1
|
Zhou L, Godse S, Sinha N, Kodidela S, Singh U, Kumar S. Darunavir Nanoformulation Suppresses HIV Pathogenesis in Macrophages and Improves Drug Delivery to the Brain in Mice. Pharmaceutics 2024; 16:555. [PMID: 38675216 PMCID: PMC11054602 DOI: 10.3390/pharmaceutics16040555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/01/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Although antiretroviral therapy (ART) can suppress peripheral HIV, patients still suffer from neuroHIV due to insufficient levels of ART drugs in the brain. Hence, this study focuses on developing a poly lactic-co-glycolic acid (PLGA) nanoparticle-based ART drug delivery system for darunavir (DRV) using an intranasal route that can overcome the limitation of drug metabolic stability and blood-brain barrier (BBB) permeability. The physicochemical properties of PLGA-DRV were characterized. The results indicated that PLGA-DRV formulation inhibits HIV replication in U1 macrophages directly and in the presence of the BBB without inducing cytotoxicity. However, the PLGA-DRV did not inhibit HIV replication more than DRV alone. Notably, the total antioxidant capacity remained unchanged upon treatment with both DRV or PLGA-DRV in U1 cells. Compared to DRV alone, PLGA-DRV further decreased reactive oxygen species, suggesting a decrease in oxidative stress by the formulation. Oxidative stress is generally increased by HIV infection, leading to increased inflammation. Although the PLGA-DRV formulation did not further reduce the inflammatory response, the formulation did not provoke an inflammatory response in HIV-infected U1 macrophages. As expected, in vitro experiments showed higher DRV permeability by PLGA-DRV than DRV alone to U1 macrophages. Importantly, in vivo experiments, especially using intranasal administration of PLGA-DRV in wild-type mice, demonstrated a significant increase in the brain-to-plasma ratio of DRV compared to the free DRV. Overall, findings from this study attest to the potential of the PLGA-DRV nanoformulation in reducing HIV pathogenesis in macrophages and enhancing drug delivery to the brain, offering a promising avenue for treating HIV-related neurological disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Santosh Kumar
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave., Memphis, TN 38163, USA (S.G.); (U.S.)
| |
Collapse
|
2
|
Gunaydin-Akyildiz A, Aksoy N, Boran T, Ilhan EN, Ozhan G. Favipiravir induces oxidative stress and genotoxicity in cardiac and skin cells. Toxicol Lett 2022; 371:9-16. [PMID: 36152797 PMCID: PMC9492396 DOI: 10.1016/j.toxlet.2022.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/07/2022] [Accepted: 09/20/2022] [Indexed: 11/03/2022]
Abstract
Favipiravir (T-705), used against influenza viruses, is approved for emergency use in many countries for the treatment of COVID-19. The frequent adverse effects of favipiravir are related with the gastrointestinal system, however, studies suggest a positive association of favipiravir on QTc prolongation, which can cause cardiotoxicity. Also, there are reports of skin reactions such as angioedema due to favipiravir. Despite the several adverse effects, studies examining the drug's effects at the molecular level are insufficient, e.g., the genotoxic and oxidative stress-inducing effects of favipiravir, which are among the primary mechanisms of drug-induced toxicity. The cytotoxicity of favipiravir was analyzed with the measurement of the ATP content in H9c2 cardiomyoblasts and CCD-1079Sk skin fibroblasts. The ATP level decreased starting from 200 µM. The inhibitory effect on the mitochondrial electron transport chain enzymes complex I and complex V was also evaluated where favipiravir showed significant enzyme inhibitory effects in the highest concentration studied. A molecular docking study evaluating the interaction between favipiravir-RTP and mitochondrial DNA polymerase (POLG1) was done. The relationship of favipiravir with oxidative stress was examined by measuring glutathione (GSH) and protein carbonyl levels which were observed higher after drug treatment compared to the control group. The genotoxicity study was done using the Comet assay and increase in DNA tail has been detected. Furthermore, 8-OHdG levels were measured higher in favipiravir treated cells indicating oxidative DNA damage. Favipiravir induced oxidative stress leading to DNA damage in cardiomyoblast cells and fibroblastic skin cells. Oxidative stress and DNA damage might eventually lead to organ-specific damage such as cardiotoxicity and dermal toxicity. Considering the increased use of favipiravir in recent years, and that oxidative stress and genotoxicity are two important indicators of drug-induced toxicity, the obtained results are worth attention.
Collapse
Affiliation(s)
- Aysenur Gunaydin-Akyildiz
- Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34093 Istanbul, Turkey.
| | - Nergis Aksoy
- Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34093 Istanbul, Turkey
| | - Tugce Boran
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey
| | - Emine Nihan Ilhan
- Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34093 Istanbul, Turkey
| | - Gul Ozhan
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey
| |
Collapse
|
3
|
Combination Antiretroviral Therapy (cART) in Diabetes Exacerbates Diabetogenic Effects on Hippocampal Microstructure, Neurogenesis and Cytokine Perturbation in Male Sprague Dawley Rats. Diagnostics (Basel) 2022; 12:diagnostics12040905. [PMID: 35453953 PMCID: PMC9029837 DOI: 10.3390/diagnostics12040905] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/14/2022] [Accepted: 04/01/2022] [Indexed: 02/04/2023] Open
Abstract
The increasing incidence of diabetes and HIV/AIDS–diabetes comorbidity in society has led to the prevalence of combination antiretroviral therapy (cART) in diabetes, with some reported neural effects. Therefore, the effects of cART and type two diabetes (T2D) on the hippocampal levels of cytokines, lipid peroxidation; histomorphology and neurogenesis were investigated. Adult male Sprague–Dawley rats were divided into four groups: DB (diabetic rats); DAV (diabetic rats treated with cART (efavirenz, emtricitabine and tenofovir); AV (normal rats treated with cART) and the NC group (with no treatment). Following ninety days of treatment, the rats were terminated, and the brains excised. Immunoassay (IL-1α, IL-6, TNFα and MDA); immunohistochemical (Ki67 and DCX) and cresyl violet histomorphology analyses were carried out on brain homogenates and sections, respectively. In comparison to the control, the results showed that cART significantly elevated the IL-6, TNFα and MDA levels, while DB and DAV significantly reduced the body weight, glucose tolerance, IL-1α, IL-6, TNFα and MDA levels. The hippocampal neuronal number was reduced in AV (dentate gyrus; DG region), in the DB group (Cornu Ammonis subregion 1; CA1 and DG regions only) and in DAV (all three hippocampal regions). Additionally, the expression of neurogenic markers Ki67 and doublecortin (DCX) were reduced in the diabetic group, with a greater reduction in the cART+T2D group compared to the control. Furthermore, the neuronal number at all hippocampal regions was negatively corelated with the diabetic parameters (FBG; fasting blood glucose, NFBG; non-fasting blood glucose, AUC; area under the glucose tolerance curve) but positively correlated with body weight. Additionally, the increase in the DG neuronal nuclei area of DB and DAV was significantly positively correlated with FBG, NFBG and AUC and inversely correlated with the estimated number of neurons and neurogenesis. These findings indicate that cART in diabetes (DAV) has similar effects as diabetes relative to the induction of oxidative stress and impairment of the cytokine immune response, but exacerbated neurotoxicity is observed in DAV, as shown by a significantly decreased DCX expression compared to DB and reduction in the number of Cornu Ammonis subregion 3 (CA3) hippocampal neurons, unlike in cART or the diabetes-alone groups.
Collapse
|
4
|
Bryant J, Andhavarapu S, Bever C, Guda P, Katuri A, Gupta U, Arvas M, Asemu G, Heredia A, Gerzanich V, Simard JM, Makar TK. 7,8-Dihydroxyflavone improves neuropathological changes in the brain of Tg26 mice, a model for HIV-associated neurocognitive disorder. Sci Rep 2021; 11:18519. [PMID: 34531413 PMCID: PMC8446048 DOI: 10.1038/s41598-021-97220-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 08/04/2021] [Indexed: 02/08/2023] Open
Abstract
The combined antiretroviral therapy era has significantly increased the lifespan of people with HIV (PWH), turning a fatal disease to a chronic one. However, this lower but persistent level of HIV infection increases the susceptibility of HIV-associated neurocognitive disorder (HAND). Therefore, research is currently seeking improved treatment for this complication of HIV. In PWH, low levels of brain derived neurotrophic factor (BDNF) has been associated with worse neurocognitive impairment. Hence, BDNF administration has been gaining relevance as a possible adjunct therapy for HAND. However, systemic administration of BDNF is impractical because of poor pharmacological profile. Therefore, we investigated the neuroprotective effects of BDNF-mimicking 7,8 dihydroxyflavone (DHF), a bioactive high-affinity TrkB agonist, in the memory-involved hippocampus and brain cortex of Tg26 mice, a murine model for HAND. In these brain regions, we observed astrogliosis, increased expression of chemokine HIV-1 coreceptors CXCR4 and CCR5, neuroinflammation, and mitochondrial damage. Hippocampi and cortices of DHF treated mice exhibited a reversal of these pathological changes, suggesting the therapeutic potential of DHF in HAND. Moreover, our data indicates that DHF increases the phosphorylation of TrkB, providing new insights about the role of the TrkB-Akt-NFkB signaling pathway in mediating these pathological hallmarks. These findings guide future research as DHF shows promise as a TrkB agonist treatment for HAND patients in adjunction to the current antiviral therapies.
Collapse
Affiliation(s)
- Joseph Bryant
- Institute of Human Virology, Baltimore, MD, 21201, USA
| | | | - Christopher Bever
- Research Service, Veterans Affairs Center, Baltimore, MD, 21201, USA
| | | | - Akhil Katuri
- Institute of Human Virology, Baltimore, MD, 21201, USA
| | - Udit Gupta
- Institute of Human Virology, Baltimore, MD, 21201, USA
| | | | - Girma Asemu
- Institute of Human Virology, Baltimore, MD, 21201, USA
| | | | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland, Baltimore, MD, 21201, USA
| | - J Marc Simard
- Research Service, Veterans Affairs Center, Baltimore, MD, 21201, USA
- Department of Neurosurgery, University of Maryland, Baltimore, MD, 21201, USA
| | - Tapas Kumar Makar
- Institute of Human Virology, Baltimore, MD, 21201, USA.
- Research Service, Veterans Affairs Center, Baltimore, MD, 21201, USA.
| |
Collapse
|
5
|
Singh S, Ghosh S, Pal VK, Munshi M, Shekar P, Narasimha Murthy DT, Mugesh G, Singh A. Antioxidant nanozyme counteracts HIV-1 by modulating intracellular redox potential. EMBO Mol Med 2021; 13:e13314. [PMID: 33793064 PMCID: PMC8103102 DOI: 10.15252/emmm.202013314] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 02/05/2021] [Accepted: 02/19/2021] [Indexed: 12/23/2022] Open
Abstract
Reactive oxygen species (ROS) regulates the replication of human immunodeficiency virus (HIV‐1) during infection. However, the application of this knowledge to develop therapeutic strategies remained unsuccessful due to the harmful consequences of manipulating cellular antioxidant systems. Here, we show that vanadium pentoxide (V2O5) nanosheets functionally mimic natural glutathione peroxidase activity to mitigate ROS associated with HIV‐1 infection without adversely affecting cellular physiology. Using genetic reporters of glutathione redox potential and hydrogen peroxide, we showed that V2O5 nanosheets catalyze ROS neutralization in HIV‐1‐infected cells and uniformly block viral reactivation and replication. Mechanistically, V2O5 nanosheets suppressed HIV‐1 by affecting the expression of pathways coordinating redox balance, virus transactivation (e.g., NF‐κB), inflammation, and apoptosis. Importantly, a combination of V2O5 nanosheets with a pharmacological inhibitor of NF‐κB (BAY11‐7082) abrogated reactivation of HIV‐1. Lastly, V2O5 nanosheets inhibit viral reactivation upon prostratin stimulation of latently infected CD4+ T cells from HIV‐infected patients receiving suppressive antiretroviral therapy. Our data successfully revealed the usefulness of V2O5 nanosheets against HIV and suggested nanozymes as future platforms to develop interventions against infectious diseases.
Collapse
Affiliation(s)
- Shalini Singh
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India.,Centre for Infectious Disease Research (CIDR), Indian Institute of Science, Bangalore, India
| | - Sourav Ghosh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, India
| | - Virender Kumar Pal
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India.,Centre for Infectious Disease Research (CIDR), Indian Institute of Science, Bangalore, India
| | - MohamedHusen Munshi
- Centre for Infectious Disease Research (CIDR), Indian Institute of Science, Bangalore, India
| | - Pooja Shekar
- Bangalore Medical College and Research Institute, Bangalore, India
| | | | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, India
| | - Amit Singh
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India.,Centre for Infectious Disease Research (CIDR), Indian Institute of Science, Bangalore, India
| |
Collapse
|
6
|
Bertrand L, Velichkovska M, Toborek M. Cerebral Vascular Toxicity of Antiretroviral Therapy. J Neuroimmune Pharmacol 2021; 16:74-89. [PMID: 31209776 PMCID: PMC7952282 DOI: 10.1007/s11481-019-09858-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/27/2019] [Indexed: 01/14/2023]
Abstract
HIV infection is associated with comorbidities that are likely to be driven not only by HIV itself, but also by the toxicity of long-term use of antiretroviral therapy (ART). Indeed, increasing evidence demonstrates that the antiretroviral drugs used for HIV treatment have toxic effects resulting in various cellular and tissue pathologies. The blood-brain barrier (BBB) is a modulated anatomophysiological interface which separates and controls substance exchange between the blood and the brain parenchyma; therefore, it is particularly exposed to ART-induced toxicity. Balancing the health risks and gains of ART has to be considered in order to maximize the positive effects of therapy. The current review discusses the cerebrovascular toxicity of ART, with the focus on mitochondrial dysfunction. Graphical Abstract Graphical representation of the interactions between HIV, antiretroviral therapy (ART), and the blood-brain barrier (BBB).
Collapse
Affiliation(s)
- Luc Bertrand
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Room 528, 1011 NW 15th Street, Miami, FL, 33136, USA
| | - Martina Velichkovska
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Room 528, 1011 NW 15th Street, Miami, FL, 33136, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Room 528, 1011 NW 15th Street, Miami, FL, 33136, USA.
| |
Collapse
|
7
|
Olufunke D, Edidiong A, Oluwatomisin F, Alani A. Therapeutic activities of naringenin on efavirenz-induced sleep-like disorder in the midbrain of white albino mice. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:1462-1470. [PMID: 33235704 PMCID: PMC7671428 DOI: 10.22038/ijbms.2020.47043.10852] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Efavirenz, has proven to be effective in suppressing human immunodeficiency virus (HIV) viral load; however, complaints of sleep disorders including hallucination, and insomnia have greatly contributed to non-adherence to antiretroviral therapy. This study aimed at investigating therapeutic activities of naringenin on efavirenz-induced sleep disorder. MATERIALS AND METHODS Sixty mice were divided into six groups of control, combination antiretroviral therapy (cART), efavirenz, naringenin, naringenin/efavirenz and naringenin/cART. Efavirenz, cART, and naringenin were administered orally and daily at 15 mg/kg, 24 mg/kg and 50 mg/kg, respectively for 28 days. Post neurobehavioral test, oxidative stress, histology and immunohistochemistry for dopamine were carried out after administration process. RESULTS Efavirenz (P<0.0001) and cART (P<0.01) significantly increased immobility during open field (P<0.01), escape time in seconds (sec) in Morris water maze (P<0.001) and numbers of head-twitch response (HTR) (P<0.0001). Similarly, there was a significant increase in malondialdehyde (MDA) (P<0.0001) and decreased superoxide dismutase (SOD) (P<0.001) and reduced glutathione (GSH) (P<0.001); however, naringenin-treated groups potentiated anti-oxidant function by reducing oxidative stress (P<0.01). Histological evaluation demonstrated severe neurodegeneration, vacuolization and pyknosis in efavirenz and cART compared to naringenin groups. Dopaminergic neurons using immunohistochemial antibody (tyrosine hydroxylase) staining showed poor immunoreactivity in efavirenz and cART in contrast to naringenin groups. CONCLUSION Efavirenz and cART have the potential of inducing sleep disorder possibly due to their capability to trigger inflammation and deplete dopamine level. However, naringenin has proven to be effective in ameliorating these damages.
Collapse
Affiliation(s)
- Dosumu Olufunke
- Department of Anatomy, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Akang Edidiong
- Department of Anatomy, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Faniyan Oluwatomisin
- Department of Anatomy, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Akanmu Alani
- Department of Haematology and Blood Transfusion, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| |
Collapse
|
8
|
Wu S, Yang S, Bloe CB, Zhuang R, Huang J, Zhang W. Identification of Key Genes and Pathways in Mouse Spinal Cord Involved in ddC-Induced Neuropathic Pain by Transcriptome Sequencing. J Mol Neurosci 2020; 71:651-661. [PMID: 32812184 DOI: 10.1007/s12031-020-01686-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/11/2020] [Indexed: 01/12/2023]
Abstract
Highly active antiretroviral therapy (HAART) works effectively in inhibiting HIV replication in patients. However, the use of nucleoside reverse transcriptase inhibitors (NRTIs) often causes side effects of neuropathic pain, and its mechanism remains to be elucidated. Therefore, we aim to explore the mechanism of NRTIs-induced neuropathic pain at the transcriptome level. C57BL/6 J mice were given intraperitoneal injection of zalcitabine (ddC) or saline (control) for 2 weeks, during which the mechanical pain threshold of the mice was detected by von Frey test. Then the L3~L5 spinal segments of the mice were isolated and subsequently used for RNA sequencing (RNA-seq) on the last day of treatment. The mechanical pain threshold of mice given ddC decreased significantly. Compared with the control group, ddC caused significant changes in the expression of 135 genes, of which 66 upregulated and 69 downregulated. Enrichment analysis showed that the functions of these genes are mainly enriched in regulation of transcription, multicellular organism development, and cell differentiation, and the pathway is mainly enriched in the cGMP-PKG signaling pathway and AMPK signaling pathway. Furthermore, key genes such as Gabrd, Kcnd3, Npcd, Insr, Lypd6, Scd2, and Mef2d were also identified. These may serve as drug targets for the prevention or treatment of NRTI-induced neuropathic pain.
Collapse
Affiliation(s)
- Shengjun Wu
- Clinical Laboratory of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Su Yang
- Clinical Laboratory of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Chris Bloe Bloe
- College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Renjie Zhuang
- The Affiliated Hospital of Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Jian Huang
- College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Wenping Zhang
- College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| |
Collapse
|
9
|
Sanna MD, Manassero G, Vercelli A, Herdegen T, Galeotti N. The isoform-specific functions of the c-Jun N-terminal kinase (JNK) in a mouse model of antiretroviral-induced painful peripheral neuropathy. Eur J Pharmacol 2020; 880:173161. [DOI: 10.1016/j.ejphar.2020.173161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/30/2020] [Accepted: 04/28/2020] [Indexed: 01/26/2023]
|
10
|
Anderson AP, Luo X, Russell W, Yin YW. Oxidative damage diminishes mitochondrial DNA polymerase replication fidelity. Nucleic Acids Res 2020; 48:817-829. [PMID: 31799610 PMCID: PMC6954441 DOI: 10.1093/nar/gkz1018] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/12/2019] [Accepted: 11/22/2019] [Indexed: 12/20/2022] Open
Abstract
Mitochondrial DNA (mtDNA) resides in a high ROS environment and suffers more mutations than its nuclear counterpart. Increasing evidence suggests that mtDNA mutations are not the results of direct oxidative damage, rather are caused, at least in part, by DNA replication errors. To understand how the mtDNA replicase, Pol γ, can give rise to elevated mutations, we studied the effect of oxidation of Pol γ on replication errors. Pol γ is a high fidelity polymerase with polymerase (pol) and proofreading exonuclease (exo) activities. We show that Pol γ exo domain is far more sensitive to oxidation than pol; under oxidative conditions, exonuclease activity therefore declines more rapidly than polymerase. The oxidized Pol γ becomes editing-deficient, displaying a 20-fold elevated mutations than the unoxidized enzyme. Mass spectrometry analysis reveals that Pol γ exo domain is a hotspot for oxidation. The oxidized exo residues increase the net negative charge around the active site that should reduce the affinity to mismatched primer/template DNA. Our results suggest that the oxidative stress induced high mutation frequency on mtDNA can be indirectly caused by oxidation of the mitochondrial replicase.
Collapse
Affiliation(s)
- Andrew P Anderson
- Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX 77030, USA
- Sealy Center for Structural Biology, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Xuemei Luo
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - William Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Y Whitney Yin
- Sealy Center for Structural Biology, University of Texas Medical Branch, Galveston, TX 77550, USA
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77550, USA
| |
Collapse
|
11
|
Cotto B, Natarajanseenivasan K, Langford D. HIV-1 infection alters energy metabolism in the brain: Contributions to HIV-associated neurocognitive disorders. Prog Neurobiol 2019; 181:101616. [PMID: 31108127 PMCID: PMC6742565 DOI: 10.1016/j.pneurobio.2019.101616] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/17/2019] [Accepted: 05/13/2019] [Indexed: 12/17/2022]
Abstract
The brain is particularly sensitive to changes in energy supply. Defects in glucose utilization and mitochondrial dysfunction are hallmarks of nearly all neurodegenerative diseases and are also associated with the cognitive decline that occurs as the brain ages. Chronic neuroinflammation driven by glial activation is commonly implicated as a contributing factor to neurodegeneration and cognitive impairment. Human immunodeficiency virus-1 (HIV-1) disrupts normal brain homeostasis and leads to a spectrum of HIV-associated neurocognitive disorders (HAND). HIV-1 activates stress responses in the brain and triggers a state of chronic neuroinflammation. Growing evidence suggests that inflammatory processes and bioenergetics are interconnected in the propagation of neuronal dysfunction. Clinical studies of people living with HIV and basic research support the notion that HIV-1 creates an environment in the CNS that interrupts normal metabolic processes at the cellular level to collectively alter whole brain metabolism. In this review, we highlight reports of abnormal brain metabolism from clinical studies and animal models of HIV-1. We also describe diverse CNS cell-specific changes in bioenergetics associated with HIV-1. Moreover, we propose that attention should be given to adjunctive therapies that combat sources of metabolic dysfunction as a mean to improve and/or prevent neurocognitive impairments.
Collapse
Affiliation(s)
- Bianca Cotto
- Lewis Katz School of Medicine at Temple University, Department of Neuroscience and Center for Neurovirology, Philadelphia, PA, 19140, USA.
| | - Kalimuthusamy Natarajanseenivasan
- Lewis Katz School of Medicine at Temple University, Department of Neuroscience and Center for Neurovirology, Philadelphia, PA, 19140, USA.
| | - Dianne Langford
- Lewis Katz School of Medicine at Temple University, Department of Neuroscience and Center for Neurovirology, Philadelphia, PA, 19140, USA.
| |
Collapse
|
12
|
Rozzi SJ, Avdoshina V, Fields JA, Mocchetti I. Human immunodeficiency virus Tat impairs mitochondrial fission in neurons. Cell Death Discov 2018. [PMID: 29531805 PMCID: PMC5841280 DOI: 10.1038/s41420-017-0013-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Human immunodeficiency virus-1 (HIV) infection of the central nervous system promotes neuronal injury that culminates in HIV-associated neurocognitive disorders. Viral proteins, including transactivator of transcription (Tat), have emerged as leading candidates to explain HIV-mediated neurotoxicity, though the mechanisms remain unclear. Tat transgenic mice or neurons exposed to Tat, which show neuronal loss, exhibit smaller mitochondria as compared to controls. To provide an experimental clue as to which mechanisms are used by Tat to promote changes in mitochondrial morphology, rat cortical neurons were exposed to Tat (100 nM) for various time points. Within 30 min, Tat caused a significant reduction in mitochondrial membrane potential, a process that is regulated by fusion and fission. To further assess whether Tat changes these processes, fission and fusion proteins dynamin-related protein 1 (Drp1) and mitofusin-2 (Mfn2), respectively, were measured. We found that Drp1 levels increased beginning at 2 h after Tat exposure while Mfn2 remained unchanged. Moreover, increased levels of an active form of Drp1 were found to be present following Tat exposure. Furthermore, Drp1 and calcineurin inhibitors prevented Tat-mediated effects on mitochondria size. These findings indicate that mitochondrial fission is likely the leading factor in Tat-mediated alterations to mitochondrial morphology. This disruption in mitochondria homeostasis may contribute to the instability of the organelle and ultimately neuronal cell death following Tat exposure.
Collapse
Affiliation(s)
- Summer J Rozzi
- 1Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC USA.,2Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Medical Center, Washington, DC USA
| | - Valeria Avdoshina
- 2Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Medical Center, Washington, DC USA
| | - Jerel A Fields
- 3Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Italo Mocchetti
- 1Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC USA.,2Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Medical Center, Washington, DC USA
| |
Collapse
|
13
|
Rozzi SJ, Avdoshina V, Fields JA, Trejo M, Ton HT, Ahern GP, Mocchetti I. Human Immunodeficiency Virus Promotes Mitochondrial Toxicity. Neurotox Res 2017; 32:723-733. [PMID: 28695547 DOI: 10.1007/s12640-017-9776-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/15/2017] [Accepted: 06/28/2017] [Indexed: 01/09/2023]
Abstract
Combined antiretroviral therapies (cART) have had remarkable success in reducing morbidity and mortality among patients infected with human immunodeficiency virus (HIV). However, mild forms of HIV-associated neurocognitive disorders (HAND), characterized by loss of synapses, remain. cART may maintain an undetectable HIV RNA load but does not eliminate the expression of viral proteins such as trans-activator of transcription (Tat) and the envelope glycoprotein gp120 in the brain. These two viral proteins are known to promote synaptic simplifications by several mechanisms, including alteration of mitochondrial function and dynamics. In this review, we aim to outline the many targets and pathways used by viral proteins to alter mitochondria dynamics, which contribute to HIV-induced neurotoxicity. A better understanding of these pathways is crucial for the development of adjunct therapies for HAND.
Collapse
Affiliation(s)
- Summer J Rozzi
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Rd NW, Washington, DC, 20057, USA
| | - Valeria Avdoshina
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Rd NW, Washington, DC, 20057, USA
| | - Jerel A Fields
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Margarita Trejo
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Hoai T Ton
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Gerard P Ahern
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Italo Mocchetti
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Rd NW, Washington, DC, 20057, USA.
| |
Collapse
|
14
|
Wenzel ED, Bachis A, Avdoshina V, Taraballi F, Tasciotti E, Mocchetti I. Endocytic Trafficking of HIV gp120 is Mediated by Dynamin and Plays a Role in gp120 Neurotoxicity. J Neuroimmune Pharmacol 2017; 12:492-503. [PMID: 28349243 DOI: 10.1007/s11481-017-9739-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 03/13/2017] [Indexed: 12/13/2022]
Abstract
Neurons that endocytose the human immunodeficiency virus-1 (HIV) protein gp120 exhibit neurite retraction and activation of caspase-3, suggesting that the endocytic process may be crucial for gp120-mediated neuronal injury. The goal of this study is to demonstrate that internalization and accumulation of gp120 play a role in its neurotoxic effects. In mammalian cells, endocytosis is primarily a dynamin-dependent process. To establish whether gp120 is endocytosed in a dynamin-dependent manner, we used fibroblasts in which deletion of dynamins was induced by tamoxifen. We observed a robust reduction of intracellular gp120 immunoreactivity in tamoxifen-treated cells. To examine whether endocytosis of gp120 is crucial for its neurotoxic effect, we blocked gp120 internalization into primary rat cortical neurons by dynasore, an inhibitor of the dynamin GTP-ase activity. We found that dynasore blocks both gp120 internalization and neurotoxicity. We then utilized gp120-loaded mesoporous silica nanoparticles to deliver gp120 intracellularly. We established that once internalized, gp120 is neurotoxic regardless of chemokine receptor activation. Our data suggest that dynamin-dependent endocytosis of gp120 is critical for its neurotoxicity.
Collapse
Affiliation(s)
- Erin D Wenzel
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Medical Center, EP09 New Research Building, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA.,Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Alessia Bachis
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Medical Center, EP09 New Research Building, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Valeria Avdoshina
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Medical Center, EP09 New Research Building, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Francesca Taraballi
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, 77030, USA.,Department of Orthopedics, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Italo Mocchetti
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Medical Center, EP09 New Research Building, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA.
| |
Collapse
|
15
|
Hung KM, Chen PC, Hsieh HC, Calkins MJ. Mitochondrial defects arise from nucleoside/nucleotide reverse transcriptase inhibitors in neurons: Potential contribution to HIV-associated neurocognitive disorders. Biochim Biophys Acta Mol Basis Dis 2016; 1863:406-413. [PMID: 27840304 DOI: 10.1016/j.bbadis.2016.11.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/04/2016] [Accepted: 11/10/2016] [Indexed: 02/07/2023]
Abstract
The cornerstone of current HIV treatment is a class of drugs called nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs). However, patients who receive long term treatment with NRTIs often develop severe side effects, which are related to mitochondrial toxicity. The potential contribution of NRTI-mediated toxicity to HIV-associated neurocognitive disorders (HAND) has not been fully explored. NRTI toxicity is thought to be mediated through mitochondrial DNA polymerase γ (pol γ) inhibition, which impairs mitochondrial DNA (mtDNA) synthesis and leads to various mitochondrial dysfunctions. To evaluate the relationship between NRTI-mediated pol γ inhibition and mitochondrial toxicity in neurons, we systematically investigated mitochondrial regulation in NRTI-treated primary cortical neurons by measuring parameters related to mtDNA content, retrograde signaling responses and mitochondrial homeostasis. The effects of four different NRTIs with variable pol γ inhibitory activity and mitochondrial toxicity were assessed. The strong pol γ inhibitor, ddI, abolished mtDNA synthesis and greatly reduced mtDNA content. However, mtDNA transcription was not as severely affected, and no defects in oxidative phosphorylation were observed. Detrimental effects on mitochondrial respiration and motility were observed after AZT treatment in the absence of mtDNA depletion or inhibition of mtDNA synthesis. The results suggest that individual NRTIs, such as ddI and AZT, have the potential to cause mitochondrial toxicity in neurons. This mitochondrial toxicity would be expected to contribute to neurotoxicity in the central nervous system, and therefore, HAND etiology may be affected by NRTI treatment.
Collapse
Affiliation(s)
- Kui-Ming Hung
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan; Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Pei-Chun Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Han-Chieh Hsieh
- Department of Neurology, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Marcus J Calkins
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan.
| |
Collapse
|
16
|
Ivanov AV, Valuev-Elliston VT, Ivanova ON, Kochetkov SN, Starodubova ES, Bartosch B, Isaguliants MG. Oxidative Stress during HIV Infection: Mechanisms and Consequences. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8910396. [PMID: 27829986 PMCID: PMC5088339 DOI: 10.1155/2016/8910396] [Citation(s) in RCA: 227] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/18/2016] [Indexed: 12/15/2022]
Abstract
It is generally acknowledged that reactive oxygen species (ROS) play crucial roles in a variety of natural processes in cells. If increased to levels which cannot be neutralized by the defense mechanisms, they damage biological molecules, alter their functions, and also act as signaling molecules thus generating a spectrum of pathologies. In this review, we summarize current data on oxidative stress markers associated with human immunodeficiency virus type-1 (HIV-1) infection, analyze mechanisms by which this virus triggers massive ROS production, and describe the status of various defense mechanisms of the infected host cell. In addition, we have scrutinized scarce data on the effect of ROS on HIV-1 replication. Finally, we present current state of knowledge on the redox alterations as crucial factors of HIV-1 pathogenicity, such as neurotoxicity and dementia, exhaustion of CD4+/CD8+ T-cells, predisposition to lung infections, and certain side effects of the antiretroviral therapy, and compare them to the pathologies associated with the nitrosative stress.
Collapse
Affiliation(s)
- Alexander V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
| | - Vladimir T. Valuev-Elliston
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
| | - Olga N. Ivanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
| | - Sergey N. Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
| | - Elizaveta S. Starodubova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
- M. P. Chumakov Institute of Poliomyelitis and Viral Encephalitides, Moscow 142782, Russia
| | - Birke Bartosch
- Cancer Research Center Lyon, INSERM U1052 and CNRS 5286, Lyon University, 69003 Lyon, France
- DevWeCan Laboratories of Excellence Network (Labex), France
| | - Maria G. Isaguliants
- Riga Stradins University, Riga LV-1007, Latvia
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
- N. F. Gamaleya Research Center of Epidemiology and Microbiology, Moscow 123098, Russia
| |
Collapse
|
17
|
Iida T, Yi H, Liu S, Huang W, Kanda H, Lubarsky DA, Hao S. Spinal CPEB-mtROS-CBP signaling pathway contributes to perineural HIV gp120 with ddC-related neuropathic pain in rats. Exp Neurol 2016; 281:17-27. [PMID: 27090160 DOI: 10.1016/j.expneurol.2016.04.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 04/03/2016] [Accepted: 04/12/2016] [Indexed: 10/21/2022]
Abstract
Human immunodeficiency virus (HIV) patients treated with nucleoside reverse transcriptase inhibitors (NRTIs), have been known to develop neuropathic pain. While there has been a major shift away from some neurotoxic NRTIs in current antiretroviral therapy, a large number of HIV patients alive today have previously received them, and many have developed painful peripheral neuropathy. The exact mechanisms by which HIV with NRTIs contribute to the development of neuropathic pain are not known. Previous studies suggest that cytoplasmic polyadenylation element-binding protein (CPEB), reactive oxygen species (ROS), and cAMP-response element-binding protein (CREB)-binding protein (CBP), are involved in the neuroimmunological diseases including inflammatory/neuropathic pain. In this study, we investigated the role of CPEB, mitochondrial ROS (mtROS), or CBP in neuropathic pain induced by HIV envelope protein gp120 combined with antiretroviral drug. The application of recombinant gp120 into the sciatic nerve plus systemic ddC (one of NRTIs) induced mechanical allodynia. Knockdown of CPEB or CBP using intrathecal antisense oligodeoxynucleotide (AS-ODN) reduced mechanical allodynia. Intrathecal mitochondrial superoxide scavenger mito-tempol (Mito-T) increased mechanical withdrawal threshold. Knockdown of CPEB using intrathecal AS-ODN, reduced the up-regulated mitochondrial superoxide in the spinal dorsal horn in rats with gp120 combined with ddC. Intrathecal Mito-T lowered the increased expression of CBP in the spinal dorsal horn. Immunostaining studies showed that neuronal CPEB positive cells were co-localized with MitoSox positive profiles, and that MitoSox positive profiles were co-localized with neuronal CBP. Our studies suggest that neuronal CPEB-mtROS-CBP pathway in the spinal dorsal horn, plays an important role in the gp120/ddC-induced neuropathic pain in rats.
Collapse
Affiliation(s)
- Takafumi Iida
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Department of Anesthesiology, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - Hyun Yi
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Shue Liu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Wan Huang
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Hirotsugu Kanda
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Department of Anesthesiology, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - David A Lubarsky
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Shuanglin Hao
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States.
| |
Collapse
|
18
|
Avdoshina V, Fields JA, Castellano P, Dedoni S, Palchik G, Trejo M, Adame A, Rockenstein E, Eugenin E, Masliah E, Mocchetti I. The HIV Protein gp120 Alters Mitochondrial Dynamics in Neurons. Neurotox Res 2016; 29:583-593. [PMID: 26936603 DOI: 10.1007/s12640-016-9608-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 12/15/2015] [Indexed: 12/17/2022]
Abstract
Neurotoxicity of human immunodeficiency virus-1 (HIV) includes synaptic simplification and neuronal apoptosis. However, the mechanisms of HIV-associated neurotoxicity remain unclear, thus precluding an effective treatment of the neurological complications. The present study was undertaken to characterize novel mechanisms of HIV neurotoxicity that may explain how HIV subjects develop neuronal degeneration. Several neurodegenerative disorders are characterized by mitochondrial dysfunction; therefore, we hypothesized that HIV promotes mitochondrial damage. We first analyzed brains from HIV encephalitis (HIVE) by electron microscopy. Several sections of HIVE subjects contained enlarged and damaged mitochondria compared to brains from HIV subjects with no neurological complications. Similar pathologies were observed in mice overexpressing the HIV protein gp120, suggesting that this viral protein may be responsible for mitochondrial pathology found in HIVE. To gain more information about the cellular mechanisms of gp120 neurotoxicity, we exposed rat cortical neurons to gp120 and we determined cellular oxygen consumption rate, mitochondrial distribution, and trafficking. Our data show that gp120 evokes impairment in mitochondrial function and distribution. These data suggest that one of the mechanisms of HIV neurotoxicity includes altered mitochondrial dynamics in neurons.
Collapse
Affiliation(s)
- Valeria Avdoshina
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Jerel Adam Fields
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Paul Castellano
- Department of Microbiology and Molecular Genetics, Public Health Research Institute Center and at the International Center for Public Health New Jersey Medical School - Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Simona Dedoni
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Guillermo Palchik
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Margarita Trejo
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Anthony Adame
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Edward Rockenstein
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Eliseo Eugenin
- Department of Microbiology and Molecular Genetics, Public Health Research Institute Center and at the International Center for Public Health New Jersey Medical School - Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Eliezer Masliah
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.,Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Italo Mocchetti
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
19
|
Sanna MD, Ghelardini C, Galeotti N. Blockade of the spinal BDNF-activated JNK pathway prevents the development of antiretroviral-induced neuropathic pain. Neuropharmacology 2016; 105:543-552. [PMID: 26898292 DOI: 10.1016/j.neuropharm.2016.02.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/22/2016] [Accepted: 02/15/2016] [Indexed: 12/26/2022]
Abstract
UNLABELLED Although antiretroviral agents have been used successfully in suppressing viral production, they have also been associated with a number of side effects. The antiretroviral toxic neuropathy induces debilitating and extremely difficult to treat pain syndromes that often lead to discontinuation of antiretroviral therapy. Due to the critical need for the identification of novel therapeutic targets to improve antiretroviral neuropathic pain management, we investigated the role of the JNK signalling pathway in the mechanism of antiretroviral painful neuropathy. Mice were exposed to zalcitabine (2',3'-dideoxycytidine, ddC) and stavudine (2',3'-didehydro-3'-deoxythymidine, d4T) that induced a persistent mechanical allodynia and a transient cold allodynia. Treatment with the JNK blocker SP600125 before antiretroviral administration abolished mechanical hypersensitivity with no effect on thermal response. A robust spinal JNK overphosphorylation was observed on post-injection day 1 and 3, along with a JNK-dependent increase in p-c-Jun and ATF3 protein levels. Co-immunoprecipitation experiments showed the presence of a heterodimeric complex between ATF3 and c-Jun indicating that these transcription factors can act together to regulate transcription through heterodimerization. A rise in BDNF and caspase-3 protein levels was detected on day 1 and BDNF sequestration prevented both caspase-3 and p-JNK increase. These data suggest that BDNF plays a role in the early stages of ddC-induced allodynia by promoting apoptotic events and the activation of a hypernociceptive JNK-mediated pathway. We illustrated the activation of a BDNF-mediated JNK pathway involved in the early events responsible for the promotion of neuropathic pain, leading to a better knowledge of the mechanisms involved in the antiretroviral neuropathy. SUMMARY JNK blockade prevents antiretroviral-induced pain hypersensitivity. This may represent a potential prophylactic treatment of neuropathic pain to improve antiretroviral tolerability.
Collapse
Affiliation(s)
- Maria Domenica Sanna
- Laboratory of Neuropsychopharmacology, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Florence, Italy
| | - Carla Ghelardini
- Laboratory of Neuropsychopharmacology, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Florence, Italy
| | - Nicoletta Galeotti
- Laboratory of Neuropsychopharmacology, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Florence, Italy.
| |
Collapse
|
20
|
Neuropathological sequelae of Human Immunodeficiency Virus and apathy: A review of neuropsychological and neuroimaging studies. Neurosci Biobehav Rev 2015; 55:147-64. [PMID: 25944459 DOI: 10.1016/j.neubiorev.2015.04.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 04/22/2015] [Accepted: 04/23/2015] [Indexed: 02/07/2023]
Abstract
Apathy remains a common neuropsychiatric disturbance in the Human Immunodeficiency Virus (HIV-1) despite advances in anti-retroviral treatment (ART). The goal of the current review is to recapitulate findings relating apathy to the deleterious biobehavioral effects of HIV-1 in the post-ART era. Available literatures demonstrate that the emergence of apathy with other neurocognitive and neuropsychiatric symptoms may be attributed to neurotoxic effects of viral proliferation, e.g., aggregative effect of Tat and gp120 on apoptosis, transport and other enzymatic reactions amongst dopaminergic neurons and neuroglia. An assortment of neuroimaging modalities converge on the severity of apathy symptoms associated with the propensity of the virus to replicate within frontal-striatal brain circuits that facilitate emotional processing. Burgeoning research into functional brain connectivity also supports the effects of microvascular and neuro-inflammatory injury linked to aging with HIV-1 on the presentation of neuropsychiatric symptoms. Summarizing these findings, we review domains of HIV-associated neurocognitive and neuropsychiatric impairment linked to apathy in HIV. Taken together, these lines of research suggest that loss of affective, cognitive and behavioral inertia is commensurate with the neuropathology of HIV-1.
Collapse
|
21
|
Glover M, Hebert VY, Nichols K, Xue SY, Thibeaux TM, Zavecz JA, Dugas TR. Overexpression of mitochondrial antioxidant manganese superoxide dismutase (MnSOD) provides protection against AZT- or 3TC-induced endothelial dysfunction. Antiviral Res 2014; 111:136-42. [PMID: 25260898 PMCID: PMC4415438 DOI: 10.1016/j.antiviral.2014.09.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 09/15/2014] [Accepted: 09/16/2014] [Indexed: 01/14/2023]
Abstract
Nucleoside reverse transcriptase inhibitors (NRTIs) are considered the backbone of current combination therapies for HIV. These therapies have significantly decreased mortality and morbidity in HIV-infected patients, but some are associated with cardiovascular complications, including endothelial dysfunction, an early marker for atherosclerosis. Our prior studies demonstrated that co-treatment of cells with an antioxidant therapy reversed NRTI-induced endothelial injury. Thus, as a proof of concept that mitochondrially-targeted antioxidants may be useful in preventing NRTI toxicity, in the current study, mice overexpressing a mitochondrial antioxidant, manganese superoxide dismutase (MnSOD), were compared with wild-type (WT) mice. Mice were treated chronically with either zidovudine (AZT), lamivudine (3TC), or tenofovir (TDF) to determine whether overexpression of MnSOD protected them from endothelial dysfunction. Endothelial function was assessed using vessel reactivity experiments on thoracic aortas as well as measures of endothelium derived factors nitric oxide (NO), endothelin-1 (ET-1), and prostacyclin. Oxidative stress was evaluated as levels of plasma 8-isoprostane. Alterations in vessel reactivity, NO, and ET-1 in WT mice treated with AZT or 3TC were noted. Overexpression of MnSOD offered protection from decreases in vessel reactivity and increases in ET-1. These findings indicate that mitochondrial oxidative stress induced by AZT or 3TC plays a major role in mediating NRTI-induced endothelial dysfunction, and suggest that the use of targeted antioxidants administered in conjunction with NRTIs may attenuate these effects.
Collapse
Affiliation(s)
- Mitzi Glover
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, United States
| | - Valeria Y Hebert
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, United States
| | - Krystle Nichols
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, United States
| | - Stephen Y Xue
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, United States
| | - Taylor M Thibeaux
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, United States
| | - James A Zavecz
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, United States
| | - Tammy R Dugas
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, United States.
| |
Collapse
|
22
|
Toxic and drug-induced peripheral neuropathies: updates on causes, mechanisms and management. Curr Opin Neurol 2014; 26:481-8. [PMID: 23995278 DOI: 10.1097/wco.0b013e328364eb07] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW This review discusses publications highlighting current research on toxic, chemotherapy-induced peripheral neuropathies (CIPNs), and drug-induced peripheral neuropathies (DIPNs). RECENT FINDINGS The emphasis in clinical studies is on the early detection and grading of peripheral neuropathies, whereas recent studies in animal models have given insights into molecular mechanisms, with the discovery of novel neuronal, axonal, and Schwann cell targets. Some substances trigger inflammatory changes in the peripheral nerves. Pharmacogenetic techniques are underway to identify genes that may help to predict individuals at higher risk of developing DIPNs. Several papers have been published on chemoprotectants; however, to date, this approach has not been shown effective in clinical trials. SUMMARY Both length and nonlength-dependent neuropathies are encountered, including small-fiber involvement. The introduction of new diagnostic techniques, such as excitability studies, skin laser Doppler flowmetry, and pharmacogenetics, holds promise for early detection and to elucidate underlying mechanisms. New approaches to improve functions and quality of life in CIPN patients are discussed. Apart from developing less neurotoxic anticancer therapies, there is still hope to identify chemoprotective agents (erythropoietin and substances involved in the endocannabinoid system are promising) able to prevent or correct painful CIPNs.
Collapse
|
23
|
Douville RN, Nath A. Human endogenous retroviruses and the nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2014; 123:465-85. [PMID: 25015500 DOI: 10.1016/b978-0-444-53488-0.00022-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Renée N Douville
- Department of Microbiology, University of Winnipeg, Winnipeg, Manitoba, Canada
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
24
|
Robertson K, Liner J, Meeker RB. Antiretroviral neurotoxicity. J Neurovirol 2012; 18:388-99. [PMID: 22811264 DOI: 10.1007/s13365-012-0120-3] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 06/26/2012] [Accepted: 06/27/2012] [Indexed: 11/25/2022]
Abstract
Combination antiretroviral therapy (CART) has proven to effectively suppress systemic HIV burden, however, poor penetration into the central nervous system (CNS) provides incomplete protection. Although the severity of HIV-associated neurocognitive disorders (HAND) has been reduced, neurological disease is expected to exert an increasing burden as HIV-infected patients live longer. Strategies to enhance penetration of antiretroviral compounds into the CNS could help to control HIV replication in this reservoir but also carries an increased risk of neurotoxicity. Efforts to target antiretroviral compounds to the CNS will have to balance these risks against the potential gain. Unfortunately, little information is available on the actions of antiretroviral compounds in the CNS, particularly at concentrations that provide effective virus suppression. The current studies evaluated the direct effects of 15 antiretroviral compounds on neurons to begin to provide basic neurotoxicity data that will serve as a foundation for the development of dosing and drug selection guidelines. Using sensitive indices of neural damage, we found a wide range of toxicities, with median toxic concentrations ranging from 2 to 10,000 ng/ml. Some toxic concentrations overlapped concentrations currently seen in the CSF but the level of toxicity was generally modest at clinically relevant concentrations. Highest neurotoxicities were associated with abacavir, efavarenz, etravirine, nevaripine, and atazanavir, while the lowest were with darunavir, emtracitabine, tenofovir, and maraviroc. No additive effects were seen with combinations used clinically. These data provide initial evidence useful for the development of treatment strategies that might reduce the risk of antiretroviral neurotoxicity.
Collapse
Affiliation(s)
- Kevin Robertson
- Department of Neurology, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
25
|
Zhang Y, Wang M, Li H, Zhang H, Shi Y, Wei F, Liu D, Liu K, Chen D. Accumulation of nuclear and mitochondrial DNA damage in the frontal cortex cells of patients with HIV-associated neurocognitive disorders. Brain Res 2012; 1458:1-11. [PMID: 22554480 DOI: 10.1016/j.brainres.2012.04.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Revised: 03/28/2012] [Accepted: 04/02/2012] [Indexed: 12/31/2022]
Abstract
Oxidative stress has been suggested to play a key role in the neuropathogenesis of HIV infection. HIV proteins (gp120, Tat) and proinflammatory cytokines can trigger the production of reactive oxygen species (ROS), resulting in DNA and RNA lesions. Among all the lesions induced by ROS, one of the most abundant lesions in DNA and RNA is 8-hydroxydeoxyguanosine (8-oxoG). Here, we studied accumulated DNA oxidative damage induced by ROS in the central nervous system (CNS) in tissue from neuro-AIDS patients. The frontal cortex of autopsy tissue from HIV-1 infected patients was adopted for analysis for HIV-1 subtype, nuclear and mitochondrial DNA lesions by immunofluorescence staining, qPCR and sequencing of PCR cloning. This study provides evidence that HIV infection in the CNS leads to nuclear and mitochondrial genomic DNA damage in the brain. High level of nuclear and mtDNA 8-oxoG damage were identified in the cortex autopsy tissue of HAND patients. Increased accumulation of mtDNA mutations and depletion occurs in brain tissue in a subset of HAND cases, and is significantly different from that observed in control cases. These findings suggest that higher level of ROS in the CNS of HAND patients would contribute to the HIV induced neuro-inflammation and apoptosis of neuronal and glial cells.
Collapse
Affiliation(s)
- Yulin Zhang
- Department of Medicine, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Antioxidant sestrin-2 redistribution to neuronal soma in human immunodeficiency virus-associated neurocognitive disorders. J Neuroimmune Pharmacol 2012; 7:579-90. [PMID: 22450766 DOI: 10.1007/s11481-012-9357-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 03/13/2012] [Indexed: 10/28/2022]
Abstract
Sestrin-2 is involved in p53-dependent antioxidant defenses and in the maintenance of metabolic homeostasis. We hypothesize that sestrin-2 expression is altered in the brains of subjects diagnosed with human immunodeficiency virus (HIV)-associated neurocognitive disorders (HAND) due to neuronal oxidative stress. We studied sestrin-2 immunoreactivity in 42 isocortex sections from HIV-1-infected subjects compared to 18 age-matched non-HIV controls and 19 advanced Alzheimer's disease (AD) cases. With HIV infection, the sestrin-2 immunoreactivity pattern shifted from neuropil predominance (N) to neuropil and neuronal-soma co-dominance (NS) and neuronal-soma predominance (S; P < 0.0001, Chi-square test for linear trend). Among HIV cases showing the NS or S pattern, HAND cases were preferentially associated with the S pattern (n = 10 of 20) compared to cognitively intact cases (n = 1 of 11; P = 0.047, Fisher's exact test). In AD brains, sestrin-2 immunoreactivity was mostly intense in the neuropil and co-localized with phospho-Tau immunoreactivity in a subset of neurofibrillary lesions. Phospho-Tau-immunoreactive neurofibrillary lesions were rare in HIV cases and their occurrence was not associated with HAND. Levels of isocortical 8-hydroxy-deoxyguanosine (marker of nucleic acid oxidation) immunoreactivity were not significantly altered in HAND cases compared to cognitively intact HIV cases. In conclusion, the sestrin-2 immunoreactivity redistribution to neuronal soma in HAND suggests unique involvement of sestrin-2 in the pathophysiology of HAND, which is different from the role of sestrin-2 in AD pathogenesis. Alternatively, the difference in sestrin-2 immunoreactivity distribution between HAND and AD may be related to different degrees of severity or stages of oxidative stress.
Collapse
|
27
|
Leung GPH. Iatrogenic mitochondriopathies: a recent lesson from nucleoside/nucleotide reverse transcriptase inhibitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 942:347-69. [PMID: 22399431 DOI: 10.1007/978-94-007-2869-1_16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The use of nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) has revolutionized the treatment of infection by human immunodeficiency virus (HIV) and hepatitis-B virus. NRTIs can suppress viral replication in the long-term, but possess significant toxicity that can seriously compromise treatment effectiveness. The major toxicity of NRTIs is mitochondrial toxicity. This manifests as serious side effects such as myopathy, peripheral neuropathy and lactic acidosis. In general, it is believed that the mitochondrial pathogenesis is closely related to the effect of NRTIs on mitochondrial DNA polymerase-γ. Depletion and mutation of mitochondrial DNA during chronic NRTI therapy may lead to cellular respiratory dysfunction and release of reactive oxidative species, resulting in cellular damage. It is now apparent that the etiology is far more complex than originally thought. It appears to involve multiple mechanisms as well as host factors such as HIV per se, inborn mitochondrial mutation, and sex. Management of mitochondrial toxicity during NRTI therapy remains a challenge. Interruption of NRTI therapy and substitution of the causative agents with alternative better-tolerated NRTIs represents the mainstay of management for mitochondrial toxicity and its clinical manifestations. A range of pharmacological approaches has been proposed as treatments and prophylaxes.
Collapse
Affiliation(s)
- George P H Leung
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
28
|
Ammaranond P, Sanguansittianan S. Mechanism of HIV antiretroviral drugs progress toward drug resistance. Fundam Clin Pharmacol 2011; 26:146-61. [PMID: 22118474 DOI: 10.1111/j.1472-8206.2011.01009.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The rapid replication rate of HIV-1 RNA and its inherent genetic variation have led to the production of many HIV-1 variants with decreased drug susceptibility. The capacity of HIV to develop drug resistance mutations is a major obstacle to long-term effective anti-HIV therapy. Incomplete suppression of viral replication with an initial drug regimen diminishes the clinical benefit to the patient and may promote the development of broader drug resistance that may cause subsequent treatment regimens to be ineffective. The increased clinical use of combination antiretroviral treatment for HIV-1 infection has led to the selection of viral strains resistant to multiple drugs, including strains resistant to all licensed nucleoside analog RT inhibitors and protease inhibitors. Therefore, it is important to understand the influence of such mutations on viral properties such as replicative fitness, fidelity, and mutation rates. Although research continues to improve our understanding of resistance, leading to refined treatment strategies and, in some cases, improved outcome, resistance to antiretroviral therapy remains a major cause of treatment failure among patients living with HIV-1.
Collapse
Affiliation(s)
- Palanee Ammaranond
- Department of Transfusion Medicine, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand.
| | | |
Collapse
|
29
|
Adibhatla RM, Hatcher JF, Gusain A. Tricyclodecan-9-yl-xanthogenate (D609) mechanism of actions: a mini-review of literature. Neurochem Res 2011; 37:671-9. [PMID: 22101393 DOI: 10.1007/s11064-011-0659-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 10/14/2011] [Accepted: 11/10/2011] [Indexed: 12/21/2022]
Abstract
Tricyclodecan-9-yl-xanthogenate (D609) is known for its antiviral and antitumor properties. D609 actions are widely attributed to inhibiting phosphatidylcholine (PC)-specific phospholipase C (PC-PLC). D609 also inhibits sphingomyelin synthase (SMS). PC-PLC and/or SMS inhibition will affect lipid second messengers 1,2-diacylglycerol (DAG) and/or ceramide. Evidence indicates either PC-PLC and/or SMS inhibition affected the cell cycle and arrested proliferation, and stimulated differentiation in various in vitro and in vivo studies. Xanthogenate compounds are also potent antioxidants and D609 reduced Aß-induced toxicity, attributed to its antioxidant properties. Zn²⁺ is necessary for PC-PLC enzymatic activity; inhibition by D609 might be attributed to its Zn²⁺ chelation. D609 has also been proposed to inhibit acidic sphingomyelinase or down-regulate hypoxia inducible factor-1α; however these are down-stream events related to PC-PLC inhibition. Characterization of the mammalian PC-PLC is limited to inhibition of enzymatic activity (frequently measured using Amplex red assay with bacterial PC-PLC as a standard). The mammalian PC-PLC has not been cloned; sequenced and structural information is unavailable. D609 showed promise in cancer studies, reduced atherosclerotic plaques (inhibition of PC-PLC) and cerebral infarction after stroke (PC-PLC or SMS). D609 actions as an antagonist to pro-inflammatory cytokines have been attributed to PC-PLC. The purpose of this review is to comprehensively evaluate the literature and summarize the findings and relevance to cell cycle and CNS pathologies.
Collapse
Affiliation(s)
- Rao Muralikrishna Adibhatla
- Department of Neurological Surgery, Clinical Science Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI 53792-3232, USA.
| | | | | |
Collapse
|
30
|
Zheng X, Ouyang H, Liu S, Mata M, Fink DJ, Hao S. TNFα is involved in neuropathic pain induced by nucleoside reverse transcriptase inhibitor in rats. Brain Behav Immun 2011; 25:1668-76. [PMID: 21741472 PMCID: PMC3191308 DOI: 10.1016/j.bbi.2011.06.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 06/13/2011] [Accepted: 06/14/2011] [Indexed: 12/11/2022] Open
Abstract
In patients with HIV/AIDS, neuropathic pain is a common neurological complication. Infection with the HIV itself may lead to neuropathic pain, and painful symptoms are enhanced when patients are treated with nucleoside reverse transcriptase inhibitors (NRTIs). The mechanisms by which NRTIs contribute to the development of neuropathic pain are not known. In the current studies, we tested the role of TNFα in antiretroviral drug-induced neuropathic pain. We administered 2',3'-dideoxycytidine (ddC, one of the NRTIs) systemically to induce mechanical allodynia. We found that ddC induced overexpression of both mRNA and proteins of GFAP and TNFα in the spinal dorsal horn. TNFα was colocalized with GFAP in the spinal dorsal horn and with NeuN in the DRG. Knockdown of TNFα with siRNA blocked the mechanical allodynia induced by ddC. Intrathecal administration of glial inhibitor or recombinant TNF soluble receptor, reversed mechanical allodynia induced by ddC. These results suggest that TNFα is involved in NRTI-induced neuropathic pain.
Collapse
Affiliation(s)
- Xuexing Zheng
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL33136,College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, P. R. China
| | - Handong Ouyang
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109
| | - Shue Liu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL33136
| | - Marina Mata
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109
| | - David J. Fink
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109
| | - Shuanglin Hao
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL33136,Correspondence to: Dr. Shuanglin Hao, Associate Professor, Research, Department of Anesthesiology, University of Miami Miller School of Medicine, 1550 NW 10th Avenue, Fox BLDG, Rm304C, Miami, FL 33136, Tel: 1-305-243-6420, Fax: 1-305-243-9160,
| |
Collapse
|
31
|
Oxidative Stress and β-Amyloid Protein in Alzheimer’s Disease. Neuromolecular Med 2011; 13:223-50. [DOI: 10.1007/s12017-011-8155-9] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 08/26/2011] [Indexed: 12/21/2022]
|
32
|
Wu D, Niu JQ, Ding YH, Wu XY, Zhong BH, Feng XW. Antiviral effects of three novel derivatives of adefovir on the replication of hepatitis B virus. Med Chem Res 2011. [DOI: 10.1007/s00044-011-9616-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
33
|
Chen CH, Chou TW, Cheng LH, Ho CW. In vitro anti-adenoviral activity of five Allium plants. J Taiwan Inst Chem Eng 2011. [DOI: 10.1016/j.jtice.2010.07.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
34
|
Jayadev S, Garden GA. Host and viral factors influencing the pathogenesis of HIV-associated neurocognitive disorders. J Neuroimmune Pharmacol 2009; 4:175-89. [PMID: 19373562 DOI: 10.1007/s11481-009-9154-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Accepted: 03/27/2009] [Indexed: 01/03/2023]
Abstract
The human immunodeficiency virus (HIV) invades the central nervous system early in the course of infection and establishes a protected viral reservoir. However, neurocognitive consequences of HIV infection, known collectively as HIV-associated neurocognitive disorders (HAND), develop in only a small portion of infected patients. The precise mechanisms of pathogenesis involved in HIV-induced central nervous system injury are still not completely understood. In particular, most theories of HAND pathogenesis cannot account for either the selective vulnerability of specific neuronal populations to HIV-induced neurodegeneration or why only a subset of patients develop clinically detectable nervous system disease. Epidemiological and virological studies have identified a variety of host and viral factors that are associated with increased risk of developing HAND. Some host factors that predispose HIV-infected patients to HAND overlap with those associated with Alzheimer's disease (AD), suggesting the possibility that common pathogenic mechanisms may participate in both diseases. Here, we will review reports of host and viral factors associated with HAND and place these studies in the context of the data employed to support current theories regarding the molecular and cellular mechanisms that lead to HIV-induced neurodegeneration with additional focus on mechanisms common to AD pathogenesis.
Collapse
Affiliation(s)
- Suman Jayadev
- Department of Neurology and Center for Neurogenetics and Neurotherapeutics, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
35
|
Fraternale A, Paoletti MF, Casabianca A, Nencioni L, Garaci E, Palamara AT, Magnani M. GSH and analogs in antiviral therapy. Mol Aspects Med 2009; 30:99-110. [DOI: 10.1016/j.mam.2008.09.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Revised: 09/15/2008] [Accepted: 09/15/2008] [Indexed: 01/07/2023]
|
36
|
Nasi M, Guaraldi G, Orlando G, Durante C, Pinti M, Nemes E, Nardini G, Passarino G, Cocchi M, Esposito R, Mussini C, Cossarizza A. Mitochondrial DNA Haplogroups and Highly Active Antiretroviral Therapy–Related Lipodystrophy. Clin Infect Dis 2008; 47:962-8. [DOI: 10.1086/591706] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
37
|
NeuroAIDS: characteristics and diagnosis of the neurological complications of AIDS. Mol Diagn Ther 2008; 12:25-43. [PMID: 18288880 DOI: 10.1007/bf03256266] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The neurological complications of AIDS (NeuroAIDS) include neurocognitive impairment and HIV-associated dementia (HAD; also known as AIDS dementia and HIV encephalopathy). HAD is the most significant and devastating central nervous system (CNS) complications associated with HIV infection. Despite recent advances in our knowledge of the clinical features, pathogenesis, and neurobiological aspects of HAD, it remains a formidable scientific and therapeutic challenge. An understanding of the mechanisms of HIV neuroinvasion, CNS proliferation, and HAD pathogenesis provide a basis for the interpretation of the diagnostic features of HAD and its milder form, HIV-associated minor cognitive/motor disorder (MCMD). Current diagnostic strategies are associated with significant limitations, but it is hoped that the use of biomarkers may assist researchers and clinicians in predicting the onset of the disease process and in evaluating the effects of new therapies.
Collapse
|
38
|
Singh AK, Gupta S, Jiang Y. Oxidative stress and protein oxidation in the brain of water drinking and alcohol drinking rats administered the HIV envelope protein, gp120. J Neurochem 2007; 104:1478-93. [PMID: 18067547 DOI: 10.1111/j.1471-4159.2007.05094.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Possible roles of oxidative stress and protein oxidation on alcohol-induced augmentation of cerebral neuropathy in gp120 administered alcohol preferring rats drinking either pure water (W rats) or a free-choice ethanol and water (E rats) for 90 days. This study showed that peripherally administered gp120 accumulated into the brain, liver, and RBCs samples from water drinking - gp120 administered rats (Wg rats) and ethanol drinking - gp120 administered rats (Eg rats), although gp120 levels in samples from Eg rats were significantly greater than the levels in samples from Wg rats. The brain samples from ethanol drinking-saline administered (EC) and Wg rats exhibited comparable levels of free radicals that were significantly lower than the levels in Eg rats. Peroxiredoxin-I (PrxI) activity in the brain samples exhibited the following pattern: Wg >> >> WC >> EC > Eg. Total protein-carbonyl and carbonylated hippocampal cholinergic neurostimulating peptide precursor protein levels, but not N-acetylaspartate or N-acetyl aspartylglutamate or total protein-thiol levels, paralleled the free radical levels in the brain of all four groups. This suggests PrxI inhibition may be more sensitive indicator of oxidative stress than measuring free radicals or metabolites. As PrxI oxidation in WC, Wg, and EC rats was reversible, while PrxI oxidation in Eg rats was not, we suggest that alcohol drinking and gp120 together hyperoxidized and inactivated PrxI that suppressed free radical neutralization in the brain of Eg rats. In conclusion, chronic alcohol drinking, by carbonylating and hyperoxidizing free radical neutralization proteins, augmented the gp120-induced oxidative stress that may be associated with an increase in severity of the brain neuropathy.
Collapse
Affiliation(s)
- Ashok K Singh
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, Minnesota 55108, USA.
| | | | | |
Collapse
|
39
|
Chang L, Yakupov R, Nakama H, Stokes B, Ernst T. Antiretroviral treatment is associated with increased attentional load-dependent brain activation in HIV patients. J Neuroimmune Pharmacol 2007; 3:95-104. [PMID: 18247124 DOI: 10.1007/s11481-007-9092-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2007] [Accepted: 09/05/2007] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The purpose of this paper was to determine whether antiretroviral medications, especially the nucleoside analogue reverse transcriptase inhibitors, lead to altered brain activation due to their potential neurotoxic effects in patients with human immunodeficiency virus (HIV) infection. METHODS Forty-two right-handed men were enrolled in three groups: seronegative controls (SN, n = 18), HIV subjects treated with antiretroviral medications (HIV+ARV, n = 12), or not treated with antiretroviral medications (HIV+NARV, n = 12). Each subject performed a set of visual attention tasks with increasing difficulty or load (tracking two, three or four balls) during functional magnetic resonance imaging. RESULTS HIV subjects, both groups combined, showed greater load-dependent increases in brain activation in the right frontal regions compared to SN (p-corrected = 0.006). HIV+ARV additionally showed greater load-dependent increases in activation compared to SN in bilateral superior frontal regions (p-corrected = 0.032) and a lower percent accuracy on the performance of the most difficult task (tracking four balls). Region of interest analyses further demonstrated that SN showed load-dependent decreases (with repeated trials despite increasing difficulty), while HIV subjects showed load-dependent increases in activation with the more difficult tasks, especially those on ARVs. INTERPRETATION These findings suggest that chronic ARV treatments may lead to greater requirement of the attentional network reserve and hence less efficient usage of the network and less practice effects in these HIV patients. As the brain has a limited reserve capacity, exhausting the reserve capacity in HIV+ARV would lead to declined performance with more difficult tasks that require more attention.
Collapse
Affiliation(s)
- L Chang
- Department of Medicine, Division of Neurology, John A. Burns School of Medicine, University of Hawaii at Manoa, The Queen's Medical Center University Tower, 1356 Lusitana Street, Honolulu, HI 96813, USA.
| | | | | | | | | |
Collapse
|
40
|
Butterfield DA, Reed T, Newman SF, Sultana R. Roles of amyloid beta-peptide-associated oxidative stress and brain protein modifications in the pathogenesis of Alzheimer's disease and mild cognitive impairment. Free Radic Biol Med 2007; 43:658-77. [PMID: 17664130 PMCID: PMC2031860 DOI: 10.1016/j.freeradbiomed.2007.05.037] [Citation(s) in RCA: 427] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2007] [Revised: 05/20/2007] [Accepted: 05/25/2007] [Indexed: 12/17/2022]
Abstract
Oxidative stress has been implicated to play a crucial role in the pathogenesis of a number of diseases, including neurodegenerative disorders, cancer, and ischemia, just to name a few. Alzheimer disease (AD) is an age-related neurodegenerative disorder that is recognized as the most common form of dementia. AD is histopathologically characterized by the presence of extracellular amyloid plaques, intracellular neurofibrillary tangles, the presence of oligomers of amyloid beta-peptide (Abeta), and synapse loss. In this review we discuss the role of Abeta in the pathogenesis of AD and also the use of redox proteomics to identify oxidatively modified brain proteins in AD and mild cognitive impairment. In addition, redox proteomics studies in in vivo models of AD centered around human Abeta(1-42) are discussed.
Collapse
|