelled distance (+596.70%) and in the number of homebase visits (+1222.90%) compared to the control animals. After chronic exposure to QP, animals had a significantly (p < 0.05) higher percentage of D2R density in the CP (7.92% ± 0.48%) versus 6.66% ± 0.28% in animals treated with saline. There were no differences for mGluR5 and GLT1 receptor density. Conclusions Chronic exposure to QP leads to hyperlocomotion and an increase in D2R density. Furthermore, as mGluR5 and GLT1 density did not seem to be directly affected, decreased levels of glutamate might have influenced the binding potential in earlier reports.
Collapse
Affiliation(s)
- Stijn Servaes
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Dorien Glorie
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Antwerp, Belgium
- Department of Nuclear Medicine, University Hospital Antwerp, Edegem, Antwerp, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Antwerp, Belgium
- * E-mail:
| |
Collapse
|
23
|
Gill AJ, Garza R, Ambegaokar SS, Gelman BB, Kolson DL. Heme oxygenase-1 promoter region (GT)n polymorphism associates with increased neuroimmune activation and risk for encephalitis in HIV infection. J Neuroinflammation 2018; 15:70. [PMID: 29510721 PMCID: PMC5838989 DOI: 10.1186/s12974-018-1102-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/21/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Heme oxygenase-1 (HO-1) is a critical cytoprotective enzyme that limits oxidative stress, inflammation, and cellular injury within the central nervous system (CNS) and other tissues. We previously demonstrated that HO-1 protein expression is decreased within the brains of HIV+ subjects and that this HO-1 reduction correlates with CNS immune activation and neurocognitive dysfunction. To define a potential CNS protective role for HO-1 against HIV, we analyzed a well-characterized HIV autopsy cohort for two common HO-1 promoter region polymorphisms that are implicated in regulating HO-1 promoter transcriptional activity, a (GT)n dinucleotide repeat polymorphism and a single nucleotide polymorphism (A(-413)T). Shorter HO-1 (GT)n repeats and the 'A' SNP allele associate with higher HO-1 promoter activity. METHODS Brain dorsolateral prefrontal cortex tissue samples from an autopsy cohort of HIV-, HIV+, and HIV encephalitis (HIVE) subjects (n = 554) were analyzed as follows: HO-1 (GT)n polymorphism allele lengths were determined by PCR and capillary electrophoresis, A(-413)T SNP alleles were determined by PCR with allele specific probes, and RNA expression of selected neuroimmune markers was analyzed by quantitative PCR. RESULTS HIV+ subjects with shorter HO-1 (GT)n alleles had a significantly lower risk of HIVE; however, shorter HO-1 (GT)n alleles did not correlate with CNS or peripheral viral loads. In HIV+ subjects without HIVE, shorter HO-1 (GT)n alleles associated significantly with lower expression of brain type I interferon response markers (MX1, ISG15, and IRF1) and T-lymphocyte activation markers (CD38 and GZMB). No significant correlations were found between the HO-1 (GT)n repeat length and brain expression of macrophage markers (CD163, CD68), endothelial markers (PECAM1, VWF), the T-lymphocyte marker CD8A, or the B-lymphocyte maker CD19. Finally, we found no significant associations between the A(-413)T SNP and HIVE diagnosis, HIV viral loads, or any neuroimmune markers. CONCLUSION Our data suggest that an individual's HO-1 promoter region (GT)n polymorphism allele repeat length exerts unique modifying risk effects on HIV-induced CNS neuroinflammation and associated neuropathogenesis. Shorter HO-1 (GT)n alleles increase HO-1 promoter activity, which could provide neuroprotection through decreased neuroimmune activation. Therapeutic strategies that induce HO-1 expression could decrease HIV-associated CNS neuroinflammation and decrease the risk for development of HIV neurological disease.
Collapse
Affiliation(s)
- Alexander J. Gill
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 280C Clinical Research Building, Philadelphia, PA 19104 USA
| | - Rolando Garza
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 280C Clinical Research Building, Philadelphia, PA 19104 USA
| | - Surendra S. Ambegaokar
- Department of Botany & Microbiology, Robbins Program in Neuroscience, Ohio Wesleyan University, Delaware, OH 43016 USA
| | - Benjamin B. Gelman
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Dennis L. Kolson
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 280C Clinical Research Building, Philadelphia, PA 19104 USA
| |
Collapse
|
24
|
Gelman BB, Endsley J, Kolson D. When do models of NeuroAIDS faithfully imitate "the real thing"? J Neurovirol 2017; 24:146-155. [PMID: 29256039 PMCID: PMC5910470 DOI: 10.1007/s13365-017-0601-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/03/2017] [Accepted: 11/08/2017] [Indexed: 02/07/2023]
Abstract
HIV-infected patients treated with antiretroviral medicines (ART) still face neurological challenges. HIV-associated neurocognitive disturbances (HAND) can occur, and latent viral DNA persisting in the central nervous system (CNS) prevents eradication of HIV. This communication focuses on how to develop experimental models of HAND and CNS HIV latency that best imitate the CNS pathophysiology in diseased humans, which we take to be “the real thing.” Models of HIV encephalitis (HIVE) with active CNS viral replication were developed in the early years of the AIDS pandemic. The clinical relevancy of such models is in sharp decline because HIVE seldom occurs in virally suppressed patients, while HAND remains common. The search for improved models of HAND should incorporate the neurochemical, neuroimmunological and neuropathological features of virally suppressed patients. Common anomalies in these patients as established in autopsy brain specimens include brain endothelial cell activation and neurochemical imbalances of synaptic transmission; classical neurodegeneration may not be as crucial. With regard to latent HIV with viral suppression, human brain specimens show that the pool of latent proviral HIV DNA in the CNS is relatively small relative to the total body pool and does not change substantially over years. The CNS pool of latent virus probably differs from lymphoid tissues, because the mononuclear phagocyte system sustains productive infection (versus lymphocytes). These and yet-to-be discovered aspects of the human CNS of virally suppressed patients need to be better defined and addressed in experimental models. To maintain clinical relevancy, models of HAND and viral latency should faithfully emulate “the real thing.”
Collapse
Affiliation(s)
- Benjamin B Gelman
- Department of Pathology, Route 0419, University of Texas Medical Branch, Galveston, TX, 77555-0419, USA.
| | - Janice Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Dennis Kolson
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104-6140, USA
| |
Collapse
|
25
|
Khodr CE, Chen L, Al-Harthi L, Hu XT. Aging alters voltage-gated calcium channels in prefrontal cortex pyramidal neurons in the HIV brain. J Neurovirol 2017; 24:113-118. [PMID: 29090376 DOI: 10.1007/s13365-017-0588-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/14/2017] [Accepted: 10/05/2017] [Indexed: 10/18/2022]
Abstract
We assessed firing and voltage-gated Ca2+ influx in medial prefrontal cortex (mPFC) pyramidal neurons from older (12 months old) HIV-1 transgenic (Tg) rats. We found that neurons from older Tg rats showed increased firing compared to non-Tg rats, but Ca2+ spikes were unchanged. However, stronger excitatory stimulation was needed to evoke Ca2+ spikes, which was associated with reduced mPFC Cav1.2 L-type Ca2+ channel (L-channel) protein. In contrast, L-channel protein was unaltered in younger (6-7 weeks old) Tg rats, which we previously found had enhanced neuronal Ca2+ influx. These studies demonstrate that aging alters HIV-induced Ca2+ channel dysfunction that affects mPFC activity.
Collapse
Affiliation(s)
- Christina E Khodr
- Department of Immunology and Microbiology, Rush University Medical Center, Cohn Research Building, Rm.414, 1735 W. Harrison Street, Chicago, IL, 60612, USA
| | - Lihua Chen
- Department of Immunology and Microbiology, Rush University Medical Center, Cohn Research Building, Rm.414, 1735 W. Harrison Street, Chicago, IL, 60612, USA
| | - Lena Al-Harthi
- Department of Immunology and Microbiology, Rush University Medical Center, Cohn Research Building, Rm.414, 1735 W. Harrison Street, Chicago, IL, 60612, USA
| | - Xiu-Ti Hu
- Department of Immunology and Microbiology, Rush University Medical Center, Cohn Research Building, Rm.414, 1735 W. Harrison Street, Chicago, IL, 60612, USA.
| |
Collapse
|
26
|
Xu C, Hermes DJ, Nwanguma B, Jacobs IR, Mackie K, Mukhopadhyay S, Lichtman AH, Ignatowska-Jankowska B, Fitting S. Endocannabinoids exert CB 1 receptor-mediated neuroprotective effects in models of neuronal damage induced by HIV-1 Tat protein. Mol Cell Neurosci 2017; 83:92-102. [PMID: 28733129 DOI: 10.1016/j.mcn.2017.07.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 07/07/2017] [Accepted: 07/14/2017] [Indexed: 01/25/2023] Open
Abstract
In the era of combined antiretroviral therapy (cART), human immunodeficiency virus type 1 (HIV-1) is considered a chronic disease that specifically targets the brain and causes HIV-1-associated neurocognitive disorders (HAND). Endocannabinoids (eCBs) elicit neuroprotective and anti-inflammatory actions in several central nervous system (CNS) disease models, but their effects in HAND remain unknown. HIV-1 does not infect neurons, but produces viral toxins, such as transactivator of transcription (Tat), that disrupt neuronal calcium equilibrium and give rise to synaptodendritic injuries and cell death, the former being highly correlated with HAND. Consequently, we tested whether the eCBs N-arachidonoylethanolamine (anandamide/AEA) and 2-arachidonoyl-glycerol (2-AG) offer neuroprotective actions in a neuronal culture model. Specifically, we examined the neuroprotective actions of these eCBs on Tat excitotoxicity in primary cultures of prefrontal cortex neurons (PFC), and whether cannabinoid receptors mediate this neuroprotection. Tat-induced excitotoxicity was reflected by increased intracellular calcium levels, synaptodendritic damage, neuronal excitability, and neuronal death. Further, upregulation of cannabinoid 1 receptor (CB1R) protein levels was noted in the presence of HIV-1 Tat. The direct application of AEA and 2-AG reduced excitotoxic levels of intracellular calcium and promoted neuronal survival following Tat exposure, which was prevented by the CB1R antagonist rimonabant, but not by the CB2R antagonist AM630. Overall, our findings indicate that eCBs protect PFC neurons from Tat excitotoxicity in vitro via a CB1R-related mechanism. Thus, the eCB system possesses promising targets for treatment of neurodegenerative disorders associated with HIV-1 infection.
Collapse
Affiliation(s)
- Changqing Xu
- Department of Psychology & Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - Douglas J Hermes
- Department of Psychology & Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - Blessing Nwanguma
- Department of Chemistry & Biochemistry, North Carolina Central University, Durham, NC, USA
| | - Ian R Jacobs
- Department of Psychology & Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - Kenneth Mackie
- Department of Psychological & Brain Science, Indiana University, Bloomington, IN, USA
| | - Somnath Mukhopadhyay
- Department of Chemistry & Biochemistry, North Carolina Central University, Durham, NC, USA
| | - Aron H Lichtman
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Sylvia Fitting
- Department of Psychology & Neuroscience, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
27
|
Xu C, Fitting S. Inhibition of GABAergic Neurotransmission by HIV-1 Tat and Opioid Treatment in the Striatum Involves μ-Opioid Receptors. Front Neurosci 2016; 10:497. [PMID: 27877102 PMCID: PMC5099255 DOI: 10.3389/fnins.2016.00497] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 10/17/2016] [Indexed: 01/07/2023] Open
Abstract
Due to combined antiretroviral therapy (cART), human immunodeficiency virus type 1 (HIV-1) is considered a chronic disease with high prevalence of mild forms of neurocognitive impairments, also referred to as HIV-associated neurocognitive disorders (HAND). Although opiate drug use can exacerbate HIV-1 Tat-induced neuronal damage, it remains unknown how and to what extent opioids interact with Tat on the GABAergic system. We conducted whole-cell recordings in mouse striatal slices and examined the effects of HIV-1 Tat in the presence and absence of morphine (1 μM) and damgo (1 μM) on GABAergic neurotransmission. Results indicated a decrease in the frequency and amplitude of spontaneous inhibitory postsynaptic currents (sIPSCs) and miniature IPSCs (mIPSCs) by Tat (5–50 nM) in a concentration-dependent manner. The significant Tat-induced decrease in IPSCs was abolished when removing extracellular and/or intracellular calcium. Treatment with morphine or damgo alone significantly decreased the frequency, but not amplitude of IPSCs. Interestingly, morphine but not damgo indicated an additional downregulation of the mean frequency of mIPSCs in combination with Tat. Pretreatment with naloxone (1 μM) and CTAP (1 μM) prevented the Tat-induced decrease in sIPSCs frequency but only naloxone prevented the combined Tat and morphine effect on mIPSCs frequency. Results indicate a Tat- or opioid-induced decrease in GABAergic neurotransmission via μ-opioid receptors with combined Tat and morphine effects involving additional opioid receptor-related mechanisms. Exploring the interactions between Tat and opioids on the GABAergic system may help to guide future research on HAND in the context of opiate drug use.
Collapse
Affiliation(s)
- Changqing Xu
- Department of Psychology and Neuroscience, University of North Carolina Chapel Hill Chapel Hill, NC, USA
| | - Sylvia Fitting
- Department of Psychology and Neuroscience, University of North Carolina Chapel Hill Chapel Hill, NC, USA
| |
Collapse
|
28
|
Wächter C, Eiden LE, Naumann N, Depboylu C, Weihe E. Loss of cerebellar neurons in the progression of lentiviral disease: effects of CNS-permeant antiretroviral therapy. J Neuroinflammation 2016; 13:272. [PMID: 27737697 PMCID: PMC5064958 DOI: 10.1186/s12974-016-0726-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 09/20/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The majority of investigations on HIV-associated neurocognitive disorders (HAND) neglect the cerebellum in spite of emerging evidence for its role in higher cognitive functions and dysfunctions in common neurodegenerative diseases. METHODS We systematically investigated the molecular and cellular responses of the cerebellum as contributors to lentiviral infection-induced neurodegeneration, in the simian immunodeficiency virus (SIV)-infected rhesus macaque model for HIV infection and HAND. Four cohorts of animals were studied: non-infected controls, SIV-infected asymptomatic animals, and SIV-infected AIDS-diseased animals with and without brain-permeant antiretroviral treatment. The antiretroviral utilized was 6-chloro-2',3'-dideoxyguanosine (6-Cl-ddG), a CNS-permeable nucleoside reverse transcriptase inhibitor. Quantitation of granule cells and Purkinje cells, of an established biomarker of SIV infection (gp41), of microglial/monocyte/macrophage markers (IBA-1, CD68, CD163), and of the astroglial marker (GFAP) were used to reveal cell-specific cerebellar responses to lentiviral infection and antiretroviral therapy (ART). The macromolecular integrity of the blood brain barrier was tested by albumin immunohistochemistry. RESULTS Productive CNS infection was observed in the symptomatic stage of disease, and correlated with extensive microglial/macrophage and astrocyte activation, and widespread macromolecular blood brain barrier defects. Signs of productive infection, and inflammation, were reversed upon treatment with 6-Cl-ddG, except for a residual low-grade activation of microglial cells and astrocytes. There was an extensive loss of granule cells in the SIV-infected asymptomatic cohort, which was further increased in the symptomatic stage of the disease and persisted after 6-Cl-ddG (administered after the onset of symptoms of AIDS). In the symptomatic stage, Purkinje cell density was reduced. Purkinje cell loss was likewise unaffected by 6-Cl-ddG treatment at this time. CONCLUSIONS Our findings suggest that neurodegenerative mechanisms are triggered by SIV infection early in the disease process, i. e., preceding large-scale cerebellar productive infection and marked neuroinflammation. These affect primarily granule cells early in disease, with later involvement of Purkinje cells, indicating differential vulnerability of the two neuronal populations. The results presented here indicate a role for the cerebellum in neuro-AIDS. They also support the conclusion that, in order to attenuate the development of motor and cognitive dysfunctions in HIV-positive individuals, CNS-permeant antiretroviral therapy combined with anti-inflammatory and neuroprotective treatment is indicated even before overt signs of CNS inflammation occur.
Collapse
Affiliation(s)
- Christian Wächter
- Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps University Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany
| | - Lee E Eiden
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - Nedye Naumann
- Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps University Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany
| | - Candan Depboylu
- Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps University Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany.,Experimental Neurology, Department of Neurology, Philipps University Marburg, Marburg, Germany
| | - Eberhard Weihe
- Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps University Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany.
| |
Collapse
|
29
|
Khodr CE, Chen L, Dave S, Al-Harthi L, Hu XT. Combined chronic blockade of hyper-active L-type calcium channels and NMDA receptors ameliorates HIV-1 associated hyper-excitability of mPFC pyramidal neurons. Neurobiol Dis 2016; 94:85-94. [PMID: 27326669 PMCID: PMC4983475 DOI: 10.1016/j.nbd.2016.06.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/04/2016] [Accepted: 06/16/2016] [Indexed: 01/09/2023] Open
Abstract
Human Immunodeficiency Virus type 1 (HIV-1) infection induces neurological and neuropsychological deficits, which are associated with dysregulation of the medial prefrontal cortex (mPFC) and other vulnerable brain regions. We evaluated the impact of HIV infection in the mPFC and the therapeutic potential of targeting over-active voltage-gated L-type Ca(2+) channels (L-channel) and NMDA receptors (NMDAR), as modeled in HIV-1 transgenic (Tg) rats. Whole-cell patch-clamp recording was used to assess the membrane properties and voltage-sensitive Ca(2+) potentials (Ca(2+) influx) in mPFC pyramidal neurons. Neurons from HIV-1 Tg rats displayed reduced rheobase, spike amplitude and inwardly-rectifying K(+) influx, increased numbers of action potentials, and a trend of aberrant firing compared to those from non-Tg control rats. Neuronal hyper-excitation was associated with abnormally-enhanced Ca(2+) influx (independent of NMDAR), which was eliminated by acute L-channel blockade. Combined chronic blockade of over-active L-channels and NMDARs with open-channel blockers abolished HIV effects on spiking, aberrant firing and Ca(2+) potential half-amplitude duration, though not the reduced inward rectification. In contrast, individual chronic blockade of over-active L-channels or NMDARs did not alleviate HIV-induced mPFC hyper-excitability. These studies demonstrate that HIV alters mPFC neuronal activity by dysregulating membrane excitability and Ca(2+) influx through the L-channels. This renders these neurons more susceptible and vulnerable to excitatory stimuli, and could contribute to HIV-associated neuropathogenesis. Combined targeting of over-active L-channels/NMDARs alleviates HIV-induced dysfunction of mPFC pyramidal neurons, emphasizing a potential novel therapeutic strategy that may effectively decrease HIV-induced Ca(2+) dysregulation in the mPFC.
Collapse
Affiliation(s)
- Christina E Khodr
- Department of Pharmacology, Rush University Medical Center, Chicago, IL 60612, United States
| | - Lihua Chen
- Department of Pharmacology, Rush University Medical Center, Chicago, IL 60612, United States
| | - Sonya Dave
- Department of Pharmacology, Rush University Medical Center, Chicago, IL 60612, United States
| | - Lena Al-Harthi
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, IL 60612, United States
| | - Xiu-Ti Hu
- Department of Pharmacology, Rush University Medical Center, Chicago, IL 60612, United States.
| |
Collapse
|