1
|
Vilca SJ, Margetts AV, Höglund L, Fleites I, Bystrom LL, Pollock TA, Bourgain-Guglielmetti F, Wahlestedt C, Tuesta LM. Microglia contribute to methamphetamine reinforcement and reflect persistent transcriptional and morphological adaptations to the drug. Brain Behav Immun 2024; 120:339-351. [PMID: 38838836 PMCID: PMC11269013 DOI: 10.1016/j.bbi.2024.05.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
Methamphetamine use disorder (MUD) is a chronic, relapsing disease that is characterized by repeated drug use despite negative consequences and for which there are currently no FDA-approved cessation therapeutics. Repeated methamphetamine (METH) use induces long-term gene expression changes in brain regions associated with reward processing and drug-seeking behavior, and recent evidence suggests that methamphetamine-induced neuroinflammation may also shape behavioral and molecular responses to the drug. Microglia, the resident immune cells in the brain, are principal drivers of neuroinflammatory responses and contribute to the pathophysiology of substance use disorders. Here, we investigated transcriptional and morphological changes in dorsal striatal microglia in response to methamphetamine-taking and during methamphetamine abstinence, as well as their functional contribution to drug-taking behavior. We show that methamphetamine self-administration induces transcriptional changes associated with protein folding, mRNA processing, immune signaling, and neurotransmission in dorsal striatal microglia. Importantly, many of these transcriptional changes persist through abstinence, a finding supported by morphological analyses. Functionally, we report that microglial ablation increases methamphetamine-taking, possibly involving neuroimmune and neurotransmitter regulation. In contrast, microglial depletion during abstinence does not alter methamphetamine-seeking. Taken together, these results suggest that methamphetamine induces both short and long-term changes in dorsal striatal microglia that contribute to altered drug-taking behavior and may provide valuable insights into the pathophysiology of MUD.
Collapse
Affiliation(s)
- Samara J Vilca
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Alexander V Margetts
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Leon Höglund
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Isabella Fleites
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Lauren L Bystrom
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Tate A Pollock
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Florence Bourgain-Guglielmetti
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Claes Wahlestedt
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Luis M Tuesta
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States.
| |
Collapse
|
2
|
Vilca SJ, Margetts AV, Fleites I, Wahlestedt C, Tuesta LM. Microglia contribute to methamphetamine reinforcement and reflect persistent transcriptional and morphological adaptations to the drug. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.19.563168. [PMID: 37961443 PMCID: PMC10634674 DOI: 10.1101/2023.10.19.563168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Methamphetamine use disorder (MUD) is a chronic, relapsing disease that is characterized by repeated drug use despite negative consequences and for which there are currently no FDA-approved cessation therapeutics. Repeated methamphetamine (METH) use induces long-term gene expression changes in brain regions associated with reward processing and drug-seeking behavior, and recent evidence suggests that methamphetamine-induced neuroinflammation may also shape behavioral and molecular responses to the drug. Microglia, the resident immune cells in the brain, are principal drivers of neuroinflammatory responses and contribute to the pathophysiology of substance use disorders. Here, we investigated transcriptional and morphological changes in dorsal striatal microglia in response to methamphetamine-taking and during methamphetamine abstinence, as well as their functional contribution to drug-taking behavior. We show that methamphetamine self-administration induces transcriptional changes associated with protein folding, mRNA processing, immune signaling, and neurotransmission in dorsal striatal microglia. Importantly, many of these transcriptional changes persist through abstinence, a finding supported by morphological analyses. Functionally, we report that microglial ablation increases methamphetamine-taking, possibly involving neuroimmune and neurotransmitter regulation, and that post-methamphetamine microglial repopulation attenuates drug-seeking following a 21-day period of abstinence. In contrast, microglial depletion during abstinence did not alter methamphetamine-seeking. Taken together, these results suggest that methamphetamine induces both short and long-term changes in dorsal striatal microglia that contribute to altered drug-taking behavior and may provide valuable insights into the pathophysiology of MUD.
Collapse
Affiliation(s)
- Samara J. Vilca
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Alexander V. Margetts
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Isabella Fleites
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Claes Wahlestedt
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Luis M. Tuesta
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136
| |
Collapse
|
3
|
Ozkizilcik A, Sharma A, Feng L, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Wiklund L, Sharma HS. Nanowired delivery of antibodies to tau and neuronal nitric oxide synthase together with cerebrolysin attenuates traumatic brain injury induced exacerbation of brain pathology in Parkinson's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 171:83-121. [PMID: 37783564 DOI: 10.1016/bs.irn.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Concussive head injury (CHI) is one of the major risk factors for developing Parkinson's disease in later life of military personnel affecting lifetime functional and cognitive disturbances. Till date no suitable therapies are available to attenuate CHI or PD induced brain pathology. Thus, further exploration of novel therapeutic agents are highly warranted using nanomedicine in enhancing the quality of life of veterans or service members of US military. Since PD or CHI induces oxidative stress and perturbs neurotrophic factors regulation associated with phosphorylated tau (p-tau) deposition, a possibility exists that nanodelivery of agents that could enhance neurotrophic factors balance and attenuate oxidative stress could be neuroprotective in nature. In this review, nanowired delivery of cerebrolysin-a balanced composition of several neurotrophic factors and active peptide fragments together with monoclonal antibodies to neuronal nitric oxide synthase (nNOS) with p-tau antibodies was examined in PD following CHI in model experiments. Our results suggest that combined administration of nanowired antibodies to nNOS and p-tau together with cerebrolysin significantly attenuated CHI induced exacerbation of PD brain pathology. This combined treatment also has beneficial effects in CHI or PD alone, not reported earlier.
Collapse
Affiliation(s)
- Asya Ozkizilcik
- Dept. Biomedical Engineering, University of Arkansas, Fayetteville, AR, United Staes
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan Road (West), Shijiazhuang, Hebei Province, P.R. China
| | - Dafin F Muresanu
- Dept. Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; ''RoNeuro'' Institute for Neurological Research and Diagnostic, Mircea Eliade Street, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Dept. Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Dept. Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ala Nozari
- Department of Anesthesiology, Boston University, Albany str, Boston MA, United States
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
4
|
Clark CE, Rigby BR. Can exposure to heat attenuate neurodegeneration in older adults with Parkinson's disease? Front Aging Neurosci 2023; 15:1239656. [PMID: 37744389 PMCID: PMC10513428 DOI: 10.3389/fnagi.2023.1239656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/25/2023] [Indexed: 09/26/2023] Open
Affiliation(s)
| | - Brandon Rhett Rigby
- School of Health Promotion and Kinesiology, Texas Woman's University, Denton, TX, United States
| |
Collapse
|
5
|
Nozari A, Sharma A, Wang Z, Feng L, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Wiklund L, Sharma HS. Co-administration of Nanowired Oxiracetam and Neprilysin with Monoclonal Antibodies to Amyloid Beta Peptide and p-Tau Thwarted Exacerbation of Brain Pathology in Concussive Head Injury at Hot Environment. ADVANCES IN NEUROBIOLOGY 2023; 32:271-313. [PMID: 37480464 DOI: 10.1007/978-3-031-32997-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Environmental temperature adversely affects the outcome of concussive head injury (CHI)-induced brain pathology. Studies from our laboratory showed that animals reared at either cold environment or at hot environment exacerbate brain pathology following CHI. Our previous experiments showed that nanowired delivery of oxiracetam significantly attenuated CHI-induced brain pathology and associated neurovascular changes. Military personnel are the most susceptible to CHI caused by explosion, blasts, missile or blunt head trauma leading to lifetime functional and cognitive impairments affecting the quality of life. Severe CHI leads to instant death and/or lifetime paralysis. Military personnel engaged in combat operations are often subjected to extreme high or low environmental temperature zones across the globe. Thus, further exploration of novel therapeutic agents at cold or hot ambient temperatures following CHI are the need of the hour. CHI is also a major risk factor for developing Alzheimer's disease by enhancing amyloid beta peptide deposits in the brain. In this review, effect of hot environment on CHI-induced brain pathology is discussed. In addition, whether nanodelivery of oxiracetam together with neprilysin and monoclonal antibodies (mAb) to amyloid beta peptide and p-tau could lead to superior neuroprotection in CHI is explored. Our results show that co-administration of oxiracetam with neprilysin and mAb to AβP and p-tau significantly induced superior neuroprotection following CHI in hot environment, not reported earlier.
Collapse
Affiliation(s)
- Ala Nozari
- Anesthesiology & Intensive Care, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Zhenguo Wang
- Shijiazhuang Pharma Group NBP Pharmaceutical Co., Ltd., Shijiazhuang, Hebei Province, China
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan, Hebei Province, China
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania
- "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, USA
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| |
Collapse
|
6
|
Lafuente JV, Sharma A, Feng L, Muresanu DF, Nozari A, Tian ZR, Buzoianu AD, Sjöquist PO, Wiklund L, Sharma HS. Nanowired Delivery of Mesenchymal Stem Cells with Antioxidant Compound H-290/51 Reduces Exacerbation of Methamphetamine Neurotoxicity in Hot Environment. ADVANCES IN NEUROBIOLOGY 2023; 32:317-352. [PMID: 37480465 DOI: 10.1007/978-3-031-32997-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Military personnel are often exposed to hot environments either for combat operations or peacekeeping missions. Hot environment is a severe stressful situation leading to profound hyperthermia, fatigue and neurological impairments. To avoid stressful environment, some people frequently use methamphetamine (METH) or other psychostimulants to feel comfortable under adverse situations. Our studies show that heat stress alone induces breakdown of the blood-brain barrier (BBB) and edema formation associated with reduced cerebral blood flow (CBF). On the other hand, METH alone induces hyperthermia and neurotoxicity. These effects of METH are exacerbated at high ambient temperatures as seen with greater breakdown of the BBB and brain pathology. Thus, a combination of METH use at hot environment may further enhance the brain damage-associated behavioral dysfunctions. METH is well known to induce severe oxidative stress leading to brain pathology. In this investigation, METH intoxication at hot environment was examined on brain pathology and to explore suitable strategies to induce neuroprotection. Accordingly, TiO2-nanowired delivery of H-290/51 (150 mg/kg, i.p.), a potent chain-breaking antioxidant in combination with mesenchymal stem cells (MSCs), is investigated in attenuating METH-induced brain damage at hot environment in model experiments. Our results show that nanodelivery of H-290/51 with MSCs significantly enhanced CBF and reduced BBB breakdown, edema formation and brain pathology following METH exposure at hot environment. These observations are the first to point out that METH exacerbated brain pathology at hot environment probably due to enhanced oxidative stress, and MSCs attenuate these adverse effects, not reported earlier.
Collapse
Affiliation(s)
- José Vicente Lafuente
- LaNCE, Department Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan, Hebei Province, China
| | - Dafin F Muresanu
- Department Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania
- "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Ala Nozari
- Anesthesiology & Intensive Care, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Z Ryan Tian
- Department Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, USA
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Per-Ove Sjöquist
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
7
|
Sharma HS, Lafuente JV, Muresanu DF, Sahib S, Tian ZR, Menon PK, Castellani RJ, Nozari A, Buzoianu AD, Sjöquist PO, Patnaik R, Wiklund L, Sharma A. Neuroprotective effects of insulin like growth factor-1 on engineered metal nanoparticles Ag, Cu and Al induced blood-brain barrier breakdown, edema formation, oxidative stress, upregulation of neuronal nitric oxide synthase and brain pathology. PROGRESS IN BRAIN RESEARCH 2021; 266:97-121. [PMID: 34689867 DOI: 10.1016/bs.pbr.2021.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Military personnel are vulnerable to environmental or industrial exposure of engineered nanoparticles (NPs) from metals. Long-term exposure of NPs from various sources affect sensory-motor or cognitive brain functions. Thus, a possibility exists that chronic exposure of NPs affect blood-brain barrier (BBB) breakdown and brain pathology by inducing oxidative stress and/or nitric oxide production. This hypothesis was examined in the rat intoxicated with Ag, Cu or Al (50-60nm) nanoparticles (50mg/kg, i.p. once daily) for 7 days. In these NPs treated rats the BBB permeability, brain edema, neuronal nitric oxide synthase (nNOS) immunoreactivity and brain oxidants levels, e.g., myeloperoxidase (MP), malondialdehyde (MD) and glutathione (GT) was examined on the 8th day. Cu and Ag but not Al nanoparticles increased the MP and MD levels by twofold in the brain although, GT showed 50% decline. At this time increase in brain water content and BBB breakdown to protein tracers were seen in areas exhibiting nNOS positive neurons and cell injuries. Pretreatment with insulin like growth factor-1 (IGF-1) in high doses (1μg/kg, i.v. but not 0.5μg/kg daily for 7 days) together with NPs significantly reduced the oxidative stress, nNOS upregulation, BBB breakdown, edema formation and cell injuries. These novel observations demonstrate that (i) NPs depending on their metal constituent (Cu, Ag but not Al) induce oxidative stress and nNOS expression leading to BBB disruption, brain edema and cell damage, and (ii) IGF-1 depending on doses exerts powerful neuroprotection against nanoneurotoxicity, not reported earlier.
Collapse
Affiliation(s)
- Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Preeti K Menon
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Per-Ove Sjöquist
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
8
|
TBHQ-Overview of Multiple Mechanisms against Oxidative Stress for Attenuating Methamphetamine-Induced Neurotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8874304. [PMID: 33354283 PMCID: PMC7735854 DOI: 10.1155/2020/8874304] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022]
Abstract
Methamphetamine is a derivative of amphetamines, a highly addictive central stimulant with multiple systemic toxicity including the brain, heart, liver, lung, and spleen. It has adverse effects such as apoptosis and breakdown of the blood-brain barrier. Methamphetamine is a fatal and toxic chemical substance, and its lethal mechanism has been widely studied in recent years. The possible mechanism is that methamphetamine can cause cardiotoxicity and neurotoxicity mainly by inducing oxidative stress so as to generate heat, eliminate people's hunger and thirst, and maintain a state of excitement so that people can continue to exercise. According to many research, there is no doubt that methamphetamine triggers neurotoxicity by inducing reactive oxygen species (ROS) production and redox imbalance. This review summarized the mechanisms of methamphetamine-induced neurotoxicity including apoptosis and blood-brain barrier breakdown through oxidative stress and analyzed several possible antioxidative mechanisms of tert-butylhydroquinone (TBHQ) which is a kind of food additive with antioxidative effects. As a nuclear factor E2-related factor 2 (Nrf2) agonist, TBHQ may inhibit neurotoxicity caused by oxidative stress through the following three mechanisms: the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase system, the astrocytes activation, and the glutathione pathway. The mechanism about methamphetamine's toxic effects and its antioxidative therapeutic drugs would become a research hotspot in this field and has very important research significance.
Collapse
|
9
|
Sancho Santos ME, Grabicová K, Steinbach C, Schmidt-Posthaus H, Šálková E, Kolářová J, Vojs Staňová A, Grabic R, Randák T. Environmental concentration of methamphetamine induces pathological changes in brown trout (Salmo trutta fario). CHEMOSPHERE 2020; 254:126882. [PMID: 32957289 DOI: 10.1016/j.chemosphere.2020.126882] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/03/2020] [Accepted: 04/23/2020] [Indexed: 06/11/2023]
Abstract
Methamphetamine, mainly consumed as an illicit drug, is a potent addictive psychostimulant that has been detected in surface water at concentrations ranging from nanograms to micrograms per litre, especially in Middle and East Europe. The aim of this study was to expose brown trout (Salmo trutta fario) to environmental (1 μg L-1) and higher (50 μg L-1) concentrations of methamphetamine for 35 days with a four-day depuration phase to assess the possible negative effects on fish health. Degenerative liver and heart alterations, similar to those described in mammals, were observed at both concentrations, although at different intensities. Apoptotic changes in hepatocytes, revealed by activated caspase-3, were found in exposed fish. The parent compound and a metabolite (amphetamine) were detected in fish tissues in both concentration groups, in the order of kidney > liver > brain > muscle > plasma. Bioconcentration factors ranged from 0.13 to 80. A therapeutic plasma concentration was reached for both compounds in the high-concentration treatment. This study indicates that chronic environmental concentrations of methamphetamine can lead to health issues in aquatic organisms.
Collapse
Affiliation(s)
- Maria Eugenia Sancho Santos
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic.
| | - Kateřina Grabicová
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic
| | - Christoph Steinbach
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic
| | - Heike Schmidt-Posthaus
- University of Bern, Centre for Fish and Wildlife Health, Department of Infectious Diseases and Pathobiology, Laenggassstrasse 122, 3001, Bern, Switzerland
| | - Eva Šálková
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic
| | - Jitka Kolářová
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic
| | - Andrea Vojs Staňová
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic; Comenius University in Bratislava, Faculty of Natural Sciences, Department of Analytical Chemistry, Ilkovicova 6, SK-842 15, Bratislava, Slovak Republic
| | - Roman Grabic
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic
| | - Tomáš Randák
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic
| |
Collapse
|
10
|
Thuringer D, Garrido C. Molecular chaperones in the brain endothelial barrier: neurotoxicity or neuroprotection? FASEB J 2019; 33:11629-11639. [PMID: 31348679 DOI: 10.1096/fj.201900895r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Brain microvascular endothelial cells (BMECs) interact with astrocytes and pericytes to form the blood-brain barrier (BBB). Their compromised function alters the BBB integrity, which is associated with early events in the pathogenesis of cancer, neurodegenerative diseases, and epilepsy. Interestingly, these conditions also induce the expression of heat shock proteins (HSPs). Here we review the contribution of major HSP families to BMEC and BBB function. Although investigators mainly report protective effects of HSPs in brain, contrasted results were obtained in BMEC, which depend both on the HSP and on its location, intra- or extracellular. The therapeutic potential of HSPs must be scrupulously analyzed before targeting them in patients to reduce the progression of brain lesions and improve neurologic outcomes in the long term.-Thuringer, D., Garrido, C. Molecular chaperones in the brain endothelial barrier: neurotoxicity or neuroprotection?
Collapse
Affiliation(s)
- Dominique Thuringer
- INSERM Unité Mixte de Recherche (UMR) 1231, Institut Fédératif de Recherche en Santé-Sciences et Techniques de l'Information et de la Communication (IFR Santé-STIC), Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France
| | - Carmen Garrido
- INSERM Unité Mixte de Recherche (UMR) 1231, Institut Fédératif de Recherche en Santé-Sciences et Techniques de l'Information et de la Communication (IFR Santé-STIC), Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
11
|
|
12
|
|
13
|
Ondruschka B, Rosinsky F, Trauer H, Schneider E, Dreßler J, Franke H. Drug- and/or trauma-induced hyperthermia? Characterization of HSP70 and myoglobin expression. PLoS One 2018; 13:e0194442. [PMID: 29566034 PMCID: PMC5864017 DOI: 10.1371/journal.pone.0194442] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 03/02/2018] [Indexed: 01/04/2023] Open
Abstract
Introduction Heat shock protein 70 (HSP70) expression could be discussed as an adaption that promotes repair and counteracts cell damage. Myoglobin is released upon muscle damage of several pathways. The purpose of the present study was to determine whether the expression of HSP70 in kidney, heart and brain and of myoglobin in the kidney were associated with the cause of death and the survival times after lethal intoxications with three of the drugs most widely used in our local area (Saxony, Germany) as well as after fatal traumatic brain injury (TBI). Methods We retrospectively collected kidney, heart and brain samples of 50 autopsy cases with toxicological proved lethal intoxication (main drugs methamphetamine, morphine, alcohol), 14 TBI cases and 15 fatalities with acute myocardial injury in age- and gender-matched compilations. Results Our main findings suggest that HSP70 is associated with hyperthermal and other stress factors of most cell populations. HSP70 expressions in kidney and heart muscle are useful for a differentiation between fatal intoxications and cases without toxicological influence (p < 0.05). There were significant differences in the cerebral expression patterns between methamphetamine- and morphine-associated deaths compared to alcohol fatalities (p < 0.05). An intensive staining of HSP70 in the pericontusional zone and the hippocampus after TBI (especially neuronal and vascular) was shown even after short survival times and may be useful as an additional marker in questions of vitality or wound age. A relevant myoglobin decoration of renal tubules was only shown for methamphetamine abuse in the study presented. Conclusion In sum, the immunohistochemical characteristics presented can be supportive for determining final death circumstances and minimal trauma survival times but are not isolated usefully for the detection of drug- or trauma-induced hyperthermia.
Collapse
Affiliation(s)
- Benjamin Ondruschka
- Institute of Legal Medicine, Medical Faculty, University of Leipzig, Leipzig, Germany
- * E-mail:
| | - Franziska Rosinsky
- Institute of Legal Medicine, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Heiner Trauer
- Institute of Legal Medicine, Medical Faculty, University of Leipzig, Leipzig, Germany
| | | | - Jan Dreßler
- Institute of Legal Medicine, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Heike Franke
- Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig, Leipzig, Germany
| |
Collapse
|
14
|
Eng JWL, Reed CB, Kokolus KM, Repasky EA. Housing temperature influences the pattern of heat shock protein induction in mice following mild whole body hyperthermia. Int J Hyperthermia 2015; 30:540-6. [PMID: 25430986 DOI: 10.3109/02656736.2014.981300] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Researchers studying the murine response to stress generally use mice housed under standard, nationally mandated conditions as controls. Few investigators are concerned whether basic physical aspects of mouse housing could be an additional source of stress, capable of influencing the subsequent impact of an experimentally applied stressor. We have recently become aware of the potential for housing conditions to impact important physiological and immunological properties in mice. MATERIALS AND METHODS Here we sought to determine whether housing mice at standard temperature (ST; 22 °C) vs. thermoneutral temperature (TT; 30 °C) influences baseline expression of heat shock proteins (HSPs) and their typical induction following a whole body heating. RESULTS There were no significant differences in baseline expression of HSPs at ST and TT. However, in several cases, the induction of Hsp70, Hsp110 and Hsp90 in tissues of mice maintained at ST was greater than at TT following 6 h of heating (which elevated core body temperature to 39.5 °C). This loss of HSP induction was also seen when mice housed at ST were treated with propranolol, a β-adrenergic receptor antagonist, used clinically to treat hypertension and stress. CONCLUSIONS Taken together, these data show that housing temperature significantly influences the expression of HSPs in mice after whole body heating and thus should be considered when stress responses are studied in mice.
Collapse
Affiliation(s)
- Jason W-L Eng
- Department of Immunology, Roswell Park Cancer Institute , Buffalo, New York , USA
| | | | | | | |
Collapse
|
15
|
Sharma HS, Muresanu DF, Lafuente JV, Sjöquist PO, Patnaik R, Sharma A. Nanoparticles Exacerbate Both Ubiquitin and Heat Shock Protein Expressions in Spinal Cord Injury: Neuroprotective Effects of the Proteasome Inhibitor Carfilzomib and the Antioxidant Compound H-290/51. Mol Neurobiol 2015; 52:882-98. [DOI: 10.1007/s12035-015-9297-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Indexed: 12/22/2022]
|
16
|
Damasceno WC, Pires W, Lima MRM, Lima NRV, Wanner SP. The dynamics of physical exercise-induced increases in thalamic and abdominal temperatures are modified by central cholinergic stimulation. Neurosci Lett 2015; 590:193-8. [PMID: 25655022 DOI: 10.1016/j.neulet.2015.01.082] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 01/14/2015] [Accepted: 01/30/2015] [Indexed: 10/24/2022]
Abstract
Evidence has shown that brain and abdominal (T abd) temperatures are regulated by distinct physiological mechanisms. Thus, the present study examined whether central cholinergic stimulation would change the dynamics of exercise-induced increases in T abd and thalamic temperature (T thal), an index of brain temperature. Adult male Wistar rats were used in all of the experiments. Two guide cannulae were implanted in the rats, one in the thalamus and the other in the right lateral cerebral ventricle, to measure T thal and to centrally inject a cholinergic agonist, respectively. Then, a temperature sensor was implanted in the abdominal cavity. On the day of the experiments, the rats received an intracerebroventricular injection of 2 μL of 10(-2)M physostigmine (Phy) or a vehicle solution (Veh) and were subjected to treadmill running until volitional fatigue occurred. T thal was measured using a thermistor connected to a multimeter, and T abd was recorded by telemetry. Phy injection delayed the exercise-induced increases in T thal (37.6 ± 0.2°C Phy vs 38.7 ± 0.1°C Veh at the 10th min of exercise) and in T abd. Despite the delayed hyperthermia, Phy did not change the rats' physical performance. In addition, the more rapid exercise-induced increase in T thal relative to Tabd in the rats treated with Veh was abolished by Phy. Collectively, our data indicate that central cholinergic stimulation affects the dynamics of exercise-induced increases in T thal and T abd. These results also provide evidence of the involvement of cholinoceptors in the modulation of brain heat loss during physical exercise.
Collapse
Affiliation(s)
- William Coutinho Damasceno
- Exercise Physiology Laboratory, Department of Physical Education, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Washington Pires
- Exercise Physiology Laboratory, Department of Physical Education, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Milene Rodrigues Malheiros Lima
- Exercise Physiology Laboratory, Department of Physical Education, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Nilo Resende Viana Lima
- Exercise Physiology Laboratory, Department of Physical Education, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Samuel Penna Wanner
- Exercise Physiology Laboratory, Department of Physical Education, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
17
|
Abstract
The blood-brain barrier (BBB) is a large regulatory and exchange interface between the brain and peripheral circulation. We propose that changes of the BBB contribute to many pathophysiological processes in the brain of subjects with chronic sleep restriction (CSR). To achieve CSR that mimics a common pattern of human sleep loss, we quantified a new procedure of sleep disruption in mice by a week of consecutive sleep recording. We then tested the hypothesis that CSR compromises microvascular function. CSR not only diminished endothelial and inducible nitric oxide synthase, endothelin1, and glucose transporter expression in cerebral microvessels of the BBB, but it also decreased 2-deoxy-glucose uptake by the brain. The expression of several tight junction proteins also was decreased, whereas the level of cyclooxygenase-2 increased. This coincided with an increase of paracellular permeability of the BBB to the small tracers sodium fluorescein and biotin. CSR for 6 d was sufficient to impair BBB structure and function, although the increase of paracellular permeability returned to baseline after 24 h of recovery sleep. This merits attention not only in neuroscience research but also in public health policy and clinical practice.
Collapse
|
18
|
Sharma HS, Menon P, Lafuente JV, Muresanu DF, Tian ZR, Patnaik R, Sharma A. Development ofin vivodrug-induced neurotoxicity models. Expert Opin Drug Metab Toxicol 2014; 10:1637-61. [DOI: 10.1517/17425255.2014.970168] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
19
|
Methamphetamine-induced toxicity: an updated review on issues related to hyperthermia. Pharmacol Ther 2014; 144:28-40. [PMID: 24836729 DOI: 10.1016/j.pharmthera.2014.05.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 05/07/2014] [Indexed: 01/30/2023]
Abstract
Reports of methamphetamine-related emergency room visits suggest that elevated body temperature is a universal presenting symptom, with lethal overdoses generally associated with extreme hyperthermia. This review summarizes the available information on methamphetamine toxicity as it pertains to elevations in body temperature. First, a brief overview of thermoregulatory mechanisms is presented. Next, central and peripheral targets that have been considered for potential involvement in methamphetamine hyperthermia are discussed. Finally, future areas of investigation are proposed, as further studies are needed to provide greater insight into the mechanisms that mediate the alterations in body temperature elicited by methamphetamine.
Collapse
|
20
|
Shen H, Mohammad A, Ramroop J, Smith SS. A stress steroid triggers anxiety via increased expression of α4βδ GABAA receptors in methamphetamine dependence. Neuroscience 2013; 254:452-75. [PMID: 23994152 DOI: 10.1016/j.neuroscience.2013.08.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 08/13/2013] [Accepted: 08/20/2013] [Indexed: 10/26/2022]
Abstract
Methamphetamine (METH) is an addictive stimulant drug. In addition to drug craving and lethargy, METH withdrawal is associated with stress-triggered anxiety. However, the cellular basis for this stress-triggered anxiety is not understood. The present results suggest that during METH withdrawal (24h) following chronic exposure (3mg/kg, i.p. for 3-5weeks) of adult, male mice, the effect of one neurosteroid released by stress, 3α,5α-THP (3α-OH-5α-pregnan-20-one), and its 3α,5β isomer reverse to trigger anxiety assessed by the acoustic startle response (ASR), in contrast to their usual anti-anxiety effects. This novel effect of 3α,5β-THP was due to increased (three-fold) hippocampal expression of α4βδ GABAA receptors (GABARs) during METH withdrawal (24h-4weeks) because anxiogenic effects of 3α,5β-THP were not seen in α4-/- mice. 3α,5β-THP reduces current at these receptors when it is hyperpolarizing, as observed during METH withdrawal. As a result, 3α,5β-THP (30nM) increased neuronal excitability, assessed with current clamp and cell-attached recordings in CA1hippocampus, one CNS site which regulates anxiety. α4βδ GABARs were first increased 1h after METH exposure and recovered 6weeks after METH withdrawal. Similar increases in α4βδ GABARs and anxiogenic effects of 3α,5β-THP were noted in rats during METH withdrawal (24h). In contrast, the ASR was increased by chronic METH treatment in the absence of 3α,5β-THP administration due to its stimulant effect. Although α4βδ GABARs were increased by chronic METH treatment, the GABAergic current recorded from hippocampal neurons at this time was a depolarizing, shunting inhibition, which was potentiated by 3α,5β-THP. This steroid reduced neuronal excitability and anxiety during chronic METH treatment, consistent with its typical effect. Flumazenil (10mg/kg, i.p., 3×) reduced α4βδ expression and prevented the anxiogenic effect of 3α,5β-THP after METH withdrawal. Our findings suggest a novel mechanism underlying stress-triggered anxiety after METH withdrawal mediated by α4βδ GABARs.
Collapse
Affiliation(s)
- H Shen
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY 11203, United States
| | | | | | | |
Collapse
|
21
|
Karch SB. Possible Strategies for the Diagnosis of Fatal Excited Delirium Syndrome. Acad Forensic Pathol 2012. [DOI: 10.23907/2012.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Excited Delirium Syndrome (ExDS) is a term traditionally used in forensic literature to describe the symptoms and signs seen in a subgroup of patients with delirium who die in an agitated state. Components of this syndrome are altered mental status, combativeness and/or aggressiveness, increased tolerance to significant pain, tachypnea, profuse sweating, severe agitation, elevated temperature, delirium, and noncompliance with law enforcement and medical personnel. The individual may display “superhuman” strength and wear clothing inappropriate for the environment. Patients with this presentation are almost guaranteed to cause difficulties for law enforcement officers and medical staff. This review is written in hopes of minimizing some of these difficulties by 1) increasing general awareness and specific knowledge about this condition, 2) explaining the neurochemical and neuroanatomical alterations that have been shown to cause those symptoms, and 3) by suggesting new lines of research that might identify easily measured biomarkers for the disease. If the disease mechanism can be deciphered, then it should be possible to devise effective strategies for treatment. It would also be of enormous value to the legal system. When defending a diagnosis before the court, physical evidence always trumps knowledge and experience. It would be far better to be able to present physical proof than to opine that the decedent's behavior was typical for the disease. In this aspect, ExDS is analogous to myocardial infarction: if a man dies suddenly, it is much easier to prove the cardiac origin of the event if an occlusive thrombus is found in a major coronary artery.
Collapse
Affiliation(s)
- Steven B. Karch
- (Royal College of Physicians, London). Royal London Hospital and Cardiac Pathology at the Stanford Transplant Laboratory. San Francisco Medical Examiner
| |
Collapse
|
22
|
Kousik SM, Napier TC, Carvey PM. The effects of psychostimulant drugs on blood brain barrier function and neuroinflammation. Front Pharmacol 2012; 3:121. [PMID: 22754527 PMCID: PMC3386512 DOI: 10.3389/fphar.2012.00121] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 06/06/2012] [Indexed: 12/17/2022] Open
Abstract
The blood brain barrier (BBB) is a highly dynamic interface between the central nervous system (CNS) and periphery. The BBB is comprised of a number of components and is part of the larger neuro(glio)vascular unit. Current literature suggests that psychostimulant drugs of abuse alter the function of the BBB which likely contributes to the neurotoxicities associated with these drugs. In both preclinical and clinical studies, psychostimulants including methamphetamine, MDMA, cocaine, and nicotine, produce BBB dysfunction through alterations in tight junction protein expression and conformation, increased glial activation, increased enzyme activation related to BBB cytoskeleton remodeling, and induction of neuroinflammatory pathways. These detrimental changes lead to increased permeability of the BBB and subsequent vulnerability of the brain to peripheral toxins. In fact, abuse of these psychostimulants, notably methamphetamine and cocaine, has been shown to increase the invasion of peripheral bacteria and viruses into the brain. Much work in this field has focused on the co-morbidity of psychostimulant abuse and human immunodeficiency virus (HIV) infection. As psychostimulants alter BBB permeability, it is likely that this BBB dysfunction results in increased penetration of the HIV virus into the brain thus increasing the risk of and severity of neuro AIDS. This review will provide an overview of the specific changes in components within the BBB associated with psychostimulant abuse as well as the implications of these changes in exacerbating the neuropathology associated with psychostimulant drugs and HIV co-morbidity.
Collapse
Affiliation(s)
- Sharanya M Kousik
- Department of Pharmacology, Rush University Medical Center Chicago, IL, USA
| | | | | |
Collapse
|
23
|
Environmental conditions modulate neurotoxic effects of psychomotor stimulant drugs of abuse. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2012; 102:147-71. [PMID: 22748829 DOI: 10.1016/b978-0-12-386986-9.00006-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Psychomotor stimulants such as methamphetamine (METH), amphetamine, and 3,4-metylenedioxymethamphetamine (MDMA or ecstasy) are potent addictive drugs. While it is known that their abuse could result in adverse health complications, including neurotoxicity, both the environmental conditions and activity states associated with their intake could strongly enhance drug toxicity, often resulting in life-threatening health complications. In this review, we analyze results of animal experiments that suggest that even moderate increases in environmental temperatures and physiological activation, the conditions typical of human raves parties, dramatically potentiate brain hyperthermic effects of METH and MDMA. We demonstrate that METH also induces breakdown of the blood-brain barrier, acute glial activation, brain edema, and structural abnormalities of various subtypes of brain cells; these effects are also strongly enhanced when the drug is used at moderately warm environmental conditions. We consider the mechanisms underlying environmental modulation of acute drug neurotoxicity and focus on the role of brain temperature, a critical homeostatic parameter that could be affected by metabolism-enhancing drugs and environmental conditions and affect neural activity and functions.
Collapse
|
24
|
Beauvais G, Atwell K, Jayanthi S, Ladenheim B, Cadet JL. Involvement of dopamine receptors in binge methamphetamine-induced activation of endoplasmic reticulum and mitochondrial stress pathways. PLoS One 2011; 6:e28946. [PMID: 22174933 PMCID: PMC3236770 DOI: 10.1371/journal.pone.0028946] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 11/17/2011] [Indexed: 01/11/2023] Open
Abstract
Single large doses of methamphetamine (METH) cause endoplasmic reticulum (ER) stress and mitochondrial dysfunctions in rodent striata. The dopamine D1 receptor appears to be involved in these METH-mediated stresses. The purpose of this study was to investigate if dopamine D1 and D2 receptors are involved in ER and mitochondrial stresses caused by single-day METH binges in the rat striatum. Male Sprague-Dawley rats received 4 injections of 10 mg/kg of METH alone or in combination with a putative D1 or D2 receptor antagonist, SCH23390 or raclopride, respectively, given 30 min prior to each METH injection. Rats were euthanized at various timepoints afterwards. Striatal tissues were used in quantitative RT-PCR and western blot analyses. We found that binge METH injections caused increased expression of the pro-survival genes, BiP/GRP-78 and P58IPK, in a SCH23390-sensitive manner. METH also caused up-regulation of ER-stress genes, Atf2, Atf3, Atf4, CHOP/Gadd153 and Gadd34. The expression of heat shock proteins (HSPs) was increased after METH injections. SCH23390 completely blocked induction in all analyzed ER stress-related proteins that included ATF3, ATF4, CHOP/Gadd153, HSPs and caspase-12. The dopamine D2-like antagonist, raclopride, exerted small to moderate inhibitory influence on some METH-induced changes in ER stress proteins. Importantly, METH caused decreases in the mitochondrial anti-apoptotic protein, Bcl-2, but increases in the pro-apoptotic proteins, Bax, Bad and cytochrome c, in a SCH23390-sensitive fashion. In contrast, raclopride provided only small inhibition of METH-induced changes in mitochondrial proteins. These findings indicate that METH-induced activation of striatal ER and mitochondrial stress pathways might be more related to activation of SCH23390-sensitive receptors.
Collapse
Affiliation(s)
- Genevieve Beauvais
- Molecular Neuropsychiatry Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland, United States of America
- Faculté de Pharmacie, Université Paris Descartes, Paris, France
| | - Kenisha Atwell
- Molecular Neuropsychiatry Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland, United States of America
| | - Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland, United States of America
| | - Bruce Ladenheim
- Molecular Neuropsychiatry Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland, United States of America
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
25
|
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
|
26
|
Low ambient temperature reveals distinct mechanisms for MDMA-induced serotonergic toxicity and astroglial Hsp27 heat shock response in rat brain. Neurochem Int 2011; 59:695-705. [PMID: 21756954 DOI: 10.1016/j.neuint.2011.06.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 06/21/2011] [Indexed: 11/23/2022]
Abstract
3,4-Methylenedioxymethamphetamine (MDMA, 'ecstasy') is a widely used recreational drug known to cause selective long-term serotonergic damage. In our recent paper we described region-specific, dose-dependent increase in the protein expression of astroglial Hsp27 and neuronal Hsp72 molecular chaperones after MDMA administration of rats. Here, we examined the possible interaction of elevated Hsp27 protein level to hyperthermic responses after MDMA administration and its separation from drug-induced serotonergic neurotoxicity. For this, 7-8 week old male Dark Agouti rats were treated with 15 mg/kg i.p. MDMA. Treatment at an ambient temperature of 22 ± 1°C caused a significant elevation of the rectal temperature, an increase of Hsp27 immunoreactive protoplasmic astrocytes in the hippocampus, the parietal and cingulate cortices, and a significant decrease in the density of tryptophan hydroxylase immunoreactive fibers in the same brain regions, 8h as well as 24h after drug administrations. In addition, serotonergic axons exhibited numerous swollen varicosities and fragmented morphology. MDMA treatment at low ambient temperature (10 ± 2°C) almost completely abolished the elevation of body temperature and the increased astroglial Hsp27 expression but failed to alter - or just slightly attenuated - the depletion in the density of tryptophan hydroxylase immunoreactive fibers. These results suggest that the increased astroglial Hsp27 protein expression is rather related to the hyperthermic response after the drug administration and it could be separated from the serotonergic neurotoxicity caused by MDMA. In addition, the induction of Hsp27 per se is uneffective to protect serotonergic fibers after MDMA administration. Our results also suggest that Tph immunohistochemistry is an early and sensitive method to demonstrate MDMA-caused vulnerability.
Collapse
|