1
|
Vincent B, Shukla M. The Common Denominators of Parkinson's Disease Pathogenesis and Methamphetamine Abuse. Curr Neuropharmacol 2024; 22:2113-2156. [PMID: 37691228 PMCID: PMC11337683 DOI: 10.2174/1570159x21666230907151226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 09/12/2023] Open
Abstract
The pervasiveness and mortality associated with methamphetamine abuse have doubled during the past decade, suggesting a possible worldwide substance use crisis. Epitomizing the pathophysiology and toxicology of methamphetamine abuse proclaims severe signs and symptoms of neurotoxic and neurobehavioral manifestations in both humans and animals. Most importantly, chronic use of this drug enhances the probability of developing neurodegenerative diseases manifolds. Parkinson's disease is one such neurological disorder, which significantly and evidently not only shares a number of toxic pathogenic mechanisms induced by methamphetamine exposure but is also interlinked both structurally and genetically. Methamphetamine-induced neurodegeneration involves altered dopamine homeostasis that promotes the aggregation of α-synuclein protofibrils in the dopaminergic neurons and drives these neurons to make them more vulnerable to degeneration, as recognized in Parkinson's disease. Moreover, the pathologic mechanisms such as mitochondrial dysfunction, oxidative stress, neuroinflammation and decreased neurogenesis detected in methamphetamine abusers dramatically resemble to what is observed in Parkinson's disease cases. Therefore, the present review comprehensively cumulates a holistic illustration of various genetic and molecular mechanisms putting across the notion of how methamphetamine administration and intoxication might lead to Parkinson's disease-like pathology and Parkinsonism.
Collapse
Affiliation(s)
- Bruno Vincent
- Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Mayuri Shukla
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, 10210, Bangkok, Thailand
| |
Collapse
|
2
|
Miller EJ, Khoshbouei H. Immunity on ice: The impact of methamphetamine on peripheral immunity. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 99:217-250. [PMID: 38467482 DOI: 10.1016/bs.apha.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Methamphetamine (METH) regulation of the dopamine transporter (DAT) and central nervous system (CNS) dopamine transmission have been extensively studied. However, our understanding of how METH influences neuroimmune communication and innate and adaptive immunity is still developing. Recent studies have shed light on the bidirectional communication between the CNS and the peripheral immune system. They have established a link between CNS dopamine levels, dopamine neuronal activity, and peripheral immunity. Akin to dopamine neurons in the CNS, a majority of peripheral immune cells also express DAT, implying that in addition to their effect in the CNS, DAT ligands such as methamphetamine may have a role in modulating peripheral immunity. For example, by directly influencing DAT-expressing peripheral immune cells and thus peripheral immunity, METH can trigger a feed-forward cascade that impacts the bidirectional communication between the CNS and peripheral immune system. In this review, we aim to discuss the current understanding of how METH modulates both innate and adaptive immunity and identify areas where knowledge gaps exist. These gaps will then be considered in guiding future research directions.
Collapse
Affiliation(s)
- Emily J Miller
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States.
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States.
| |
Collapse
|
3
|
Chilunda V, Weiselberg J, Martinez-Meza S, Mhamilawa LE, Cheney L, Berman JW. Methamphetamine induces transcriptional changes in cultured HIV-infected mature monocytes that may contribute to HIV neuropathogenesis. Front Immunol 2022; 13:952183. [PMID: 36059515 PMCID: PMC9433802 DOI: 10.3389/fimmu.2022.952183] [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: 05/24/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
HIV-associated neurocognitive impairment (HIV-NCI) persists in 15-40% of people with HIV (PWH) despite effective antiretroviral therapy. HIV-NCI significantly impacts quality of life, and there is currently no effective treatment for it. The development of HIV-NCI is complex and is mediated, in part, by the entry of HIV-infected mature monocytes into the central nervous system (CNS). Once in the CNS, these cells release inflammatory mediators that lead to neuroinflammation, and subsequent neuronal damage. Infected monocytes may infect other CNS cells as well as differentiate into macrophages, thus contributing to viral reservoirs and chronic neuroinflammation. Substance use disorders in PWH, including the use of methamphetamine (meth), can exacerbate HIV neuropathogenesis. We characterized the effects of meth on the transcriptional profile of HIV-infected mature monocytes using RNA-sequencing. We found that meth mediated an upregulation of gene transcripts related to viral infection, cell adhesion, cytoskeletal arrangement, and extracellular matrix remodeling. We also identified downregulation of several gene transcripts involved in pathogen recognition, antigen presentation, and oxidative phosphorylation pathways. These transcriptomic changes suggest that meth increases the infiltration of mature monocytes that have a migratory phenotype into the CNS, contributing to dysregulated inflammatory responses and viral reservoir establishment and persistence, both of which contribute to neuronal damage. Overall, our results highlight potential molecules that may be targeted for therapy to limit the effects of meth on HIV neuropathogenesis.
Collapse
Affiliation(s)
- Vanessa Chilunda
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Jessica Weiselberg
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Samuel Martinez-Meza
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Lwidiko E. Mhamilawa
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Parasitology and Medical Entomology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Department of Women’s and Children’s Health, International Maternal and Child Health (IMCH), Uppsala University, Uppsala, Sweden
| | - Laura Cheney
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Medicine, Division of Infectious Diseases, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, United States
| | - Joan W. Berman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
4
|
Targeting Aβ and p-Tau Clearance in Methamphetamine-Induced Alzheimer’s Disease-Like Pathology: Roles of Syntaxin 17 in Autophagic Degradation in Primary Hippocampal Neurons. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3344569. [PMID: 35633882 PMCID: PMC9132709 DOI: 10.1155/2022/3344569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/25/2022] [Accepted: 04/27/2022] [Indexed: 11/28/2022]
Abstract
Methamphetamine (Meth), a central nervous system (CNS) stimulant with strong neurotoxicity, causes progressive cognitive impairment with characterized neurodegenerative changes. However, the mechanism underlying Meth-induced pathological changes remains poorly understood. In the current study, Meth elicited a striking accumulation of the pathological proteins hyperphosphorylated tau (p-tau) and amyloid beta (Aβ) in primary hippocampal neurons, while the activation of autophagy dramatically ameliorated the high levels of these pathological proteins. Interestingly, after the Meth treatment, Aβ was massively deposited in autophagosomes, which were remarkably trapped in early endosomes. Mechanistically, syntaxin 17 (Stx17), a key soluble n-ethylmaleimide-sensitive fusion protein (NSF) attachment protein receptor (SNARE) protein responsible for autophagosome and mature endosome/lysosome fusion, was significantly downregulated and hindered in combination with autophagosomes. Notably, adenovirus overexpression of Stx17 in primary neurons facilitated autophagosome-mature endosome/lysosome fusion, which dramatically reversed the Meth-induced increases in the levels of p-tau, Aβ, beta-secretase (Bace-1), and C-terminal fragments (CTFs). Immunofluorescence assays showed that Stx17 retarded the Meth-induced Aβ, p-tau, and Bace-1 accumulation in autophagosomes and facilitated the translocation of these pathological proteins to lysosomes, which indicated the importance of Stx17 via enhanced autophagosome-mature endosome/lysosome fusion. Therefore, the current study reveals a novel mechanism involving Meth-induced high levels of pathological proteins in neurons. Targeting Stx17 may provide a novel therapeutic strategy for Meth-induced neurodegenerative changes.
Collapse
|
5
|
Tao L, Hu M, Zhang X, Wang X, Zhang Y, Chen X, Tang J, Wang J. Methamphetamine-mediated dissemination of β-amyloid: Disturbances in endocytosis, transport and clearance of β-amyloid in microglial BV2 cells. Toxicol Appl Pharmacol 2022; 447:116090. [DOI: 10.1016/j.taap.2022.116090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/27/2022] [Accepted: 05/22/2022] [Indexed: 12/01/2022]
|
6
|
Grabowska K, Macur K, Zieschang S, Zaman L, Haverland N, Schissel A, Morsey B, Fox HS, Ciborowski P. HIV-1 and methamphetamine alter galectins -1, -3, and -9 in human monocyte-derived macrophages. J Neurovirol 2022; 28:99-112. [PMID: 35175539 PMCID: PMC9076712 DOI: 10.1007/s13365-021-01025-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/30/2021] [Accepted: 10/27/2021] [Indexed: 12/05/2022]
Abstract
Macrophages are key elements of the innate immune system. Their HIV-1 infection is a complex process that involves multiple interacting factors and various steps and is further altered by exposure of infected cells to methamphetamine (Meth), a common drug of abuse in people living with HIV. This is reflected by dynamic changes in the intracellular and secreted proteomes of these cells. Quantification of these changes poses a challenge for experimental design and associated analytics. In this study, we measured the effect of Meth on expression of intracellular and secreted galectins-1, -3, and -9 in HIV-1 infected human monocyte-derived macrophages (hMDM) using SWATH-MS, which was further followed by MRM targeted mass spectrometry validation. Cells were exposed to Meth either prior to or after infection. Our results are the first to perform comprehensive quantifications of galectins in primary hMDM cells during HIV-1 infection and Meth exposure a building foundation for future studies on the molecular mechanisms underlying cellular pathology of hMDM resulting from viral infection and a drug of abuse—Meth.
Collapse
Affiliation(s)
- Kinga Grabowska
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.,Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Katarzyna Macur
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.,Core Facility Laboratories, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Sarah Zieschang
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lubaba Zaman
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nicole Haverland
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andrew Schissel
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brenda Morsey
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard S Fox
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pawel Ciborowski
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
7
|
Miller DR, Bu M, Gopinath A, Martinez LR, Khoshbouei H. Methamphetamine Dysregulation of the Central Nervous System and Peripheral Immunity. J Pharmacol Exp Ther 2021; 379:372-385. [PMID: 34535563 PMCID: PMC9351721 DOI: 10.1124/jpet.121.000767] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/16/2021] [Indexed: 11/22/2022] Open
Abstract
Methamphetamine (METH) is a potent psychostimulant that increases extracellular monoamines, such as dopamine and norepinephrine, and affects multiple tissue and cell types in the central nervous system (CNS) and peripheral immune cells. The reinforcing properties of METH underlie its significant abuse potential and dysregulation of peripheral immunity and central nervous system functions. Together, the constellation of METH's effects on cellular targets and regulatory processes has led to immune suppression and neurodegeneration in METH addicts and animal models of METH exposure. Here we extensively review many of the cell types and mechanisms of METH-induced dysregulation of the central nervous and peripheral immune systems. SIGNIFICANCE STATEMENT: Emerging research has begun to show that methamphetamine regulates dopaminergic neuronal activity. In addition, METH affects non-neuronal brain cells, such as microglia and astrocytes, and immunological cells of the periphery. Concurrent disruption of bidirectional communication between dopaminergic neurons and glia in the CNS and peripheral immune cell dysregulation gives rise to a constellation of dysfunctional neuronal, cell, and tissue types. Therefore, understanding the pathophysiology of METH requires consideration of the multiple targets at the interface between basic and clinical neuroscience.
Collapse
Affiliation(s)
- Douglas R Miller
- Department of Neuroscience, College of Medicine (D.R.M., M.B., A.G., H.K.), and Department of Oral Biology, College of Dentistry (L.R.M.), University of Florida, Gainesville, Florida
| | - Mengfei Bu
- Department of Neuroscience, College of Medicine (D.R.M., M.B., A.G., H.K.), and Department of Oral Biology, College of Dentistry (L.R.M.), University of Florida, Gainesville, Florida
| | - Adithya Gopinath
- Department of Neuroscience, College of Medicine (D.R.M., M.B., A.G., H.K.), and Department of Oral Biology, College of Dentistry (L.R.M.), University of Florida, Gainesville, Florida
| | - Luis R Martinez
- Department of Neuroscience, College of Medicine (D.R.M., M.B., A.G., H.K.), and Department of Oral Biology, College of Dentistry (L.R.M.), University of Florida, Gainesville, Florida
| | - Habibeh Khoshbouei
- Department of Neuroscience, College of Medicine (D.R.M., M.B., A.G., H.K.), and Department of Oral Biology, College of Dentistry (L.R.M.), University of Florida, Gainesville, Florida
| |
Collapse
|
8
|
Mystery of methamphetamine-induced autophagosome accumulation in hippocampal neurons: loss of syntaxin 17 in defects of dynein-dynactin driving and autophagosome-late endosome/lysosome fusion. Arch Toxicol 2021; 95:3263-3284. [PMID: 34374793 DOI: 10.1007/s00204-021-03131-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 08/04/2021] [Indexed: 01/07/2023]
Abstract
Methamphetamine (METH), a psychoactive-stimulant facilitates massive accumulation of autophagosomes and causes autophagy-associated neuronal death. However, the underlying mechanisms involving METH-induced auto-phagosome accumulation remain poorly understood. In the current study, autophagic flux was tracked by mRFP-GFP-LC3 adenovirus, 900 μM METH treatment was found to significantly disrupt autophagic flux, which was further validated by remarkable increase of co-localized of LC3 and SQSTM1/p62, enhancement of LC3-II and SQSTM1/p62 protein levels, and massive autophagosome puncta aggregation. With the cycloheximide (CHX) treatment, METH treatment was displayed a significant inhibition of SQSTM1/p62 degradation. Therefore, the mRNAs associated with vesicle degradation were screened, and syntaxin 17 (Stx17) and dynein-dynactin mRNA levels significantly decreased, an effect was proved in protein level as well. Intriguingly, METH induced autophagosome accumulation and autophagic flux disturbance was incredibly retarded by overexpression of Stx17, which was validated by the restoration of the fusion autophagosome-late endosome/lysosome fusion. Moreover, Stx17 overexpression obviously impeded the METH-induced decrease of co-localization of the retrograded motor protein dynein/dynactin and autophagosome-late endosome, though the dynein/dynactin proteins were not involved in autophagosome-late endosome/lysosome fusion. Collectively, our findings unravel the mechanism of METH-induced autophagosome accumulation involving autophagosome-late endosome/lysosome fusion deficiency and that autophagy-enhancing mechanisms such as the overexpression of Stx17 may be therapeutic strategies for the treatment of METH-induced neuronal damage.
Collapse
|
9
|
Macur K, Zieschang S, Lei S, Morsey B, Jaquet S, Belshan M, Fox HS, Ciborowski P. SWATH-MS and MRM: Quantification of Ras-related proteins in HIV-1 infected and methamphetamine-exposed human monocyte-derived macrophages (hMDM). Proteomics 2021; 21:e2100005. [PMID: 34051048 PMCID: PMC9977323 DOI: 10.1002/pmic.202100005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/30/2022]
Abstract
HIV-1 infection of macrophages is a multistep and multifactorial process that has been shown to be enhanced by exposure to methamphetamine (Meth). In this study, we sought to identify the underlying mechanisms of this effect by quantifying the effect of Meth on the proteome of HIV-1-infected macrophages using sequential windowed acquisition of all theoretical fragment ion mass spectra (SWATH-MS) approach. The analyses identified several members of the Rab family of proteins as being dysregulated by Meth treatment, which was confirmed by bioinformatic analyses that indicated substantial alteration of vesicular transport pathways. Validation of the SWATH-MS was performed using an MRM based approach, which confirmed that Meth exposure affects expression of the Rab proteins. However, the pattern of expression changes were highly dynamic, and displayed high donor-to-donor variability. Surprisingly a similar phenomenon was observed for Actin. Our results demonstrate that Meth affects vesicular transport pathways, suggesting a possible molecular mechanism underlying its effect on HIV infection hMDM and a potential broader effect of Meth on cellular homeostasis.
Collapse
Affiliation(s)
- Katarzyna Macur
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE,Core Facility Laboratories, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Poland
| | - Sarah Zieschang
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Shulei Lei
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Brenda Morsey
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Spencer Jaquet
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Michael Belshan
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, NE
| | - Howard S. Fox
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Pawel Ciborowski
- Department of Pharmacology and Experimental Neuroscience, School of Medicine, University of Nebraska Medical Center, Omaha, NE,Corresponding author: Dr. Pawel Ciborowski, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985800 University of Nebraska Medical Center, Omaha, NE 68198-5800, phone +1 (402) 559-3733, fax +1 (402) 559-7495
| |
Collapse
|
10
|
Yang T, Zang S, Wang Y, Zhu Y, Jiang L, Chen X, Zhang X, Cheng J, Gao R, Xiao H, Wang J. Methamphetamine induced neuroinflammation in mouse brain and microglial cell line BV2: Roles of the TLR4/TRIF/Peli1 signaling axis. Toxicol Lett 2020; 333:150-158. [DOI: 10.1016/j.toxlet.2020.07.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/29/2020] [Accepted: 07/26/2020] [Indexed: 11/28/2022]
|
11
|
Chen L, Yu P, Zhang L, Zou Y, Zhang Y, Jiang L, Gao R, Xiao H, Qian Y, Wang J. Methamphetamine exposure induces neuropathic protein β-Amyloid expression. Toxicol In Vitro 2019; 54:304-309. [DOI: 10.1016/j.tiv.2018.10.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/17/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022]
|
12
|
Papageorgiou M, Raza A, Fraser S, Nurgali K, Apostolopoulos V. Methamphetamine and its immune-modulating effects. Maturitas 2018; 121:13-21. [PMID: 30704560 DOI: 10.1016/j.maturitas.2018.12.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/29/2018] [Accepted: 12/04/2018] [Indexed: 12/22/2022]
Abstract
The recreational use of methamphetamine (METH, or ice) is a global burden. It pervades and plagues contemporary society; it has been estimated that there are up to 35 million users worldwide. METH is a highly addictive psychotropic compound which acts on the central nervous system, and chronic use can induce psychotic behavior. METH has the capacity to modulate immune cells, giving the drug long-term effects which may manifest as neuropsychiatric disorders, and that increase susceptibility to communicable diseases, such as HIV. In addition, changes to the cytokine balance have been associated with compromise of the blood-brain barrier, resulting to alterations to brain plasticity, creating lasting neurotoxicity. Immune-related signaling pathways are key to further evaluating how METH impacts host immunity through these neurological and peripheral modifications. Combining this knowledge with current data on inflammatory responses will improve understanding of how the adaptive and innate immunity responds to METH, how this can activate premature-ageing processes and how METH exacerbates disturbances that lead to non-communicable age-related diseases, including cardiovascular disease, stroke, depression and dementia.
Collapse
Affiliation(s)
- Marco Papageorgiou
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Ali Raza
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Sarah Fraser
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia; Department of Medicine, The University of Melbourne, Regenerative Medicine and StemCells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia.
| | | |
Collapse
|
13
|
Zhu J, Zang S, Chen X, Jiang L, Gu A, Cheng J, Zhang L, Wang J, Xiao H. Involvement of the delayed rectifier outward potassium channel Kv2.1 in methamphetamine-induced neuronal apoptosis via the p38 mitogen-activated protein kinase signaling pathway. J Appl Toxicol 2018; 38:696-704. [PMID: 29297590 DOI: 10.1002/jat.3576] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/06/2017] [Accepted: 11/11/2017] [Indexed: 01/08/2023]
Abstract
Methamphetamine (Meth) is an illicit psychostimulant with high abuse potential and severe neurotoxicity. Recent studies have shown that dysfunctions in learning and memory induced by Meth may partially reveal the mechanisms of neuronal channelopathies. Kv2.1, the primary delayed rectifying potassium channel in neurons, is responsible for mediating apoptotic current surge. However, whether Kv2.1 is involved in Meth-mediated neural injury remains unknown. In the present study, the treatment of primary cultured hippocampal neurons with Meth indicated that Meth induced a time- and dose-dependent augmentation of Kv2.1 protein expression, accompanied by elevated cleaved-caspase 3 and declined bcl-2/bax ratio. The blockage of Kv2.1 with the inhibitor GxTx-1E or the knockdown of the channel noticeably abrogated the pro-apoptotic effects mediated by Meth, demonstrating the specific roles of Kv2.1 in Meth-mediated neural damage. Additionally, the p38 mitogen-activated protein kinase (MAPK) signaling was demonstrated to be involved in Meth-mediated Kv2.1 upregulation and in the subsequent pro-apoptotic effects, as treatment with a p38 MAPK inhibitor significantly attenuated Meth-mediated Kv2.1 upregulation and cell apoptosis. Of note, PRE-084, a sigma-1 receptor agonist, obviously attenuated Meth-induced upregulation of Kv2.1 expression, neural apoptosis and p38 MAPK activation. Taken together, these results reveal a novel mechanism involved in Meth-induced neural death with implications for therapeutic interventions for Meth users.
Collapse
Affiliation(s)
- Jingying Zhu
- Key Lab of Modern Toxicology (NJMU), Ministry of Education; Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, Jiangsu, 211166, China.,Wuxi Center for Disease Control and Prevention, 499 Jincheng Road, Liangxi District, Wuxi, Jiangsu, 214023, China
| | - Songsong Zang
- Key Lab of Modern Toxicology (NJMU), Ministry of Education; Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, Jiangsu, 211166, China
| | - Xufeng Chen
- Department of Emergency Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Lei Jiang
- Department of Emergency Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Aihua Gu
- Key Lab of Modern Toxicology (NJMU), Ministry of Education; Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, Jiangsu, 211166, China
| | - Jie Cheng
- Key Lab of Modern Toxicology (NJMU), Ministry of Education; Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, Jiangsu, 211166, China
| | - Li Zhang
- Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Jun Wang
- Key Lab of Modern Toxicology (NJMU), Ministry of Education; Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, Jiangsu, 211166, China
| | - Hang Xiao
- Key Lab of Modern Toxicology (NJMU), Ministry of Education; Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, Jiangsu, 211166, China
| |
Collapse
|
14
|
Höper T, Mussotter F, Haase A, Luch A, Tralau T. Application of proteomics in the elucidation of chemical-mediated allergic contact dermatitis. Toxicol Res (Camb) 2017; 6:595-610. [PMID: 30090528 PMCID: PMC6062186 DOI: 10.1039/c7tx00058h] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/07/2017] [Indexed: 12/23/2022] Open
Abstract
Allergic contact dermatitis (ACD) is a widespread hypersensitivity reaction of the skin. The cellular mechanisms underlying its development are complex and involve close interaction of different cell types of the immune system. It is this very complexity which has long prevented straightforward replacement of the corresponding regulatory in vivo tests. Recent efforts have already resulted in the development of several in vitro testing alternatives that address key steps of ACD. Yet identification of suitable biomarkers is still a subject of intense research. Search strategies for the latter encompass transcriptomics, proteomics as well as metabolomics approaches. The scope of this review shall be the application and use of proteomics in the context of ACD. This includes highlighting relevant aspects of the molecular and cellular mechanisms underlying ACD, the exploitation of these mechanisms for testing and biomarkers (e.g., in the context of the OECD's adverse outcome pathway initiative) as well as an outlook on emerging proteome targets, for example during the allergen-induced activation of dendritic cells (DCs).
Collapse
Affiliation(s)
- Tessa Höper
- German Federal Institute for Risk Assessment , Department of Chemical and Product Safety , Max-Dohrn-Strasse 8-10 , 10589 Berlin , Germany .
| | - Franz Mussotter
- German Federal Institute for Risk Assessment , Department of Chemical and Product Safety , Max-Dohrn-Strasse 8-10 , 10589 Berlin , Germany .
| | - Andrea Haase
- German Federal Institute for Risk Assessment , Department of Chemical and Product Safety , Max-Dohrn-Strasse 8-10 , 10589 Berlin , Germany .
| | - Andreas Luch
- German Federal Institute for Risk Assessment , Department of Chemical and Product Safety , Max-Dohrn-Strasse 8-10 , 10589 Berlin , Germany .
| | - Tewes Tralau
- German Federal Institute for Risk Assessment , Department of Chemical and Product Safety , Max-Dohrn-Strasse 8-10 , 10589 Berlin , Germany .
| |
Collapse
|
15
|
Aalinkeel R, Mangum CS, Abou-Jaoude E, Reynolds JL, Liu M, Sundquist K, Parikh NU, Chaves LD, Mammen MJ, Schwartz SA, Mahajan SD. Galectin-1 Reduces Neuroinflammation via Modulation of Nitric Oxide-Arginase Signaling in HIV-1 Transfected Microglia: a Gold Nanoparticle-Galectin-1 “Nanoplex” a Possible Neurotherapeutic? J Neuroimmune Pharmacol 2016; 12:133-151. [DOI: 10.1007/s11481-016-9723-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/06/2016] [Indexed: 10/20/2022]
|
16
|
Figueroa G, Parira T, Laverde A, Casteleiro G, El-Mabhouh A, Nair M, Agudelo M. Characterization of Human Monocyte-derived Dendritic Cells by Imaging Flow Cytometry: A Comparison between Two Monocyte Isolation Protocols. J Vis Exp 2016. [PMID: 27805582 DOI: 10.3791/54296] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Dendritic cells (DCs) are antigen presenting cells of the immune system that play a crucial role in lymphocyte responses, host defense mechanisms, and pathogenesis of inflammation. Isolation and study of DCs have been important in biological research because of their distinctive features. Although they are essential key mediators of the immune system, DCs are very rare in blood, accounting for approximately 0.1 - 1% of total blood mononuclear cells. Therefore, alternatives for isolation methods rely on the differentiation of DCs from monocytes isolated from peripheral blood mononuclear cells (PBMCs). The utilization of proper isolation techniques that combine simplicity, affordability, high purity, and high yield of cells is imperative to consider. In the current study, two distinct methods for the generation of DCs will be compared. Monocytes were selected by adherence or negatively enriched using magnetic separation procedure followed by differentiation into DCs with IL-4 and GM-CSF. Monocyte and MDDC viability, proliferation, and phenotype were assessed using viability dyes, MTT assay, and CD11c/ CD14 surface marker analysis by imaging flow cytometry. Although the magnetic separation method yielded a significant higher percentage of monocytes with higher proliferative capacity when compared to the adhesion method, the findings have demonstrated the ability of both techniques to simultaneously generate monocytes that are capable of proliferating and differentiating into viable CD11c+ MDDCs after seven days in culture. Both methods yielded > 70% CD11c+ MDDCs. Therefore, our results provide insights that contribute to the development of reliable methods for isolation and characterization of human DCs.
Collapse
Affiliation(s)
- Gloria Figueroa
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University
| | - Tiyash Parira
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University
| | - Alejandra Laverde
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University
| | - Gianna Casteleiro
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University
| | | | - Madhavan Nair
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University
| | - Marisela Agudelo
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University;
| |
Collapse
|
17
|
Chen X, Xing J, Jiang L, Qian W, Wang Y, Sun H, Wang Y, Xiao H, Wang J, Zhang J. Involvement of calcium/calmodulin-dependent protein kinase II in methamphetamine-induced neural damage. J Appl Toxicol 2016; 36:1460-7. [PMID: 26923100 DOI: 10.1002/jat.3301] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 01/04/2016] [Accepted: 01/05/2016] [Indexed: 11/11/2022]
Abstract
Methamphetamine (METH), an illicit drug, is widely abused in many parts of the world. Mounting evidence shows that METH exposure contributes to neurotoxicity, particularly for the monoaminergic neurons. However, to date, only a few studies have tried to unravel the mechanisms involved in METH-induced non-monoaminergic neural damage. Therefore, in the present study, we tried to explore the mechanisms for METH-induced neural damage in cortical neurons. Our results showed that METH significantly increased intracellular [Ca(2) (+) ]i in Ca(2) (+) -containing solution rather than Ca(2) (+) -free solution. Moreover, METH also upregulated calmodulin (CaM) expression and activated CaM-dependent protein kinase II (CaMKII). Significantly, METH-induced neural damage can be partially retarded by CaM antagonist W7 as well as CaMKII blocker KN93. In addition, L-type Ca(2) (+) channel was also proved to be involved in METH-induced cell damage, as nifedipine, the L-type Ca(2) (+) channel-specific inhibitor, markedly attenuated METH-induced neural damage. Collectively, our results suggest that Ca(2) (+) -CaM-CaMKII is involved in METH-mediated neurotoxicity, and it might suggest a potential target for the development of therapeutic strategies for METH abuse. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Xufeng Chen
- Department of Emergency Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Jingjing Xing
- Department of Emergency Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Lei Jiang
- Department of Emergency Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Wenyi Qian
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, Jiangsu, 211166, China
| | - Yixin Wang
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, Jiangsu, 211166, China
| | - Hao Sun
- Department of Emergency Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Yu Wang
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, Jiangsu, 211166, China
| | - Hang Xiao
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, Jiangsu, 211166, China
| | - Jun Wang
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, Jiangsu, 211166, China.
| | - Jinsong Zhang
- Department of Emergency Medicine, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
18
|
Mediouni S, Marcondes MCG, Miller C, McLaughlin JP, Valente ST. The cross-talk of HIV-1 Tat and methamphetamine in HIV-associated neurocognitive disorders. Front Microbiol 2015; 6:1164. [PMID: 26557111 PMCID: PMC4615951 DOI: 10.3389/fmicb.2015.01164] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/07/2015] [Indexed: 12/15/2022] Open
Abstract
Antiretroviral therapy has dramatically improved the lives of human immunodeficiency virus 1 (HIV-1) infected individuals. Nonetheless, HIV-associated neurocognitive disorders (HAND), which range from undetectable neurocognitive impairments to severe dementia, still affect approximately 50% of the infected population, hampering their quality of life. The persistence of HAND is promoted by several factors, including longer life expectancies, the residual levels of virus in the central nervous system (CNS) and the continued presence of HIV-1 regulatory proteins such as the transactivator of transcription (Tat) in the brain. Tat is a secreted viral protein that crosses the blood–brain barrier into the CNS, where it has the ability to directly act on neurons and non-neuronal cells alike. These actions result in the release of soluble factors involved in inflammation, oxidative stress and excitotoxicity, ultimately resulting in neuronal damage. The percentage of methamphetamine (MA) abusers is high among the HIV-1-positive population compared to the general population. On the other hand, MA abuse is correlated with increased viral replication, enhanced Tat-mediated neurotoxicity and neurocognitive impairments. Although several strategies have been investigated to reduce HAND and MA use, no clinically approved treatment is currently available. Here, we review the latest findings of the effects of Tat and MA in HAND and discuss a few promising potential therapeutic developments.
Collapse
Affiliation(s)
- Sonia Mediouni
- Department of Infectious Diseases, The Scripps Research Institute , Jupiter, FL, USA
| | | | - Courtney Miller
- Department of Metabolism and Aging, The Scripps Research Institute , Jupiter, FL, USA ; Department of Neuroscience, The Scripps Research Institute , Jupiter, FL, USA
| | - Jay P McLaughlin
- Department of Pharmacodynamics, University of Florida , Gainesville, FL, USA
| | - Susana T Valente
- Department of Infectious Diseases, The Scripps Research Institute , Jupiter, FL, USA
| |
Collapse
|
19
|
Zhang Y, Bottinelli D, Lisacek F, Luban J, Strambio-De-Castillia C, Varesio E, Hopfgartner G. Optimization of human dendritic cell sample preparation for mass spectrometry-based proteomic studies. Anal Biochem 2015; 484:40-50. [PMID: 25983236 PMCID: PMC4732721 DOI: 10.1016/j.ab.2015.05.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 05/04/2015] [Accepted: 05/06/2015] [Indexed: 12/11/2022]
Abstract
Dendritic cells (DCs) are specialized leukocytes that orchestrate the adaptive immune response. Mass spectrometry (MS)-based proteomic study of these cells presents technical challenges, especially when the DCs are human in origin due to the paucity of available biological material. Here, to maximize MS coverage of the global human DC proteome, different cell disruption methods, lysis conditions, protein precipitation, and protein pellet solubilization and denaturation methods were compared. Mechanical disruption of DC cell pellets under cryogenic conditions, coupled with the use of RIPA (radioimmunoprecipitation assay) buffer, was shown to be the method of choice based on total protein extraction and on the solubilization and identification of nuclear proteins. Precipitation by acetone was found to be more efficient than that by 10% trichloroacetic acid (TCA)/acetone, allowing in excess of 28% more protein identifications. Although being an effective strategy to eliminate the detergent residue, the acetone wash step caused a loss of protein identifications. However, this potential drawback was overcome by adding 1% sodium deoxycholate into the dissolution buffer, which enhanced both solubility of the precipitated proteins and digestion efficiency. This in turn resulted in 6 to 11% more distinct peptides and 14 to 19% more total proteins identified than using 0.5M triethylammonium bicarbonate alone, with the greatest increase (34%) for hydrophobic proteins.
Collapse
Affiliation(s)
- Ying Zhang
- Life Sciences Mass Spectrometry, School of Pharmaceutical Sciences, University of Geneva, CH-1211 Geneva 4, Switzerland
| | - Dario Bottinelli
- Life Sciences Mass Spectrometry, School of Pharmaceutical Sciences, University of Geneva, CH-1211 Geneva 4, Switzerland
| | - Frédérique Lisacek
- Proteome Informatics Group, SIB Swiss Institute of Bioinformatics, CH-1211 Geneva 4, Switzerland; Faculty of Sciences, University of Geneva, CH-1211 Geneva 4, Switzerland
| | - Jeremy Luban
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | - Emmanuel Varesio
- Life Sciences Mass Spectrometry, School of Pharmaceutical Sciences, University of Geneva, CH-1211 Geneva 4, Switzerland
| | - Gérard Hopfgartner
- Life Sciences Mass Spectrometry, School of Pharmaceutical Sciences, University of Geneva, CH-1211 Geneva 4, Switzerland.
| |
Collapse
|
20
|
Sriram U, Haldar B, Cenna JM, Gofman L, Potula R. Methamphetamine mediates immune dysregulation in a murine model of chronic viral infection. Front Microbiol 2015; 6:793. [PMID: 26322025 PMCID: PMC4531300 DOI: 10.3389/fmicb.2015.00793] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 07/21/2015] [Indexed: 02/03/2023] Open
Abstract
Methamphetamine (METH) is a highly addictive psychostimulant that not only affects the brain and cognitive functions but also greatly impacts the host immune system, rendering the body susceptible to infections and exacerbating the severity of disease. Although there is gathering evidence about METH abuse and increased incidence of HIV and other viral infections, not much is known about the effects on the immune system in a chronic viral infection setting. We have used the lymphocytic choriomeningitis virus (LCMV) chronic mouse model of viral infection in a chronic METH environment and demonstrate that METH significantly increases CD3 marker on splenocytes and programmed death-1 (PD-1) expression on T cells, a cell surface signaling molecule known to inhibit T cell function and cause exhaustion in a lymphoid organ. Many of these METH effects were more pronounced during early stage of infection, which are gradually attenuated during later stages of infection. An essential cytokine for T-lymphocyte homeostasis, Interleukin-2 (IL-2) in serum was prominently reduced in METH-exposed infected mice. In addition, the serum pro-inflammatory (TNF, IL12 p70, IL1β, IL-6, and KC-GRO) and Th2 (IL-2, IL-10, and IL-4) cytokine profiles were also altered in the presence of METH. Interestingly CXCR3, an inflammatory chemokine receptor, showed significant increase in the METH treated LCMV infected mice. Similarly, compared to only infected mice, epidermal growth factor receptor (EGFR) in METH exposed LCMV infected mice were up regulated. Collectively, our data suggest that METH alters systemic, peripheral immune responses and modulates key markers on T cells involved in pathogenesis of chronic viral infection.
Collapse
Affiliation(s)
- Uma Sriram
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine Philadelphia, PA, USA
| | - Bijayesh Haldar
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine Philadelphia, PA, USA
| | - Jonathan M Cenna
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine Philadelphia, PA, USA
| | - Larisa Gofman
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine Philadelphia, PA, USA
| | - Raghava Potula
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine Philadelphia, PA, USA ; Center for Substance Abuse Research, Temple University School of Medicine Philadelphia, PA, USA
| |
Collapse
|
21
|
Parikh NU, Aalinkeel R, Reynolds JL, Nair BB, Sykes DE, Mammen MJ, Schwartz SA, Mahajan SD. Galectin-1 suppresses methamphetamine induced neuroinflammation in human brain microvascular endothelial cells: Neuroprotective role in maintaining blood brain barrier integrity. Brain Res 2015; 1624:175-187. [PMID: 26236024 DOI: 10.1016/j.brainres.2015.07.033] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 05/19/2015] [Accepted: 07/07/2015] [Indexed: 11/18/2022]
Abstract
Methamphetamine (Meth) abuse can lead to the breakdown of the blood-brain barrier (BBB) integrity leading to compromised CNS function. The role of Galectins in the angiogenesis process in tumor-associated endothelial cells (EC) is well established; however no data are available on the expression of Galectins in normal human brain microvascular endothelial cells and their potential role in maintaining BBB integrity. We evaluated the basal gene/protein expression levels of Galectin-1, -3 and -9 in normal primary human brain microvascular endothelial cells (BMVEC) that constitute the BBB and examined whether Meth altered Galectin expression in these cells, and if Galectin-1 treatment impacted the integrity of an in-vitro BBB. Our results showed that BMVEC expressed significantly higher levels of Galectin-1 as compared to Galectin-3 and -9. Meth treatment increased Galectin-1 expression in BMVEC. Meth induced decrease in TJ proteins ZO-1, Claudin-3 and adhesion molecule ICAM-1 was reversed by Galectin-1. Our data suggests that Galectin-1 is involved in BBB remodeling and can increase levels of TJ proteins ZO-1 and Claudin-3 and adhesion molecule ICAM-1 which helps maintain BBB tightness thus playing a neuroprotective role. Galectin-1 is thus an important regulator of immune balance from neurodegeneration to neuroprotection, which makes it an important therapeutic agent/target in the treatment of drug addiction and other neurological conditions.
Collapse
Affiliation(s)
- Neil U Parikh
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York at Buffalo, Clinical Translational Research Center, 875 Ellicott Street, Buffalo, NY 14203, USA
| | - R Aalinkeel
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York at Buffalo, Clinical Translational Research Center, 875 Ellicott Street, Buffalo, NY 14203, USA
| | - J L Reynolds
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York at Buffalo, Clinical Translational Research Center, 875 Ellicott Street, Buffalo, NY 14203, USA
| | - B B Nair
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York at Buffalo, Clinical Translational Research Center, 875 Ellicott Street, Buffalo, NY 14203, USA
| | - D E Sykes
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York at Buffalo, Clinical Translational Research Center, 875 Ellicott Street, Buffalo, NY 14203, USA
| | - M J Mammen
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York at Buffalo, Clinical Translational Research Center, 875 Ellicott Street, Buffalo, NY 14203, USA
| | - S A Schwartz
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York at Buffalo, Clinical Translational Research Center, 875 Ellicott Street, Buffalo, NY 14203, USA
| | - S D Mahajan
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York at Buffalo, Clinical Translational Research Center, 875 Ellicott Street, Buffalo, NY 14203, USA.
| |
Collapse
|
22
|
Donnelly MR, Ciborowski P. Proteomics, biomarkers, and HIV-1: A current perspective. Proteomics Clin Appl 2015; 10:110-25. [PMID: 26033875 PMCID: PMC4666820 DOI: 10.1002/prca.201500002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 03/17/2015] [Accepted: 05/27/2015] [Indexed: 01/24/2023]
Abstract
Despite more than three decades of extensive research, HIV‐1 infection although well controlled with cART, remains incurable. Multifactorial complexity of the viral life‐cycle poses great challenges in understanding molecular mechanisms underlying this infection and the development of biomarkers, which we hope will lead us to its eradication. For a more in‐depth understanding of how the virus interacts with host target cells, T cells and macrophages, proteomic profiling techniques that offer strategies to investigate the proteome in its entirety were employed. Here, we review proteomic studies related to HIV‐1 infection and discuss perspectives and limitations of proteomic and systems biology approaches in future studies.
Collapse
Affiliation(s)
- Maire Rose Donnelly
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Pawel Ciborowski
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
23
|
Salamanca SA, Sorrentino EE, Nosanchuk JD, Martinez LR. Impact of methamphetamine on infection and immunity. Front Neurosci 2015; 8:445. [PMID: 25628526 PMCID: PMC4290678 DOI: 10.3389/fnins.2014.00445] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 12/17/2014] [Indexed: 12/21/2022] Open
Abstract
The prevalence of methamphetamine (METH) use is estimated at ~35 million people worldwide, with over 10 million users in the United States. METH use elicits a myriad of social consequences and the behavioral impact of the drug is well understood. However, new information has recently emerged detailing the devastating effects of METH on host immunity, increasing the acquisition of diverse pathogens and exacerbating the severity of disease. These outcomes manifest as modifications in protective physical and chemical defenses, pro-inflammatory responses, and the induction of oxidative stress pathways. Through these processes, significant neurotoxicities arise, and, as such, chronic abusers with these conditions are at a higher risk for heightened consequences. METH use also influences the adaptive immune response, permitting the unrestrained development of opportunistic diseases. In this review, we discuss recent literature addressing the impact of METH on infection and immunity, and identify areas ripe for future investigation.
Collapse
Affiliation(s)
- Sergio A Salamanca
- Department of Biomedical Sciences, Long Island University-Post Brookville, NY, USA
| | - Edra E Sorrentino
- Department of Biomedical Sciences, Long Island University-Post Brookville, NY, USA
| | - Joshua D Nosanchuk
- Microbiology and Immunology, Albert Einstein College of Medicine Bronx, NY, USA ; Medicine (Division of Infectious Diseases), Albert Einstein College of Medicine Bronx, NY, USA
| | - Luis R Martinez
- Department of Biomedical Sciences, NYIT College of Osteopathic Medicine, New York Institute of Technology Old Westbury, NY, USA
| |
Collapse
|
24
|
Kim HS, Kang D, Moon MH, Kim HJ. Identification of pancreatic cancer-associated tumor antigen from HSP-enriched tumor lysate-pulsed human dendritic cells. Yonsei Med J 2014; 55:1014-27. [PMID: 24954332 PMCID: PMC4075362 DOI: 10.3349/ymj.2014.55.4.1014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 10/23/2013] [Accepted: 11/04/2013] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Vaccine strategies utilizing dendritic cells (DCs) to elicit anti-tumor immunity are the subject of intense research. Although we have shown that DCs pulsed with heat-treated tumor lysate (HTL) induced more potent anti-tumor immunity than DCs pulsed with conventional tumor lysate (TL), the underlying molecular mechanism is unclear. In order to explore the molecular basis of this approach and to identify potential antigenic peptides from pancreatic cancer, we analyzed and compared the major histocompatibility complex (MHC) ligands derived from TL- and HTL-pulsed dendritic cells by mass spectrophotometry. MATERIALS AND METHODS Human monocyte-derived dendritic cells were pulsed with TL or HTL prior to maturation induction. To delineate differences of MHC-bound peptide repertoire eluted from DCs pulsed with TL or HTL, nanoflow liquid chromatography-electrospray ionization-tandem mass spectrometry (nLC-ESI-MS-MS) was employed. RESULTS HTL, but not TL, significantly induced DC function, assessed by phenotypic maturation, allostimulation capacity and IFN-γ secretion by stimulated allogeneic T cells. DCs pulsed with TL or HTL displayed pancreas or pancreatic cancer-related peptides in context of MHC class I and II molecules. Some of the identified peptides had not been previously reported as expressed in pancreatic cancer or cancer of other tissue types. CONCLUSION Our partial lists of MHC-associated peptides revealed the differences between peptide profiles eluted from HTL-and TL-loaded DCs, implying that induced heat shock proteins in HTL chaperone tumor-derived peptides enhanced their delivery to DCs and promoted cross-presentation by DC. These findings may aid in identifying novel tumor antigens or biomarkers and in designing future vaccination strategies.
Collapse
Affiliation(s)
- Han-Soo Kim
- Innovative Cell and Gene Therapy Center, International St. Mary's Hospital, Incheon, Korea
| | - Dukjin Kang
- Center for Bioanalysis, Division of Metrology for Quality of Life, Korea Research Institute of Standards and Science, Daejeon, Korea
| | | | - Hyung Jik Kim
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea.
| |
Collapse
|
25
|
Wang J, Qian W, Liu J, Zhao J, Yu P, Jiang L, Zhou J, Gao R, Xiao H. Effect of methamphetamine on the microglial damage: role of potassium channel Kv1.3. PLoS One 2014; 9:e88642. [PMID: 24533129 PMCID: PMC3922974 DOI: 10.1371/journal.pone.0088642] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 01/07/2014] [Indexed: 11/29/2022] Open
Abstract
Methamphetamine (Meth) abusing represents a major public health problem worldwide. Meth has long been known to induce neurotoxicity. However, the mechanism is still remained poorly understood. Growing evidences indicated that the voltage-gated potassium channels (Kv) were participated in neuronal damage and microglia function. With the whole cell patch clamp, we found that Meth significantly increased the outward K+ currents, therefore, we explored whether Kv1.3, one of the major K+ channels expressed in microglia, was involved in Meth-induced microglia damage. Our study showed that Meth significantly increased the cell viability in a dose dependent manner, while the Kv blocker, tetraethylamine (TEA), 4-Aminopyridine (4-AP) and Kv1.3 specific antagonist margatoxin (MgTx), prevented against the damage mediated by Meth. Interestingly, treatment of cells with Meth resulted in increasing expression of Kv1.3 rather than Kv1.5, at both mRNA and protein level, which is partially blocked by MgTx. Furthermore, Meth also stimulated a significant increased expression of IL-6 and TNF-α at protein level, which was significantly inhibited by MgTx. Taken together, these results demonstrated that Kv1.3 was involved in Meth-mediated microglial damage, providing the potential target for the development of therapeutic strategies for Meth abuse.
Collapse
Affiliation(s)
- Jun Wang
- Key Lab of Modern Toxicology, Ministry of Education. Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wenyi Qian
- Key Lab of Modern Toxicology, Ministry of Education. Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jingli Liu
- Key Lab of Modern Toxicology, Ministry of Education. Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Departments of Experimental Medicine, Nanjing Drum Tower Hospital, Nanjing Medical University, Nanjing, China
| | - Jingjing Zhao
- Key Lab of Modern Toxicology, Ministry of Education. Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Pan Yu
- Key Lab of Modern Toxicology, Ministry of Education. Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lei Jiang
- Department of Emergency Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Zhou
- Key Lab of Modern Toxicology, Ministry of Education. Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Rong Gao
- Key Lab of Modern Toxicology, Ministry of Education. Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hang Xiao
- Key Lab of Modern Toxicology, Ministry of Education. Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- * E-mail:
| |
Collapse
|
26
|
Wang X, Wang Y, Ye L, Li J, Zhou Y, Sakarcan S, Ho W. Modulation of intracellular restriction factors contributes to methamphetamine-mediated enhancement of acquired immune deficiency syndrome virus infection of macrophages. Curr HIV Res 2012; 10:407-14. [PMID: 22591364 DOI: 10.2174/157016212802138797] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 02/10/2012] [Accepted: 02/12/2012] [Indexed: 12/13/2022]
Abstract
Epidemiological studies have demonstrated that the use of methamphetamine (METH), a sympathomimetic stimulant, is particularly common among patients infected with HIV. In vitro studies have determined that METH enhances HIV infection of CD4+ T cells, monocyte-derived dendritic cells, and macrophages. In addition, animal studies have also showed that METH treatment increases brain viral load of SIV-infected monkeys and promotes HIV replication and viremia in HIV/hu-CycT1 transgenic mice. However, the mechanisms (s) of METH actions on HIV remain to be determined. In this study, we investigated the impact of METH on intracellular restriction factors against HIV and SIV. We demonstrated that METH treatment of human blood mononuclear phagocytes significantly affected the expression of anti-HIV microRNAs and several key elements (RIG-I, IRF-3/5, SOCS-2, 3 and PIAS-1, 3, X, Y) in the type I IFN pathway. The suppression of these innate restriction factors was associated with a reduced production of type I IFNs and the enhancement of HIV or SIV infection of macrophages. These findings indicate that METH use impairs intracellular innate antiviral mechanism(s) in macrophages, contributing to cell susceptibility to the acquired immune deficiency syndrome (AIDS) virus infection.
Collapse
Affiliation(s)
- Xu Wang
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Understanding changes in the expression of specific proteins and/or alterations in their posttranslational modifications is crucial to elucidating the molecular mechanisms underlying disease states such as alcoholic liver disease. Protein separation and analysis techniques such as two-dimensional electrophoresis and mass spectrometry can be used for identifying biomarker proteins that are altered during progression of alcoholic liver disease. In this chapter, we outline methods for resolving liver tissue proteins from a rodent model of alcoholic liver disease using two-dimensional electrophoresis and identifying differentially expressed proteins using mass spectrometry. In addition, since oxidative stress strongly correlates with alcoholic liver disease, we also describe methods for identifying oxidatively modified proteins from liver tissue. We specifically focus on identifying proteins that are carbonylated as protein carbonylation is a permanent modification and considered deleterious to cells. The combination of two-dimensional electrophoresis for protein resolution, mass spectrometry for protein identification, and affinity-based methods for enriching and identifying carbonylated proteins is a powerful methodology for protein biomarker identification.
Collapse
|
28
|
Reynolds JL, Law WC, Mahajan SD, Aalinkeel R, Nair B, Sykes DE, Yong KT, Hui R, Prasad PN, Schwartz SA. Nanoparticle based galectin-1 gene silencing, implications in methamphetamine regulation of HIV-1 infection in monocyte derived macrophages. J Neuroimmune Pharmacol 2012; 7:673-85. [PMID: 22689223 PMCID: PMC3419803 DOI: 10.1007/s11481-012-9379-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 06/03/2012] [Indexed: 12/22/2022]
Abstract
Galectin-1, an adhesion molecule, is expressed in macrophages and implicated in human immunodeficiency virus (HIV-1) viral adsorption. In this study, we investigated the effects of methamphetamine on galectin-1 production in human monocyte derived macrophages (MDM) and the role of galectin-1 in methamphetamine potentiation of HIV-1 infection. Herein we show that levels of galectin-1 gene and protein expression are significantly increased by methamphetamine. Furthermore, concomitant incubation of MDM with galectin-1 and methamphetamine facilitates HIV-1 infection compared to galectin-1 alone or methamphetamine alone. We utilized a nanotechnology approach that uses gold nanorod (GNR)-galectin-1 siRNA complexes (nanoplexes) to inhibit gene expression for galectin-1. Nanoplexes significantly silenced gene expression for galectin-1 and reversed the effects of methamphetamine on galectin-1 gene expression. Moreover, the effects of methamphetamine on HIV-1 infection were attenuated in the presence of the nanoplex in MDM.
Collapse
Affiliation(s)
- Jessica L Reynolds
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, State University of New York at Buffalo, Innovation Center, 640 Ellicott Street, Buffalo, NY 14203, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Serum proteomics of methamphetamine addicts and up-regulation of complement factor H related to methamphetamine addiction. Neurosci Lett 2012; 525:23-8. [DOI: 10.1016/j.neulet.2012.07.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 07/12/2012] [Accepted: 07/13/2012] [Indexed: 12/22/2022]
|
30
|
Reynolds JL, Law WC, Mahajan SD, Aalinkeel R, Nair B, Sykes DE, Mammen MJ, Yong KT, Hui R, Prasad PN, Schwartz SA. Morphine and galectin-1 modulate HIV-1 infection of human monocyte-derived macrophages. THE JOURNAL OF IMMUNOLOGY 2012; 188:3757-65. [PMID: 22430735 DOI: 10.4049/jimmunol.1102276] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Morphine is a widely abused, addictive drug that modulates immune function. Macrophages are a primary reservoir of HIV-1; therefore, they play a role in the development of this disease, as well as impact the overall course of disease progression. Galectin-1 is a member of a family of β-galactoside-binding lectins that are soluble adhesion molecules and that mediate direct cell-pathogen interactions during HIV-1 viral adhesion. Because the drug abuse epidemic and the HIV-1 epidemic are closely interrelated, we propose that increased expression of galectin-1 induced by morphine may modulate HIV-1 infection of human monocyte-derived macrophages (MDMs). In this article, we show that galectin-1 gene and protein expression are potentiated by incubation with morphine. Confirming previous studies, morphine alone or galectin-1 alone enhance HIV-1 infection of MDMs. Concomitant incubation with exogenous galectin-1 and morphine potentiated HIV-1 infection of MDMs. We used a nanotechnology approach that uses gold nanorod-galectin-1 small interfering RNA complexes (nanoplexes) to inhibit gene expression for galectin-1. We found that nanoplexes silenced gene expression for galectin-1, and they reversed the effects of morphine on galectin-1 expression. Furthermore, the effects of morphine on HIV-1 infection were reduced in the presence of the nanoplex.
Collapse
Affiliation(s)
- Jessica L Reynolds
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, State University of New York at Buffalo, Buffalo, NY 14203, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Methamphetamine toxicity and its implications during HIV-1 infection. J Neurovirol 2011; 17:401-15. [PMID: 21786077 DOI: 10.1007/s13365-011-0043-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 06/22/2011] [Indexed: 10/18/2022]
Abstract
Over the past two decades methamphetamine (MA) abuse has seen a dramatic increase. The abuse of MA is particularly high in groups that are at higher risk for HIV-1 infection, especially men who have sex with men (MSM). This review is focused on MA toxicity in the CNS as well as in the periphery. In the CNS, MA toxicity is comprised of numerous effects, including, but not limited to, oxidative stress produced by dysregulation of the dopaminergic system, hyperthermia, apoptosis, and neuroinflammation. Multiple lines of evidence demonstrate that these effects exacerbate the neurodegenerative damage caused by CNS infection of HIV perhaps because both MA and HIV target the frontostriatal regions of the brain. MA has also been demonstrated to increase viral load in the CNS of SIV-infected macaques. Using transgenic animal models, as well as cultured cells, the HIV proteins Tat and gp120 have been demonstrated to have neurotoxic properties that are aggravated by MA. In addition, MA has been shown to exhibit detrimental effects on the blood-brain barrier (BBB) that have the potential to increase the probability of CNS infection by HIV. Although the effects of MA in the periphery have not been as extensively studied as have the effects on the CNS, recent reports demonstrate the potential effects of MA on HIV infection in the periphery including increased expression of HIV co-receptors and increased expression of inflammatory cytokines.
Collapse
|
32
|
Wang X, Ye L, Zhou Y, Liu MQ, Zhou DJ, Ho WZ. Inhibition of anti-HIV microRNA expression: a mechanism for opioid-mediated enhancement of HIV infection of monocytes. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 178:41-7. [PMID: 21224041 DOI: 10.1016/j.ajpath.2010.11.042] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Revised: 08/24/2010] [Accepted: 09/09/2010] [Indexed: 11/16/2022]
Abstract
Several micro RNAs (miRNAs) have the ability to inhibit HIV replication in target cells. Thus, we investigated the impact of opioids (morphine and heroin), widely abused drugs among people infected with HIV, on the expression of cellular anti-HIV miRNAs in monocytes. We found that morphine-treated monocytes expressed lower levels of cellular anti-HIV miRNAs than untreated cells. In addition, morphine treatment of monocytes compromised type I interferon (IFN)-induced anti-HIV miRNA expression. These findings paralleled the observation that morphine treatment of monocytes enhanced HIV replication. These morphine-mediated actions on the anti-HIV miRNAs and HIV could be antagonized by the opioid receptor antagonists (naltrexone or Cys2, Tyr3, Arg5, Pen7-amide). Furthermore, the in vitro impact of morphine on miRNA expression was confirmed by the in vivo observation that heroin-dependent subjects had significantly lower levels of anti-HIV miRNAs (miRNA-28, 125b, 150, and 382) in peripheral blood mononuclear cells than the healthy subjects. These in vitro and in vivo findings indicate that opioid use impairs intracellular innate anti-HIV mechanism(s) in monocytes, contributing to cell susceptibility to HIV infection.
Collapse
Affiliation(s)
- Xu Wang
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | | | |
Collapse
|
33
|
Potula R, Hawkins BJ, Cenna JM, Fan S, Dykstra H, Ramirez SH, Morsey B, Brodie MR, Persidsky Y. Methamphetamine causes mitrochondrial oxidative damage in human T lymphocytes leading to functional impairment. THE JOURNAL OF IMMUNOLOGY 2010; 185:2867-76. [PMID: 20668216 DOI: 10.4049/jimmunol.0903691] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Methamphetamine (METH) abuse is known to be associated with an inordinate rate of infections. Although many studies have described the association of METH exposure and immunosuppression, so far the underlying mechanism still remains elusive. In this study, we present evidence that METH exposure resulted in mitochondrial oxidative damage and caused dysfunction of primary human T cells. METH treatment of T lymphocytes led to a rise in intracellular calcium levels that enhanced the generation of reactive oxygen species. TCR-CD28 linked calcium mobilization and subsequent uptake by mitochondria in METH-treated T cells correlated with an increase in mitochondrion-derived superoxide. Exposure to METH-induced mitochondrial dysfunction in the form of marked decrease in mitochondrial membrane potential, increased mitochondrial mass, enhanced protein nitrosylation and diminished protein levels of complexes I, III, and IV of the electron transport chain. These changes paralleled reduced IL-2 secretion and T cell proliferative responses after TCR-CD28 stimulation indicating impaired T cell function. Furthermore, antioxidants attenuated METH-induced mitochondrial damage by preserving the protein levels of mitochondrial complexes I, III, and IV. Altogether, our data indicate that METH can cause T cell dysfunction via induction of oxidative stress and mitochondrial injury as underlying mechanism of immune impairment secondary to METH abuse.
Collapse
Affiliation(s)
- Raghava Potula
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Reynolds JL, Mahajan SD, Aalinkeel R, Nair B, Sykes DE, Schwartz SA. Proteomic analyses of the effects of drugs of abuse on monocyte-derived mature dendritic cells. Immunol Invest 2010; 38:526-50. [PMID: 19811410 DOI: 10.1080/08820130902874110] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Drug abuse has become a global health concern. Understanding how drug abuse modulates the immune system and how the immune system responds to pathogens associated with drug abuse, such hepatitis C virus (HCV) and human immunodeficiency virus (HIV-1), can be assessed by an integrated approach comparing proteomic analyses and quantitation of gene expression. Two-dimensional (2D) difference gel electrophoresis was used to determine the molecular mechanisms underlying the proteomic changes that alter normal biological processes when monocyte-derived mature dendritic cells were treated with cocaine or methamphetamine. Both drugs differentially regulated the expression of several functional classes of proteins including those that modulate apoptosis, protein folding, protein kinase activity, and metabolism and proteins that function as intracellular signal transduction molecules. Proteomic data were validated using a combination of quantitative, real-time PCR and Western blot analyses. These studies will help to identify the molecular mechanisms, including the expression of several functionally important classes of proteins that have emerged as potential mediators of pathogenesis. These proteins may predispose immunocompetent cells, including dendritic cells, to infection with viruses such as HCV and HIV-1, which are associated with drug abuse.
Collapse
Affiliation(s)
- Jessica L Reynolds
- Departments of Medicine, Division of Allergy, Immunology and Rheumatology, State University of New York at Buffalo, Buffalo General Hospital, Buffalo, New York, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Ciborowski P. Biomarkers of HIV-1-associated neurocognitive disorders: challenges of proteomic approaches. Biomark Med 2009; 3:771-85. [PMID: 20477714 PMCID: PMC3544489 DOI: 10.2217/bmm.09.63] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
HIV-1 enters the brain shortly after infection, which may lead to neurological complications and in the most severe cases to encephalitis, dementia and death. The introduction of antiretroviral therapy reduced the incidence of the most severe conditions, nevertheless, approximately half of those infected with this virus will suffer to various degrees from HIV-1-associated neurocognitive disorders. Despite many years of research, there are no biomarkers that can objectively measure and, more importantly, predict the onset and the tempo of HIV-1-associated neurocognitive disorders. Here we review biomarker candidates of neurocognitive impairment due to HIV infection of the brain that have been proposed during the last two decades, and discuss perspectives and limitations of proteomic approaches in the search for new, more sensitive and specific biomarkers.
Collapse
Affiliation(s)
- Pawel Ciborowski
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA.
| |
Collapse
|
36
|
Ferreira GB, Mathieu C, Overbergh L. Understanding dendritic cell biology and its role in immunological disorders through proteomic profiling. Proteomics Clin Appl 2009; 4:190-203. [PMID: 21137043 DOI: 10.1002/prca.200900162] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 10/08/2009] [Accepted: 10/19/2009] [Indexed: 01/10/2023]
Abstract
Dendritic cells (DC) have always been present on the bright spot of immune research. They have been extensively studied for the last 35 years, and much is known about their different phenotypes, stimulatory capacity, and role in the immune system. During the last 15 years, great attention has been given to studies on global gene and protein expression profiles during the differentiation and maturation processes of these cells. It is well understood that studying the proteome, together with information on the role of protein post-translational modifications (PTM), will reveal the real dynamics of a living cell. The rapid increase of proteomic studies during the last decade describing the differentiation and maturation process in DCs, as well as modifications brought by the use of different compounds that either increase or decrease their immunogenicity, reflects the importance of understanding the molecular processes behind the functional properties of these cells. In the present review, we will give an overview of proteomic studies focusing on DCs. Thereby we will concentrate on the importance of these studies in understanding DC behavior from a molecular point of view and how these findings have aided in understanding the differences in functional properties of these cells.
Collapse
|
37
|
Reynolds JL, Mahajan SD, Aalinkeel R, Nair B, Sykes DE, Agosto-Mujica A, Hsiao CB, Schwartz SA. Modulation of the proteome of peripheral blood mononuclear cells from HIV-1-infected patients by drugs of abuse. J Clin Immunol 2009; 29:646-56. [PMID: 19543960 DOI: 10.1007/s10875-009-9309-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Accepted: 05/28/2009] [Indexed: 01/13/2023]
Abstract
INTRODUCTION We used proteomic analyses to assess how drug abuse modulates immunologic responses to infections with the human immunodeficiency virus type 1 (HIV-1). METHODS Two-dimensional difference gel electrophoresis was utilized to determine changes in the proteome of peripheral blood mononuclear cells (PBMC) isolated from HIV-1-positive donors that occurred after treatment with cocaine or methamphetamine. Both drugs differentially regulated the expression of several functional classes of proteins. We further isolated specific subpopulations of PBMC to determine which subpopulations were selectively affected by treatment with drugs of abuse. Monocytes, B cells, and T cells were positively or negatively selected from PBMC isolated from HIV-1-positive donors. RESULTS Our results demonstrate that cocaine and methamphetamine modulate gene expression primarily in monocytes and T cells, the primary targets of HIV-1 infection. Proteomic data were validated with quantitative, real-time polymerase chain reaction. These studies elucidate the molecular mechanisms underlying the effects of drugs of abuse on HIV-1 infections. Several functionally relevant classes of proteins were identified as potential mediators of HIV-1 pathogenesis and disease progression associated with drug abuse.
Collapse
Affiliation(s)
- Jessica L Reynolds
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Buffalo General Hospital, University at Buffalo, State University of New York at Buffalo, 311 MultiLab Research Building, Buffalo, NY,14203, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Liang H, Wang X, Chen H, Song L, Ye L, Wang SH, Wang YJ, Zhou L, Ho WZ. Methamphetamine enhances HIV infection of macrophages. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:1617-24. [PMID: 18458095 DOI: 10.2353/ajpath.2008.070971] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Epidemiological studies have demonstrated that the use of methamphetamine (meth), a sympathomimetic stimulant, is particularly common among patients infected with HIV. However, there is a lack of direct evidence that meth promotes HIV infection of target cells. This study examined whether meth is able to enhance HIV infection of macrophages, the primary target site for the virus. Meth treatment resulted in a significant and dose-dependent increase of HIV reverse transcriptase activity in human blood monocyte-derived macrophages. Dopamine D1 receptor antagonists (SCH23390 and SKF83566) blocked this meth-mediated increase in the HIV infectivity of macrophages. Investigation of the underlying mechanisms of meth action showed that meth up-regulated the expression of the HIV entry co-receptor CCR5 on macrophages. Additionally, meth inhibited the expression of endogenous interferon-alpha and signal transducer and activator of transcription-1 in macrophages. These findings provide direct in vitro evidence to support the possibility that meth may function as a cofactor in the immunopathogenesis of HIV infection and may lead to the future development of innate immunity-based intervention for meth users with HIV infection.
Collapse
Affiliation(s)
- Hao Liang
- Division of Allergy and Immunology, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Shen Y, Senzer NN, Nemunaitis JJ. Use of Proteomics Analysis for Molecular Precision Approaches in Cancer Therapy. Drug Target Insights 2008. [DOI: 10.4137/dti.s649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
| | - Neil N. Senzer
- LEAD Therapeutics, Inc., San Bruno, CA
- Mary Crowley Cancer Research Centers, Dallas, TX
| | | |
Collapse
|
40
|
Mercier S, St-Pierre C, Pelletier I, Ouellet M, Tremblay MJ, Sato S. Galectin-1 promotes HIV-1 infectivity in macrophages through stabilization of viral adsorption. Virology 2007; 371:121-9. [PMID: 18028978 DOI: 10.1016/j.virol.2007.09.034] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2007] [Revised: 07/23/2007] [Accepted: 09/22/2007] [Indexed: 11/17/2022]
Abstract
Following primary infection with human immunodeficiency virus type-1 (HIV-1), macrophages are thought to play an important role, as they are one of the first target cells the virus encounters and can also sustain a significant production of viruses over extended periods of time. While the interaction between the primary cellular receptor CD4 and the virus-encoded external envelope glycoprotein gp120 initiates the infection process, it has been suggested that various host factors are exploited by HIV-1 to facilitate adsorption onto the cell surface. Macrophages and other cells found at the infection site can secrete a soluble mammalian lectin, galectin-1, which binds to beta-galactoside residues through its carbohydrate recognition domain. Being a dimer, galectin-1 can cross-link ligands expressed on different constituents to mediate adhesion between cells or between cells and pathogens. We report here that galectin-1, but not galectin-3, increased HIV-1 infectivity in monocyte-derived macrophages (MDMs). This phenomenon was likely due to an enhancement of virus adsorption kinetics, which facilitates HIV-1 entry. The fusion inhibitors T-20 and TAK779 remained effective at reducing infection even in the presence of galectin-1, indicating that the galectin-1-mediated effect is occurring at a step prior to fusion. Together, our data suggest that galectin-1 can facilitate HIV-1 infection in MDMs by promoting early events of the virus replicative cycle (i.e. adsorption).
Collapse
Affiliation(s)
- Simon Mercier
- Research Center for Infectious Diseases, CHUL Research Center, Quebec, Canada
| | | | | | | | | | | |
Collapse
|