1
|
Zhang W, Cao F, Li M, Xu Z, Sun J, Huang Z, Shi P. The involvement of calcium in the toxic effect of 4-methylethcathinone on SH-SY5Y cells. J Appl Toxicol 2024; 44:553-563. [PMID: 37950502 DOI: 10.1002/jat.4560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/05/2023] [Accepted: 10/18/2023] [Indexed: 11/12/2023]
Abstract
Neurotoxicity induced by psychoactive substances is often accompanied by an imbalance of intracellular calcium ions. It is unclear whether calcium ions play a role in the toxicity induced by psychoactive substances. In the present study, we aimed to evaluate the occurrence of calcium dysregulation and its contribution to cytotoxicity in human neurotypic SH-SY5Y cells challenged with a recently developed psychoactive substance 4-methylethcathinone (4-MEC). An increase in the intracellular calcium was detected by inductively coupled plasma atomic emission spectrometry and Fluo-3 AM dye in SH-SY5Y cells after being treated with 4-MEC. The increase of intracellular Ca2+ level mediated G0/G1 cell cycle arrest and ROS/endoplasmic reticulum stress-autophagy signaling pathways to achieve the toxicity of 4-MEC. In particular, N-acetyl-L-cysteine, a classical antioxidant, was found to be a potential treatment for 4-MEC-induced toxicity. Taken together, our results demonstrate that an increase in intracellular calcium content is one of the mechanisms of 4-MEC-induced toxicity. This study provides a molecular basis for the toxicity mechanism and therapeutic intervention of psychoactive substances.
Collapse
Affiliation(s)
- Wei Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Fangqi Cao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai, China
| | - Ming Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Zhiwen Xu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Jing Sun
- Qinghai Key Laboratory of Qinghai-Tibet Plateau Biological Resources, Northwest Institute of Plateau Biology, The Chinese Academy of Sciences, Xining, China
| | - Zhiwei Huang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Ping Shi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
2
|
Seaman RW, Lamon K, Whitton N, Latimer B, Sulima A, Rice KC, Murnane KS, Collins GT. Impacts of Self-Administered 3,4-Methylenedioxypyrovalerone (MDPV) Alone, and in Combination with Caffeine, on Recognition Memory and Striatal Monoamine Neurochemistry in Male Sprague Dawley Rats: Comparisons with Methamphetamine and Cocaine. Brain Sci 2024; 14:258. [PMID: 38539646 PMCID: PMC10969043 DOI: 10.3390/brainsci14030258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Recent data suggest that 3,4-methylenedioxypyrovalerone (MDPV) has neurotoxic effects; however, the cognitive and neurochemical consequences of MDPV self-administration remain largely unexplored. Furthermore, despite the fact that drug preparations that contain MDPV often also contain caffeine, little is known regarding the toxic effects produced by the co-use of these two stimulants. The current study investigated the degree to which self-administered MDPV or a mixture of MDPV+caffeine can produce deficits in recognition memory and alter neurochemistry relative to prototypical stimulants. Male Sprague Dawley rats were provided 90 min or 12 h access to MDPV, MDPV+caffeine, methamphetamine, cocaine, or saline for 6 weeks. Novel object recognition (NOR) memory was evaluated prior to any drug self-administration history and 3 weeks after the final self-administration session. Rats that had 12 h access to methamphetamine and those that had 90 min or 12 h access to MDPV+caffeine exhibited significant deficits in NOR, whereas no significant deficits were observed in rats that self-administered cocaine or MDPV. Striatal monoamine levels were not systematically affected. These data demonstrate synergism between MDPV and caffeine with regard to producing recognition memory deficits, highlighting the importance of recapitulating the manner in which drugs are used (e.g., in mixtures containing multiple stimulants, binge-like patterns of intake).
Collapse
Affiliation(s)
- Robert W. Seaman
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Kariann Lamon
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Psychiatry and Behavioral Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Nicholas Whitton
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Psychiatry and Behavioral Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Brian Latimer
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Psychiatry and Behavioral Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Agnieszka Sulima
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kenner C. Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kevin S. Murnane
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Psychiatry and Behavioral Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Gregory T. Collins
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| |
Collapse
|
3
|
Seaman RW, Lamon K, Whitton N, Latimer B, Sulima A, Rice KC, Murnane KS, Collins GT. Impacts of Self-Administered 3,4-Methylenedioxypyrovalerone (MDPV) Alone, and in Combination with Caffeine, on Recognition Memory and Striatal Monoamine Neurochemistry in Male Sprague-Dawley Rats: Comparisons with Methamphetamine and Cocaine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578247. [PMID: 38352595 PMCID: PMC10862826 DOI: 10.1101/2024.01.31.578247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Recent data suggest that 3,4-methylenedioxypyrovalerone (MDPV) has neurotoxic effects; however, the cognitive and neurochemical consequences of MDPV self-administration remain largely unexplored. Furthermore, despite the fact that drug preparations that contain MDPV often also contain caffeine, little is known regarding the toxic effects produced by the co-use of these two stimulants. The current study investigated the degree to which self-administered MDPV, or a mixture of MDPV+caffeine can produce deficits in recognition memory and alter neurochemistry relative to prototypical stimulants. Male Sprague-Dawley rats were provided 90-min or 12-h access to MDPV, MDPV+caffeine, methamphetamine, cocaine, or saline for 6 weeks. Novel object recognition (NOR) memory was evaluated prior to any drug self-administration history and 3 weeks after the final self-administration session. Rats that had 12-h access to methamphetamine and those that had 90-min or 12-h access to MDPV+caffeine exhibited significant deficits in NOR, whereas no significant deficits were observed in rats that self-administered cocaine or MDPV. Striatal mono-amine levels were not systematically affected. These data demonstrate synergism between MDPV and caffeine with regard to producing recognition memory deficits and lethality, highlighting the importance of recapitulating the manner in which drugs are used (e.g., in mixtures containing multiple stimulants, binge-like patterns of intake).
Collapse
Affiliation(s)
- Robert W Seaman
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Kariann Lamon
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
- Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
- Department of Psychiatry and Behavioral Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
| | - Nicholas Whitton
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
- Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
- Department of Psychiatry and Behavioral Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
| | - Brian Latimer
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
- Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
- Department of Psychiatry and Behavioral Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
| | - Agnieszka Sulima
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| | - Kenner C Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| | - Kevin S Murnane
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
- Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
- Department of Psychiatry and Behavioral Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
| | - Gregory T Collins
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- South Texas Veterans Health Care System, San Antonio, TX, United States
| |
Collapse
|
4
|
Roda E, De Luca F, Priori EC, Ratto D, Pinelli S, Corradini E, Mozzoni P, Poli D, Mazzini G, Bottone MG, Gatti AM, Marti M, Locatelli CA, Rossi P, Bottai D. The Designer Drug αPHP Affected Cell Proliferation and Triggered Deathly Mechanisms in Murine Neural Stem/Progenitor Cells. BIOLOGY 2023; 12:1225. [PMID: 37759624 PMCID: PMC10525791 DOI: 10.3390/biology12091225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/08/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
Increasing reports of neurological and psychiatric outcomes due to psychostimulant synthetic cathinones (SCs) have recently raised public concern. However, the understanding of neurotoxic mechanisms is still lacking, particularly for the under-investigated αPHP, one of the major MDPV derivatives. In particular, its effects on neural stem/progenitor cell cultures (NSPCs) are still unexplored. Therefore, in the current in vitro study, the effects of increasing αPHP concentrations (25-2000 μM), on cell viability/proliferation, morphology/ultrastructure, genotoxicity and cell death pathways, have been evaluated after exposure in murine NSPCs, using a battery of complementary techniques, i.e., MTT and clonogenic assay, flow cytometry, immunocytochemistry, TEM, and patch clamp. We revealed that αPHP was able to induce a dose-dependent significant decrease of the viability, proliferation and clonal capability of the NSPCs, paralleled by the resting membrane potential depolarization and apoptotic/autophagic/necroptotic pathway activation. Moreover, ultrastructural alterations were clearly observed. Overall, our current findings demonstrate that αPHP, damaging NSPCs and the morpho-functional fundamental units of adult neurogenic niches may affect neurogenesis, possibly triggering long-lasting, irreversible CNS damage. The present investigation could pave the way for a broadened understanding of SCs toxicology, needed to establish an appropriate treatment for NPS and the potential consequences for public health.
Collapse
Affiliation(s)
- Elisa Roda
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy (C.A.L.)
| | - Fabrizio De Luca
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Erica Cecilia Priori
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Daniela Ratto
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Silvana Pinelli
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Emilia Corradini
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Paola Mozzoni
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Diana Poli
- INAIL Research, Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Via Fontana Candida, 1, 00078 Monte Porzio Catone, Italy
| | - Giuliano Mazzini
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
- Institute of Molecular Genetics—CNR (National Research Council), 27100 Pavia, Italy
| | - Maria Grazia Bottone
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Anna Maria Gatti
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy (C.A.L.)
| | - Matteo Marti
- Department of Translational Medicine, Section of Legal Medicine, LTTA Center and University Center of Gender Medicine, University of Ferrara, 44121 Ferrara, Italy;
- Collaborative Centre for the Italian National Early Warning System, Department of Anti-Drug Policies, Presidency of the Council of Ministers, 44121 Ferrara, Italy
| | - Carlo Alessandro Locatelli
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy (C.A.L.)
| | - Paola Rossi
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Daniele Bottai
- Department of Pharmaceutical Sciences, Section of Pharmacology and Biosciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy;
| |
Collapse
|
5
|
Semi-Preparative Separation, Absolute Configuration, Stereochemical Stability and Effects on Human Neuronal Cells of MDPV Enantiomers. Molecules 2023; 28:molecules28052121. [PMID: 36903367 PMCID: PMC10003790 DOI: 10.3390/molecules28052121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
Synthetic cathinones, such as 3,4-methylenedioxypyrovalerone (MDPV), are widely abused due to their psychostimulant effects. As they are chiral molecules, studies of their stereochemical stability (racemization can occur in certain temperatures and acidic/basic environments) and of their biological and/or toxicity effects (enantiomers might display different properties) are of great relevance. In this study, the liquid chromatography (LC) semi-preparative enantioresolution of MDPV was optimized to collect both enantiomers with high recovery rates and enantiomeric ratio (e.r.) values. The absolute configuration of the MDPV enantiomers was determined by electronic circular dichroism (ECD) with the aid of theoretical calculations. The first eluted enantiomer was identified as S-(-)-MDPV and the second eluted enantiomer was identified as R-(+)-MDPV. A racemization study was performed by LC-UV, showing enantiomers' stability up to 48 h at room temperature and 24 h at 37 °C. Racemization was only affected by higher temperatures. The potential enantioselectivity of MDPV in cytotoxicity and in the expression of neuroplasticity-involved proteins-brain-derived neurotrophic factor (BDNF) and cyclin-dependent kinase 5 (Cdk5)-was also evaluated using SH-SY5Y neuroblastoma cells. No enantioselectivity was observed.
Collapse
|
6
|
Lv X, Fan Z, Cao F, Liu W, Huang Z, Shi P. Clioquinol induces autophagy by down-regulation of calreticulin in human neurotypic SH-SY5Y cells. Chem Biol Interact 2023; 369:110268. [PMID: 36396104 DOI: 10.1016/j.cbi.2022.110268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/04/2022] [Accepted: 11/12/2022] [Indexed: 11/16/2022]
Abstract
Clioquinol (CQ) is considered as a promising drug of neurodegenerative diseases. However, the underlying mechanism is unclear. Our previous study has proved that CQ induces S-phase cell cycle arrest through the elevation of intracellular calcium concentration ([Ca2+]i) with high levels of SERCA2. Furthermore, it could induce autophagy in an intracellular calcium independent manner in human neurotypic SH-SY5Y cells. In this study, the involvement of calreticulin (CRT) in autophagy induced by CQ was investigated. Our results illustrated the endoplasmic reticulum (ER) stress induced by CQ and DTT led to the cell death in different manners. DTT, an ER stress positive control, induced UPR accompanied with up-regulation of CRT and apoptosis, while CQ inhibited UPR accompanied with down-regulation of CRT,resulting in autophagy. Then, overexpression of CRT was shown to cause UPR and decrease [Ca2+]i, leading to cell apoptosis and inhibition of S-phase arrest induced by CQ. While the UPR was alleviated and autophagy was further enhanced in CRT deficient cells by using targeted siRNA. Meanwhile, down-regulation of CRT resulted in [Ca2+]i overload and induction of S-phase arrest. Finally, we found that the effect of CQ on the HT22 cells was similar to that on the SH-SY5Y cells. Our data showed for the first time that CQ decreased expression of CRT, leading to autophagy, an increase of [Ca2+]i, and cell S-phase arrest in the neurotypic cells. The present study describes the cellular signal pathways regulating autophagy by CQ and highlights the potential therapeutic application of CQ in neurodegenerative disorders.
Collapse
Affiliation(s)
- Xiaoguang Lv
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Zheyu Fan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Fangqi Cao
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Zhongshan North No 1 Road, Shanghai, 200083, China
| | - Wenbin Liu
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Zhongshan North No 1 Road, Shanghai, 200083, China.
| | - Zhiwei Huang
- Key Lab of Science & Technology of Eco-textile, Ministry of Education, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Ping Shi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
7
|
Jie L, Hong RT, Zhang YJ, Sha LL, Chen W, Ren XF. Melatonin Alleviates Liver Fibrosis by Inhibiting Autophagy. Curr Med Sci 2022; 42:498-504. [PMID: 35583587 DOI: 10.1007/s11596-022-2530-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 06/18/2021] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Melatonin has been reported to suppress inflammation and alleviate liver fibrosis, but its effect on autophagy in liver fibrosis has not been studied. This study investigated the effect of melatonin on autophagy in an animal model of liver fibrosis and the hepatic stellate cell (HSC)-T6 cell line. METHODS The model was established in rats through carbon tetrachloride treatment, and melatonin was administered at three doses (2.5, 5.0, and 10.0 mg/kg). Haematoxylin and eosin staining and Van Gieson's staining were performed to examine the pathological changes of liver. The expression of alpha-smooth muscle actin (α-SMA) and Beclin1 in liver tissues was detected by immunohistochemical staining. The protein levels of α-SMA, Beclin1 and LC3 in the animal model were detected by Western blot analysis, and gene levels of Beclin1 and LC3 were detected by quantitative real-time PCR (qRT-PCR) in the animal model. HSC-T6 cells were activated by platelet-derived growth factor-BB (PDGF-BB). The expression of α-SMA, Beclin1 and collagen I was detected by Western blot analysis, and the gene expression of Beclin1 and LC3 was detected by qRT-PCR. RESULTS Melatonin reduced the expression of α-SMA, Beclin1 and LC3 in liver tissues. In addition, melatonin inhibited the activation of HSC-T6 cells and the expression of α-SMA, Beclin1 and LC3 in these cells. These results revealed that melatonin could inhibit autophagy and HSC activation. CONCLUSION Melatonin might ameliorate liver fibrosis by regulating autophagy, suggesting that melatonin is a potential therapeutic agent for liver fibrosis.
Collapse
Affiliation(s)
- Lei Jie
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Digestive Disease of Anhui Province, Hefei, 230022, China
| | - Ru-Tao Hong
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Digestive Disease of Anhui Province, Hefei, 230022, China.
| | - Yu-Jie Zhang
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Digestive Disease of Anhui Province, Hefei, 230022, China
| | - Lu-Lin Sha
- Department of Critical Care Medicine of Cardiothoracic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Wei Chen
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Digestive Disease of Anhui Province, Hefei, 230022, China
| | - Xiao-Fei Ren
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Digestive Disease of Anhui Province, Hefei, 230022, China
| |
Collapse
|
8
|
Karam CS, Williams BL, Morozova I, Yuan Q, Panarsky R, Zhang Y, Hodgkinson CA, Goldman D, Kalachikov S, Javitch JA. Functional Genomic Analysis of Amphetamine Sensitivity in Drosophila. Front Psychiatry 2022; 13:831597. [PMID: 35250674 PMCID: PMC8894854 DOI: 10.3389/fpsyt.2022.831597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/24/2022] [Indexed: 11/14/2022] Open
Abstract
Abuse of psychostimulants, including amphetamines (AMPHs), is a major public health problem with profound psychiatric, medical, and psychosocial complications. The actions of these drugs at the dopamine transporter (DAT) play a critical role in their therapeutic efficacy as well as their liability for abuse and dependence. To date, however, the mechanisms that mediate these actions are not well-understood, and therapeutic interventions for AMPH abuse have been limited. Drug exposure can induce broad changes in gene expression that can contribute to neuroplasticity and effect long-lasting changes in neuronal function. Identifying genes and gene pathways perturbed by drug exposure is essential to our understanding of the molecular basis of drug addiction. In this study, we used Drosophila as a model to examine AMPH-induced transcriptional changes that are DAT-dependent, as those would be the most relevant to the stimulatory effects of the drug. Using this approach, we found genes involved in the control of mRNA translation to be significantly upregulated in response to AMPH in a DAT-dependent manner. To further prioritize genes for validation, we explored functional convergence between these genes and genes we identified in a genome-wide association study of AMPH sensitivity using the Drosophila Genetic Reference Panel. We validated a number of these genes by showing that they act specifically in dopamine neurons to mediate the behavioral effects of AMPH. Taken together, our data establish Drosophila as a powerful model that enables the integration of behavioral, genomic and transcriptomic data, followed by rapid gene validation, to investigate the molecular underpinnings of psychostimulant action.
Collapse
Affiliation(s)
- Caline S Karam
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Brenna L Williams
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Irina Morozova
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, United States.,Department of Chemical Engineering, Columbia University, New York, NY, United States
| | - Qiaoping Yuan
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Rony Panarsky
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Yuchao Zhang
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Colin A Hodgkinson
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Sergey Kalachikov
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, United States.,Department of Chemical Engineering, Columbia University, New York, NY, United States
| | - Jonathan A Javitch
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States.,Department of Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| |
Collapse
|
9
|
Dias da Silva D, Silva JP, Carmo H, Carvalho F. Neurotoxicity of psychoactive substances: A mechanistic overview. CURRENT OPINION IN TOXICOLOGY 2021. [DOI: 10.1016/j.cotox.2021.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
10
|
Sogos V, Caria P, Porcedda C, Mostallino R, Piras F, Miliano C, De Luca MA, Castelli MP. Human Neuronal Cell Lines as An In Vitro Toxicological Tool for the Evaluation of Novel Psychoactive Substances. Int J Mol Sci 2021; 22:ijms22136785. [PMID: 34202634 PMCID: PMC8268582 DOI: 10.3390/ijms22136785] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 12/18/2022] Open
Abstract
Novel psychoactive substances (NPS) are synthetic substances belonging to diverse groups, designed to mimic the effects of scheduled drugs, resulting in altered toxicity and potency. Up to now, information available on the pharmacology and toxicology of these new substances is very limited, posing a considerable challenge for prevention and treatment. The present in vitro study investigated the possible mechanisms of toxicity of two emerging NPS (i) 4′-methyl-alpha-pyrrolidinoexanophenone (3,4-MDPHP), a synthetic cathinone, and (ii) 2-chloro-4,5-methylenedioxymethamphetamine (2-Cl-4,5-MDMA), a phenethylamine. In addition, to apply our model to the class of synthetic opioids, we evaluated the toxicity of fentanyl, as a reference compound for this group of frequently abused substances. To this aim, the in vitro toxic effects of these three compounds were evaluated in dopaminergic-differentiated SH-SY5Y cells. Following 24 h of exposure, all compounds induced a loss of viability, and oxidative stress in a concentration-dependent manner. 2-Cl-4,5-MDMA activates apoptotic processes, while 3,4-MDPHP elicits cell death by necrosis. Fentanyl triggers cell death through both mechanisms. Increased expression levels of pro-apoptotic Bax and caspase 3 activity were observed following 2-Cl-4,5-MDMA and fentanyl, but not 3,4-MDPHP exposure, confirming the different modes of cell death.
Collapse
Affiliation(s)
- Valeria Sogos
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (V.S.); (P.C.); (C.P.); (R.M.); (F.P.); (M.A.D.L.)
| | - Paola Caria
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (V.S.); (P.C.); (C.P.); (R.M.); (F.P.); (M.A.D.L.)
| | - Clara Porcedda
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (V.S.); (P.C.); (C.P.); (R.M.); (F.P.); (M.A.D.L.)
| | - Rafaela Mostallino
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (V.S.); (P.C.); (C.P.); (R.M.); (F.P.); (M.A.D.L.)
| | - Franca Piras
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (V.S.); (P.C.); (C.P.); (R.M.); (F.P.); (M.A.D.L.)
| | - Cristina Miliano
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA;
| | - Maria Antonietta De Luca
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (V.S.); (P.C.); (C.P.); (R.M.); (F.P.); (M.A.D.L.)
| | - M. Paola Castelli
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (V.S.); (P.C.); (C.P.); (R.M.); (F.P.); (M.A.D.L.)
- Guy Everett Laboratory, University of Cagliari, 09042 Monserrato, Italy
- Center of Excellence “Neurobiology of Addiction”, University of Cagliari, 09042 Monserrato, Italy
- Correspondence: ; Tel.: +39-070-6754065
| |
Collapse
|
11
|
Jafarinejad-Farsangi S, Hashemi MS, Yazdi Rouholamini SE, Gharbi S, Ansari-Asl Z, Jafari E, Shiralizadeh Dezfuli A, Shahrokhi-Farjah M. Curcumin loaded on graphene nanosheets induced cell death in mammospheres from MCF-7 and primary breast tumor cells. Biomed Mater 2021; 16. [PMID: 34020433 DOI: 10.1088/1748-605x/ac0400] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 05/21/2021] [Indexed: 12/30/2022]
Abstract
Elimination of tumor cells is still a therapeutic challenge for breast cancer (BC) in men and women. Mammospheres serve as valuablein vitrotools for evaluating tumor behavior and sensitivity to anticancer treatments. Graphene nanosheets with unique physicochemical properties have been considered as potential biomedical approaches for drug delivery, bioimaging, and therapy. Graphene oxide (GO) and graphene quantum dots (GQDs) are suitable nanocarriers for hydrophobic and low bioaccessible anti-tumor materials like curcumin. Despite extensive studies on the potential application of graphene nanosheets in medicine, our knowledge of how different cells function and respond to these nanoparticles remains limited. Here, we evaluated cell death in mammospheres from MCF-7 and primary tumor cells in response to curcumin loaded on graphene nanosheets. Mammospheres were exposed to graphene oxide-curcumin (GO-Cur) and graphene quantum dots-curcumin (GQDs-Cur), and the incidence of cell death was evaluated by Hoechst 33342/propidium iodide double staining and flow cytometry. Besides, the expression of miR-21, miR-29a, Bax, and Bcl-2 genes were assessed using RT-qPCR. We observed, GO, and GQDs had no cytotoxic effect on Kerman male breast cancer/71 (KMBC/71) and MCF-7 tumor cells, while curcumin induced death in more than 50% of tumor cells. GO-Cur and GQDs-Cur synergistically enhanced anti-tumor activity of curcumin. Moreover, GQDs-Cur induced cell death in almost all cells of KMBC/71 mammospheres (99%;p< 0.0001). In contrast, GO-Cur induced cell death in only 21% of MCF-7 mammosphere cells (p< 0.0001). Also, the expression pattern of miR-21, miR-29a, and Bax/Bcl-2 ratio in KMBC/71 and MCF-7 mammospheres was different in response to GO-Cur and GQDs-Cur. Although KMBC/71 and MCF-7 tumor cells had similar clinical features and displayed similar responses to curcumin, more investigations are needed to clarify the detailed molecular mechanisms underlying observed differences in response to GO-Cur and GQDs-Cur.
Collapse
Affiliation(s)
| | - Mahnaz Sadat Hashemi
- Student Research Committee, School of Medicine, Kerman University of Medical Science, Kerman, Iran
| | - Seyede Elmira Yazdi Rouholamini
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Sdigheh Gharbi
- Department of Biology, Faculty of Science, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Zeinab Ansari-Asl
- Department of Chemistry, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Elham Jafari
- Pathology and Stem Cells Research Center, Kerman University of Medical Science, Kerman, Iran
| | | | - Mariam Shahrokhi-Farjah
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
12
|
Lenzi M, Cocchi V, Gasperini S, Arfè R, Marti M, Hrelia P. Evaluation of Cytotoxic and Mutagenic Effects of the Synthetic Cathinones Mexedrone, α-PVP and α-PHP. Int J Mol Sci 2021; 22:ijms22126320. [PMID: 34204826 PMCID: PMC8231654 DOI: 10.3390/ijms22126320] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 11/16/2022] Open
Abstract
Mexedrone, α-PVP and α-PHP are synthetic cathinones. They can be considered amphetamine-like substances with a stimulating effect. Actually, studies showing their impact on DNA are totally absent. Therefore, in order to fill this gap, aim of the present work was to evaluate their mutagenicity on TK6 cells. On the basis of cytotoxicity and cytostasis results, we selected the concentrations (35–100 µM) to be used in the further analysis. We used the micronucleus (MN) as indicator of genetic damage and analyzed the MNi frequency fold increase by flow cytometry. Mexedrone demonstrated its mutagenic potential contrary to the other two compounds; we then proceeded by repeating the analyzes in the presence of extrinsic metabolic activation in order to check if it was possible to totally exclude the mutagenic capacity for α-PVP and α-PHP. The results demonstrated instead the mutagenicity of their metabolites. We then evaluated reactive oxygen species (ROS) induction as a possible mechanism at the basis of the highlighted effects but the results did not show a statistically significant increase in ROS levels for any of the tested substances. Anyway, our outcomes emphasize the importance of mutagenicity evaluation for a complete assessment of the risk associated with synthetic cathinones exposure.
Collapse
Affiliation(s)
- Monia Lenzi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (M.L.); (V.C.); (S.G.); (P.H.)
| | - Veronica Cocchi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (M.L.); (V.C.); (S.G.); (P.H.)
| | - Sofia Gasperini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (M.L.); (V.C.); (S.G.); (P.H.)
| | - Raffaella Arfè
- Section of Legal Medicine and LTTA Centre, Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy;
| | - Matteo Marti
- Section of Legal Medicine and LTTA Centre, Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy;
- Collaborative Center for the Italian National Early Warning System, Department of Anti-Drug Policies, Presidency of the Council of Ministers, 44121 Ferrara, Italy
- Correspondence:
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (M.L.); (V.C.); (S.G.); (P.H.)
| |
Collapse
|
13
|
Soares J, Costa VM, Bastos MDL, Carvalho F, Capela JP. An updated review on synthetic cathinones. Arch Toxicol 2021; 95:2895-2940. [PMID: 34100120 DOI: 10.1007/s00204-021-03083-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023]
Abstract
Cathinone, the main psychoactive compound found in the plant Catha edulis Forsk. (khat), is a β-keto analogue of amphetamine, sharing not only the phenethylamine structure, but also the amphetamine-like stimulant effects. Synthetic cathinones are derivatives of the naturally occurring cathinone that largely entered the recreational drug market at the end of 2000s. The former "legal status", impressive marketing strategies and their commercial availability, either in the so-called "smartshops" or via the Internet, prompted their large spread, contributing to their increasing popularity in the following years. As their popularity increased, the risks posed for public health became clear, with several reports of intoxications and deaths involving these substances appearing both in the social media and scientific literature. The regulatory measures introduced thereafter to halt these trending drugs of abuse have proved to be of low impact, as a continuous emergence of new non-controlled derivatives keep appearing to replace those prohibited. Users resort to synthetic cathinones due to their psychostimulant properties but are often unaware of the dangers they may incur when using these substances. Therefore, studies aimed at unveiling the pharmacological and toxicological properties of these substances are imperative, as they will provide increased expertise to the clinicians that face this problem on a daily basis. The present work provides a comprehensive review on history and legal status, chemistry, pharmacokinetics, pharmacodynamics, adverse effects and lethality in humans, as well as on the current knowledge of the neurotoxic mechanisms of synthetic cathinones.
Collapse
Affiliation(s)
- Jorge Soares
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
| | - Vera Marisa Costa
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Maria de Lourdes Bastos
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - João Paulo Capela
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
- FP-ENAS (Fernando Pessoa Energy, Environment and Health Research Unit), CEBIMED (Biomedical Research Centre), Faculty of Health Sciences, University of Fernando Pessoa, Porto, Portugal.
| |
Collapse
|
14
|
Rudin D, Liechti ME, Luethi D. Molecular and clinical aspects of potential neurotoxicity induced by new psychoactive stimulants and psychedelics. Exp Neurol 2021; 343:113778. [PMID: 34090893 DOI: 10.1016/j.expneurol.2021.113778] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/25/2021] [Accepted: 05/29/2021] [Indexed: 12/20/2022]
Abstract
New psychoactive stimulants and psychedelics continue to play an important role on the illicit new psychoactive substance (NPS) market. Designer stimulants and psychedelics both affect monoaminergic systems, although by different mechanisms. Stimulant NPS primarily interact with monoamine transporters, either as inhibitors or as substrates. Psychedelic NPS most potently interact with serotonergic receptors and mediate their mind-altering effects mainly through agonism at serotonin 5-hydroxytryptamine-2A (5-HT2A) receptors. Rarely, designer stimulants and psychedelics are associated with potentially severe adverse effects. However, due to the high number of emerging NPS, it is not possible to investigate the toxicity of each individual substance in detail. The brain is an organ particularly sensitive to substance-induced toxicity due to its high metabolic activity. In fact, stimulant and psychedelic NPS have been linked to neurological and cognitive impairments. Furthermore, studies using in vitro cell models or rodents indicate a variety of mechanisms that could potentially lead to neurotoxic damage in NPS users. Cytotoxicity, mitochondrial dysfunction, and oxidative stress may potentially contribute to neurotoxicity of stimulant NPS in addition to altered neurochemistry. Serotonin 5-HT2A receptor-mediated toxicity, oxidative stress, and activation of mitochondrial apoptosis pathways could contribute to neurotoxicity of some psychedelic NPS. However, it remains unclear how well the current preclinical data of NPS-induced neurotoxicity translate to humans.
Collapse
Affiliation(s)
- Deborah Rudin
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Basel, Switzerland; Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Dino Luethi
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Basel, Switzerland; Institute of Pharmacology, Medical University of Vienna, Vienna, Austria; Institute of Applied Physics, TU Wien, Vienna, Austria.
| |
Collapse
|
15
|
Coccini T, De Simone U, Lonati D, Scaravaggi G, Marti M, Locatelli CA. MAM-2201, One of the Most Potent-Naphthoyl Indole Derivative-Synthetic Cannabinoids, Exerts Toxic Effects on Human Cell-Based Models of Neurons and Astrocytes. Neurotox Res 2021; 39:1251-1273. [PMID: 33945101 DOI: 10.1007/s12640-021-00369-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/26/2021] [Accepted: 04/25/2021] [Indexed: 01/04/2023]
Abstract
Considering the consequences on human health, in general population and workplace, associated with the use of new psychoactive substances and their continuous placing on the market, novel in vitro models for neurotoxicology research, applying human-derived CNS cells, may provide a means to understand the mechanistic basis of molecular and cellular alterations in brain. Cytotoxic effects of MAM-2201, a potent-naphthoyl indole derivative-synthetic cannabinoid, have been evaluated applying a panel of human cell-based models of neurons and astrocytes, testing different concentrations (1-30 µM) and exposure times (3-24-48 h). MAM-2201 induced toxicity in primary neuron-like cells (hNLCs), obtained from transdifferentiation of mesenchymal stem cells derived from human umbilical cord. Effects occurred in a concentration- and time-dependent manner. The lowest concentration affecting cell viability, metabolic function, apoptosis, morphology, and neuronal markers (MAP-2, NSE) was 5 μM, and even 1 μM induced apoptosis. Effects appeared early (3 h) and persisted after 24 and 48 h. Similar behavior was evidenced for human D384-astrocytes treated with MAM-2201. Differently, human SH-SY5Y-neurons, both differentiated and undifferentiated, were not sensitive to MAM-2201. On D384, the different altered endpoints were reversed, attenuated, or not antagonized by AM251 indicating that CB1 receptors may partially mediate MAM-2201-induced cytotoxicity. While in hNLCs, all toxic effects caused by MAM-2201 were apparently unrelated to CB-receptors since they were not evidenced by immunofluorescence. The present in vitro findings demonstrate the cytotoxicity of MAM-2201 on human primary neurons (hNLCs) and astrocytes cell line (D384), and support the use of these cellular models as species-specific in vitro tools suitable to clarify the neurotoxicity mechanisms of synthetic cannabinoids.
Collapse
Affiliation(s)
- T Coccini
- Laboratory of Clinical and Experimental Toxicology, and Poison Control Centre and National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 10, 27100, Pavia, Italy.
| | - U De Simone
- Laboratory of Clinical and Experimental Toxicology, and Poison Control Centre and National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 10, 27100, Pavia, Italy
| | - D Lonati
- Laboratory of Clinical and Experimental Toxicology, and Poison Control Centre and National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 10, 27100, Pavia, Italy
| | - G Scaravaggi
- Laboratory of Clinical and Experimental Toxicology, and Poison Control Centre and National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 10, 27100, Pavia, Italy
| | - M Marti
- Department of Morphology, Surgery and Experimental Medicine, Section of Legal Medicine and LTTA Center, University of Ferrara, Ferrara, Italy.,Collaborative Center for the Italian National Early Warning System, Department of Anti-Drug Policies, Presidency of the Council of Ministers, Rome, Italy
| | - C A Locatelli
- Laboratory of Clinical and Experimental Toxicology, and Poison Control Centre and National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 10, 27100, Pavia, Italy
| |
Collapse
|
16
|
Suhito IR, Angeline N, Lee KH, Kim H, Park CG, Luo Z, Kim TH. A Spheroid-Forming Hybrid Gold Nanostructure Platform That Electrochemically Detects Anticancer Effects of Curcumin in a Multicellular Brain Cancer Model. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2002436. [PMID: 32954643 DOI: 10.1002/smll.202002436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/27/2020] [Indexed: 05/26/2023]
Abstract
In this study, a multifunctional platform that enables the highly efficient formation of 3D multicellular cancer spheroids and precise real-time assessments of the anticancer effects of curcumin in a brain tumor coculture model is reported. A highly conductive gold nanostructure (HCGN) is fabricated to facilitate cancer spheroid formation without using anti-cell adhesion molecules. A neuroblastoma (SH-SY5Y) and glioblastoma (U-87MG) coculture model is generated on HCGN with a specific cell-to-cell ratio (SH-SY5Y: U-87MG = 1:1), and their redox behaviors are successfully measured without destroying the distinct 3D structure of the multicellular spheroids. Using electrochemical signals as an indicator of spheroid viability, the effects of potential anticancer compounds on cocultured spheroids are further assessed. Remarkably, decreased cell viability in 3D spheroids caused by a low concentration of curcumin (30 µM) is detectable using the electrochemical method (29.4%) but not with a conventional colorimetric assay (CCK-8). The detection is repeated more than ten times for both short- (63 h) and long-term cultivation (144 h) without damaging the spheroids, enabling real-time, non-destructive pharmacokinetic analysis of various drug candidates. Therefore, it can be concluded that the hybrid platform is a highly promising, precise, and high-throughput drug screening tool based on 3D cell cultivation.
Collapse
Affiliation(s)
- Intan Rosalina Suhito
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Novi Angeline
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Kwang-Ho Lee
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Huijung Kim
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Zhengtang Luo
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Kowloon, Hong Kong, 999077, China
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
- Integrative Research Center for Two-Dimensional Functional Materials, Institute of Interdisciplinary Convergence Research, Chung-Ang University, Seoul, 06974, Republic of Korea
| |
Collapse
|
17
|
Acute MDPV Binge Paradigm on Mice Emotional Behavior and Glial Signature. Pharmaceuticals (Basel) 2021; 14:ph14030271. [PMID: 33809599 PMCID: PMC8002122 DOI: 10.3390/ph14030271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/11/2021] [Accepted: 03/13/2021] [Indexed: 11/17/2022] Open
Abstract
3,4-Methylenedioxypyrovalerone (MDPV), a widely available synthetic cathinone, is a popular substitute for classical controlled drugs of abuse, such as methamphetamine (METH). Although MDPV poses public health risks, its neuropharmacological profile remains poorly explored. This study aimed to provide evidence on that direction. Accordingly, C57BL/6J mice were exposed to a binge MDPV or METH regimen (four intraperitoneal injections every 2 h, 10 mg/kg). Locomotor, exploratory, and emotional behavior, in addition to striatal neurotoxicity and glial signature, were assessed within 18–24 h, a known time-window encompassing classical amphetamine dopaminergic neurotoxicity. MDPV resulted in unchanged locomotor activity (open field test) and emotional behavior (elevated plus maze, splash test, tail suspension test). Additionally, striatal TH (METH neurotoxicity hallmark), Iba-1 (microglia), GFAP (astrocyte), RAGE, and TLR2/4/7 (immune modulators) protein densities remained unchanged after MDPV-exposure. Expectedly, and in sheer contrast with MDPV, METH resulted in decrease general locomotor activity paralleled by a significant striatal TH depletion, astrogliosis, and microglia arborization alterations (Sholl analysis). This comparative study newly highlights that binge MDPV-exposure comes without evident behavioral, neurochemical, and glial changes at a time-point where METH-induced striatal neurotoxicity is clearly evident. Nevertheless, neuropharmacological MDPV signature needs further profiling at different time-points, regimens, and brain regions.
Collapse
|
18
|
S-(+)-Pentedrone and R-(+)-methylone as the most oxidative and cytotoxic enantiomers to dopaminergic SH-SY5Y cells: Role of MRP1 and P-gp in cathinones enantioselectivity. Toxicol Appl Pharmacol 2021; 416:115442. [PMID: 33609514 DOI: 10.1016/j.taap.2021.115442] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/25/2021] [Accepted: 01/30/2021] [Indexed: 11/23/2022]
Abstract
Cathinone derivatives are the most representative group within new drugs market, which have been described as neurotoxic. Since cathinones, as pentedrone and methylone, are available as racemates, it is our aim to study the neuronal cytotoxicity induced by each enantiomer. Therefore, a dopaminergic SH-SY5Y cell line was used to evaluate the hypothesis of enantioselectivity of pentedrone and methylone enantiomers on cytotoxicity, oxidative stress, and membrane efflux transport (confirmed by in silico studies). Our study demonstrated enantioselectivity of these cathinones, being the S-(+)-pentedrone and R-(+)-methylone the most oxidative enantiomers and also the most cytotoxic, suggesting the oxidative stress as main cytotoxic mechanism, as previously described in in vitro studies. Additionally, the efflux transporter multidrug resistance associated protein 1 (MRP1) seems to play, together with GSH, a selective protective role against the cytotoxicity induced by R-(-)-pentedrone enantiomer. It was also observed an enantioselectivity in the binding to P-glycoprotein (P-gp), another efflux protein, being the R-(-)-pentedrone and S-(-)-methylone the most transported enantiomeric compounds. These results were confirmed, in silico, by docking studies, revealing that R-(-)-pentedrone is the enantiomer with highest affinity to MRP1 and S-(-)-methylone and R-(-)-pentedrone are the enantiomers with highest affinity to P-gp. In conclusion, our data demonstrated that pentedrone and methylone present enantioselectivity in their cytotoxicity, which seems to involve different oxidative reactivity as well as different affinity to the P-gp and MRP1 that together with GSH play a protective role.
Collapse
|
19
|
From street to lab: in vitro hepatotoxicity of buphedrone, butylone and 3,4-DMMC. Arch Toxicol 2021; 95:1443-1462. [PMID: 33550444 DOI: 10.1007/s00204-021-02990-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/21/2021] [Indexed: 12/19/2022]
Abstract
Synthetic cathinones are among the most popular new psychoactive substances, being abused for their stimulant properties, which are similar to those of amphetamine and 3,4-methylenedioxymethamphetamine (MDMA). Considering that the liver is a likely target for cathinones-induced toxicity, and for their metabolic activation/detoxification, we aimed to determine the hepatotoxicity of three commonly abused synthetic cathinones: butylone, α-methylamino-butyrophenone (buphedrone) and 3,4-dimethylmethcathinone (3,4-DMMC). We characterized their cytotoxic profile in primary rat hepatocytes (PRH) and in the HepaRG and HepG2 cell lines. PRH was the most sensitive cell model, showing the lowest EC50 values for all three substances (0.158 mM for 3,4-DMMC; 1.21 mM for butylone; 1.57 mM for buphedrone). Co-exposure of PRH to the synthetic cathinones and CYP450 inhibitors (selective and non-selective) proved that hepatic metabolism reduced the toxicity of buphedrone but increased that of butylone and 3,4-DMMC. All compounds were able to increase oxidative stress, disrupting mitochondrial homeostasis and inducing apoptotic and necrotic features, while also increasing the occurrence of acidic vesicular organelles in PRH, compatible with autophagic activation. In conclusion, butylone, buphedrone and 3,4-DMMC have hepatotoxic potential, and their toxicity lies in the interference with a number of homeostatic processes, while being influenced by their metabolic fate.
Collapse
|
20
|
Harraz MM, Guha P, Kang IG, Semenza ER, Malla AP, Song YJ, Reilly L, Treisman I, Cortés P, Coggiano MA, Veeravalli V, Rais R, Tanda G, Snyder SH. Cocaine-induced locomotor stimulation involves autophagic degradation of the dopamine transporter. Mol Psychiatry 2021; 26:370-382. [PMID: 33414501 PMCID: PMC8625012 DOI: 10.1038/s41380-020-00978-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 11/18/2020] [Accepted: 12/01/2020] [Indexed: 01/29/2023]
Abstract
Cocaine exerts its stimulant effect by inhibiting dopamine reuptake leading to increased dopamine signaling. This action is thought to reflect binding of cocaine to the dopamine transporter (DAT) to inhibit its function. However, cocaine is a relatively weak inhibitor of DAT, and many DAT inhibitors do not share the behavioral actions of cocaine. We previously showed that toxic levels of cocaine induce autophagic neuronal cell death. Here, we show that subnanomolar concentrations of cocaine elicit neural autophagy in vitro and in vivo. Autophagy inhibitors reduce the locomotor stimulant effect of cocaine in mice. Cocaine-induced autophagy degrades transporters for dopamine but not serotonin in the nucleus accumbens. Autophagy inhibition impairs cocaine conditioned place preference in mice. Our findings indicate that autophagic degradation of DAT modulates behavioral actions of cocaine.
Collapse
Affiliation(s)
- Maged M Harraz
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Prasun Guha
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - In Guk Kang
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Evan R Semenza
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Adarsha P Malla
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Young Jun Song
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Luke Reilly
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Isaac Treisman
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Pedro Cortés
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mark A Coggiano
- Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Department of Health and Human Services, Baltimore, MD, 21224, USA
| | - Vijayabhaskar Veeravalli
- Department of Neurology and Johns Hopkins Drug Discovery (JHDD) Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Rana Rais
- Department of Neurology and Johns Hopkins Drug Discovery (JHDD) Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Gianluigi Tanda
- Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Department of Health and Human Services, Baltimore, MD, 21224, USA
| | - Solomon H Snyder
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
21
|
Cocchi V, Gasperini S, Hrelia P, Tirri M, Marti M, Lenzi M. Novel Psychoactive Phenethylamines: Impact on Genetic Material. Int J Mol Sci 2020; 21:ijms21249616. [PMID: 33348640 PMCID: PMC7766159 DOI: 10.3390/ijms21249616] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/04/2020] [Accepted: 12/15/2020] [Indexed: 12/16/2022] Open
Abstract
Psychedelic and stimulating phenethylamines belong to the family of new psychoactive substances (NPS). The acute toxicity framework has begun to be investigated, while studies showing genotoxic potential are very limited or not available. Therefore, in order to fill this gap, the aim of the present work was to evaluate the genotoxicity by treating TK6 cells with 2C-H, 2C-I, 2C-B, 25B-NBOMe, and the popular 3,4-Methylenedioxymethylamphetamine (MDMA). On the basis of cytotoxicity and cytostasis results, we selected the concentrations (6.25–35 µM) to be used in genotoxicity analysis. We used the micronucleus (MN) as indicator of genetic damage and analyzed the MNi frequency fold increase by an automated flow cytometric protocol. All substances, except MDMA, resulted genotoxic; therefore, we evaluated reactive oxygen species (ROS) induction as a possible mechanism at the basis of the demonstrated genotoxicity. The obtained results showed a statistically significant increase in ROS levels for all genotoxic phenethylamines confirming this hypothesis. Our results highlight the importance of genotoxicity evaluation for a complete assessment of the risk associated also with NPS exposure. Indeed, the subjects who do not have hazardous behaviors or require hospitalization by using active but still “safe” doses could run into genotoxicity and in the well-known long-term effects associated.
Collapse
Affiliation(s)
- Veronica Cocchi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (V.C.); (S.G.); (M.L.)
| | - Sofia Gasperini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (V.C.); (S.G.); (M.L.)
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (V.C.); (S.G.); (M.L.)
- Correspondence:
| | - Micaela Tirri
- Department of Translational Medicine, Section of Legal Medicine and LTTA Center, University of Ferrara, 44121 Ferrara, Italy; (M.T.); (M.M.)
| | - Matteo Marti
- Department of Translational Medicine, Section of Legal Medicine and LTTA Center, University of Ferrara, 44121 Ferrara, Italy; (M.T.); (M.M.)
- Collaborative Center for the Italian National Early Warning System, Department of Anti-Drug Policies, Presidency of the Council of Ministers, 44121 Ferrara, Italy
| | - Monia Lenzi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (V.C.); (S.G.); (M.L.)
| |
Collapse
|
22
|
Simão AY, Gonçalves J, Gradillas A, García A, Restolho J, Fernández N, Rodilla JM, Barroso M, Duarte AP, Cristóvão AC, Gallardo E. Evaluation of the Cytotoxicity of Ayahuasca Beverages. Molecules 2020; 25:molecules25235594. [PMID: 33260723 PMCID: PMC7730595 DOI: 10.3390/molecules25235594] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
Ayahuasca is a beverage consumed at shamanic ceremonies and currently has gained popularity on recreational scenarios. It contains beta-carboline alkaloids and N,N-dimethyltryptamine, which possesses hallucinogenic effects. Only a few studies have elicited the psychoactive effects and the dose of such compounds on neurological dopaminergic cells or animals. In this work, we aimed to study the cytotoxic effects of these compounds present in ayahuasca beverages and on five different teas (Banisteriopsis caapi, Psychotria viridis, Peganum harmala, Mimosa tenuiflora and Dc Ab (commercial name)) preparations on dopaminergic immortalized cell lines. Moreover, a characterization of the derivative alkaloids was also performed. All the extracts were characterized by chromatographic systems and the effect of those compounds in cell viability and total protein levels were analyzed in N27 dopaminergic neurons cell line. This is the first article where cytotoxicity of ayahuasca tea is studied on neurological dopaminergic cells. Overall, results showed that both cell viability and protein contents decreased when cells were exposed to the individual compounds, as well as to the teas and to the two mixtures based on the traditional ayahuasca beverages.
Collapse
Affiliation(s)
- Ana Y. Simão
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.Y.S.); (J.G.); (J.R.); (A.P.D.)
- Laboratório de Fármaco-Toxicologia, UBIMedical, Universidade da Beira Interior, Estrada Municipal 506, 6200-284 Covilhã, Portugal
| | - Joana Gonçalves
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.Y.S.); (J.G.); (J.R.); (A.P.D.)
- Laboratório de Fármaco-Toxicologia, UBIMedical, Universidade da Beira Interior, Estrada Municipal 506, 6200-284 Covilhã, Portugal
| | - Ana Gradillas
- CEMBIO, Center for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Campus Monteprincipe, Boadilla del Monte, 28668 Madrid, Spain; (A.G.); (A.G.)
| | - Antonia García
- CEMBIO, Center for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Campus Monteprincipe, Boadilla del Monte, 28668 Madrid, Spain; (A.G.); (A.G.)
| | - José Restolho
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.Y.S.); (J.G.); (J.R.); (A.P.D.)
| | - Nicolás Fernández
- Cátedra de Toxicología y Química Legal, Laboratorio de Asesoramiento Toxicológico Analítico (CENATOXA), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Ciudad Autónoma de Buenos Aires (CABA), Buenos Aires C1113AAD, Argentina;
| | - Jesus M. Rodilla
- Materiais Fibrosos e Tecnologias Ambientais—FibEnTech, Departamento de Química, Universidade da Beira Interior, Rua Marquês D’Ávila e Bolama, 6201-001 Covilhã, Portugal;
| | - Mário Barroso
- Instituto Nacional de Medicina Legal e Ciências Forenses, Serviço de Química e Toxicologia Forenses, Delegação do Sul, Rua Manuel Bento de Sousa n.°3, 1169-201 Lisboa, Portugal;
| | - Ana Paula Duarte
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.Y.S.); (J.G.); (J.R.); (A.P.D.)
- Laboratório de Fármaco-Toxicologia, UBIMedical, Universidade da Beira Interior, Estrada Municipal 506, 6200-284 Covilhã, Portugal
| | - Ana C. Cristóvão
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.Y.S.); (J.G.); (J.R.); (A.P.D.)
- NEUROSOV, UBIMedical, Universidade da Beira Interior, Estrada Municipal 506, 6200-284 Covilhã, Portugal
- Correspondence: (A.C.C.); (E.G.); Tel.: +351-275-329-002/3 (A.C.C. & E.G.)
| | - Eugenia Gallardo
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.Y.S.); (J.G.); (J.R.); (A.P.D.)
- Laboratório de Fármaco-Toxicologia, UBIMedical, Universidade da Beira Interior, Estrada Municipal 506, 6200-284 Covilhã, Portugal
- Correspondence: (A.C.C.); (E.G.); Tel.: +351-275-329-002/3 (A.C.C. & E.G.)
| |
Collapse
|
23
|
In vivo toxicometabolomics reveals multi-organ and urine metabolic changes in mice upon acute exposure to human-relevant doses of 3,4-methylenedioxypyrovalerone (MDPV). Arch Toxicol 2020; 95:509-527. [PMID: 33215236 DOI: 10.1007/s00204-020-02949-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/05/2020] [Indexed: 01/08/2023]
Abstract
3,4-Methylenedioxypyrovalerone (MDPV) is consumed worldwide, despite its potential to cause toxicity in several organs and even death. There is a recognized need to clarify the biological pathways through which MDPV elicits general and target-organ toxicity. In this work, a comprehensive untargeted GC-MS-based metabolomics analysis was performed, aiming to detect metabolic changes in putative target organs (brain, heart, kidneys and liver) but also in urine of mice after acute exposure to human-relevant doses of MDPV. Male CD-1 mice received binge intraperitoneal administrations of saline or MDPV (2.5 mg/kg or 5 mg/kg) every 2 h, for a total of three injections. Twenty-four hours after the first administration, target organs, urine and blood samples were collected for metabolomics, biochemical and histological analysis. Hepatic and renal tissues of MDPV-treated mice showed moderate histopathological changes but no significant differences were found in plasma and tissue biochemical markers of organ injury. In contrast, the multivariate analysis significantly discriminated the organs and urine of MDPV-treated mice from the control (except for the lowest dose in the brain), allowing the identification of a panoply of metabolites. Those levels were significantly deviated in relation to physiological conditions and showed an organ specific response towards the drug. Kidneys and liver showed the greatest metabolic changes. Metabolites related with energetic metabolism, antioxidant defenses and inflammatory response were significantly changed in the liver of MDPV-dosed animals, while the kidneys seem to have developed an adaptive response against oxidative stress caused by MDPV. On the other hand, the dysregulation of metabolites that contribute to metabolic acidosis was also observed in this organ. The heart showed an increase of fatty acid biosynthesis, possibly as an adaptation to maintain the cardiac energy homeostasis. In the brain, changes in 3-hydroxybutyric acid levels may reflect the activation of a neurotoxic pathway. However, the increase in metabolites with neuroprotective properties seems to counteract this change. Metabolic profiling of urine from MDPV-treated mice suggested that glutathione-dependent antioxidant pathways may be particularly involved in the compensatory mechanism to counteract oxidative stress induced by MDPV. Overall, this study reports, for the first time, the metabolic profile of liver, kidneys, heart, brain, and urine of MDPV-dosed mice, providing unique insights into the biological pathways of toxicity. Our findings also underline the value of toxicometabolomics as a robust and sensitive tool for detecting adaptive/toxic cellular responses upon exposure to a physiologically relevant dose of a toxic agent, earlier than conventional toxicity tests.
Collapse
|
24
|
4′-Fluoropyrrolidinononanophenone elicits neuronal cell apoptosis through elevating production of reactive oxygen and nitrogen species. Forensic Toxicol 2020. [DOI: 10.1007/s11419-020-00550-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
25
|
Vaz I, Carvalho T, Valente MJ, Castro A, Araújo AM, Bastos ML, Carvalho M. The interplay between autophagy and apoptosis mediates toxicity triggered by synthetic cathinones in human kidney cells. Toxicol Lett 2020; 331:42-52. [PMID: 32464236 DOI: 10.1016/j.toxlet.2020.05.025] [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: 03/29/2020] [Revised: 05/06/2020] [Accepted: 05/21/2020] [Indexed: 12/21/2022]
Abstract
Synthetic cathinones abuse remains a serious public health problem. Kidney injury has been reported in intoxications associated with synthetic cathinones, but the molecular mechanisms involved have not been explored yet. In this study, the potential in vitro nephrotoxic effects of four commonly abused cathinone derivatives, namely pentedrone, 3,4-dimethylmethcatinone (3,4-DMMC), methylone and 3,4-methylenedioxypyrovalerone (MDPV), were assessed in the human kidney HK-2 cell line. All four derivatives elicited cell death in a concentration- and time-dependent manner, in the following order of potency: 3,4-DMMC >> MDPV > methylone ≈ pentedrone. 3,4-DMMC and methylone were selected to further elucidate the mechanisms behind synthetic cathinones-induced cell death. Both drugs elicited apoptotic cell death and prompted the formation of acidic vesicular organelles and autophagosomes in HK-2 cells. Moreover, the autophagy inhibitor 3-methyladenine significantly potentiated cell death, indicating that autophagy may serve as a cell survival mechanism that protects renal cells against synthetic cathinones toxicity. Both drugs triggered a rise in reactive oxygen and nitrogen species formation, which was completely prevented by antioxidant treatment with N‑acetyl‑L‑cysteine or ascorbic acid. Importantly, these antioxidant agents significantly aggravated renal cell death induced by cathinone derivatives, most likely due to their autophagy-blocking properties. Taken together, our results support an intricate control of cell survival/death modulated by oxidative stress, apoptosis and autophagy in synthetic cathinones-induced renal injury.
Collapse
Affiliation(s)
- I Vaz
- UFP Energy, Environment and Health Research Unit (FP-ENAS), University Fernando Pessoa, Praça Nove de Abril, 349, 4249-004, Porto, Portugal
| | - T Carvalho
- UFP Energy, Environment and Health Research Unit (FP-ENAS), University Fernando Pessoa, Praça Nove de Abril, 349, 4249-004, Porto, Portugal
| | - M J Valente
- UCIBIO, REQUIMTE, Laboratory of Biochemistry, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - A Castro
- UFP Energy, Environment and Health Research Unit (FP-ENAS), University Fernando Pessoa, Praça Nove de Abril, 349, 4249-004, Porto, Portugal
| | - A M Araújo
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - M L Bastos
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - M Carvalho
- UFP Energy, Environment and Health Research Unit (FP-ENAS), University Fernando Pessoa, Praça Nove de Abril, 349, 4249-004, Porto, Portugal; UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
26
|
Soares J, Costa VM, Gaspar H, Santos S, Bastos MDL, Carvalho F, Capela JP. Adverse outcome pathways induced by 3,4-dimethylmethcathinone and 4-methylmethcathinone in differentiated human SH-SY5Y neuronal cells. Arch Toxicol 2020; 94:2481-2503. [PMID: 32382956 DOI: 10.1007/s00204-020-02761-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/22/2020] [Indexed: 12/27/2022]
Abstract
Cathinones (β-keto amphetamines), widely abused in recreational settings, have been shown similar or even worse toxicological profile than classical amphetamines. In the present study, the cytotoxicity of two β-keto amphetamines [3,4-dimethylmethcathinone (3,4-DMMC) and 4-methylmethcathinone (4-MMC)], was evaluated in differentiated dopaminergic SH-SY5Y cells in comparison to methamphetamine (METH). MTT reduction and NR uptake assays revealed that both cathinones and METH induced cytotoxicity in a concentration- and time-dependent manner. Pre-treatment with trolox (antioxidant) partially prevented the cytotoxicity induced by all tested drugs, while N-acetyl-L-cysteine (NAC; antioxidant and glutathione precursor) and GBR 12909 (dopamine transporter inhibitor) partially prevented the cytotoxicity induced by cathinones, as evaluated by the MTT reduction assay. Unlike METH, cathinones induced oxidative stress evidenced by the increase on intracellular levels of reactive oxygen species (ROS), and also by the decrease of intracellular glutathione levels. Trolox prevented, partially but significantly, the ROS generation elicited by cathinones, while NAC inhibited it completely. All tested drugs induced mitochondrial dysfunction, since they led to mitochondrial membrane depolarization and to intracellular ATP depletion. Activation of caspase-3, indicative of apoptosis, was seen both for cathinones and METH, and confirmed by annexin V and propidium iodide positive staining. Autophagy was also activated by all drugs tested. Pre-incubation with bafilomycin A1, an inhibitor of the vacuolar H+-ATPase, only protected against the cytotoxicity induced by METH, which indicates dissimilar toxicological pathways for the tested drugs. In conclusion, the mitochondrial impairment and oxidative stress observed for the tested cathinones may be key factors for their neurotoxicity, but different outcome pathways seem to be involved in the adverse effects, when compared to METH.
Collapse
Affiliation(s)
- Jorge Soares
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
| | - Vera Marisa Costa
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Helena Gaspar
- BioISI - Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
- MARE - Marine and Environmental Sciences Centre, Polytechnic of Leiria, Peniche, Portugal
| | - Susana Santos
- Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Maria de Lourdes Bastos
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - João Paulo Capela
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
- FP-ENAS (Fernando Pessoa Energy, Environment and Health Research Unit), CEBIMED (Biomedical Research Centre), Faculty of Health Sciences, University of Fernando Pessoa, Porto, Portugal.
| |
Collapse
|
27
|
Zhou X, Bouitbir J, Liechti ME, Krähenbühl S, Mancuso RV. Hyperthermia Increases Neurotoxicity Associated with Novel Methcathinones. Cells 2020; 9:cells9040965. [PMID: 32295288 PMCID: PMC7227000 DOI: 10.3390/cells9040965] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/29/2020] [Accepted: 04/09/2020] [Indexed: 12/16/2022] Open
Abstract
Hyperthermia is one of the severe acute adverse effects that can be caused by the ingestion of recreational drugs, such as methcathinones. The effect of hyperthermia on neurotoxicity is currently not known. The primary aim of our study was therefore to investigate the effects of hyperthermia (40.5 °C) on the neurotoxicity of methcathinone (MC), 4-chloromethcathinone (4-CMC), and 4-methylmethcathinone (4-MMC) in SH-SY5Y cells. We found that 4-CMC and 4-MMC were cytotoxic (decrease in cellular ATP and plasma membrane damage) under both hyper- (40.5 °C) and normothermic conditions (37 °C), whereby cells were more sensitive to the toxicants at 40.5 °C. 4-CMC and 4-MMC impaired the function of the mitochondrial electron transport chain and increased mitochondrial formation of reactive oxygen species (ROS) in SH-SY5Y cells, which were accentuated under hyperthermic conditions. Hyperthermia was associated with a rapid expression of the 70 kilodalton heat shock protein (Hsp70), which partially prevented cell death after 6 h of exposure to the toxicants. After 24 h of exposure, autophagy was stimulated by the toxicants and by hyperthermia but could only partially prevent cell death. In conclusion, hyperthermic conditions increased the neurotoxic properties of methcathinones despite the stimulation of protective mechanisms. These findings may be important for the understanding of the mechanisms and clinical consequences of the neurotoxicity associated with these compounds.
Collapse
Affiliation(s)
- Xun Zhou
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, 4031 Basel, Switzerland; (X.Z.); (J.B.); (M.E.L.); (R.V.M.)
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Jamal Bouitbir
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, 4031 Basel, Switzerland; (X.Z.); (J.B.); (M.E.L.); (R.V.M.)
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
- Swiss Centre for Applied Human Toxicology, 4031 Basel, Switzerland
| | - Matthias E. Liechti
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, 4031 Basel, Switzerland; (X.Z.); (J.B.); (M.E.L.); (R.V.M.)
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Stephan Krähenbühl
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, 4031 Basel, Switzerland; (X.Z.); (J.B.); (M.E.L.); (R.V.M.)
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
- Swiss Centre for Applied Human Toxicology, 4031 Basel, Switzerland
- Correspondence: ; Tel.: +41-61-265-4715
| | - Riccardo V. Mancuso
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, 4031 Basel, Switzerland; (X.Z.); (J.B.); (M.E.L.); (R.V.M.)
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| |
Collapse
|
28
|
Luethi D, Liechti ME. Designer drugs: mechanism of action and adverse effects. Arch Toxicol 2020; 94:1085-1133. [PMID: 32249347 PMCID: PMC7225206 DOI: 10.1007/s00204-020-02693-7] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 02/25/2020] [Indexed: 12/18/2022]
Abstract
Psychoactive substances with chemical structures or pharmacological profiles that are similar to traditional drugs of abuse continue to emerge on the recreational drug market. Internet vendors may at least temporarily sell these so-called designer drugs without adhering to legal statutes or facing legal consequences. Overall, the mechanism of action and adverse effects of designer drugs are similar to traditional drugs of abuse. Stimulants, such as amphetamines and cathinones, primarily interact with monoamine transporters and mostly induce sympathomimetic adverse effects. Agonism at μ-opioid receptors and γ-aminobutyric acid-A (GABAA) or GABAB receptors mediates the pharmacological effects of sedatives, which may induce cardiorespiratory depression. Dissociative designer drugs primarily act as N-methyl-D-aspartate receptor antagonists and pose similar health risks as the medically approved dissociative anesthetic ketamine. The cannabinoid type 1 (CB1) receptor is thought to drive the psychoactive effects of synthetic cannabinoids, which are associated with a less desirable effect profile and more severe adverse effects compared with cannabis. Serotonergic 5-hydroxytryptamine-2A (5-HT2A) receptors mediate alterations of perception and cognition that are induced by serotonergic psychedelics. Because of their novelty, designer drugs may remain undetected by routine drug screening, thus hampering evaluations of adverse effects. Intoxication reports suggest that several designer drugs are used concurrently, posing a high risk for severe adverse effects and even death.
Collapse
Affiliation(s)
- Dino Luethi
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Währinger Strasse 13a, 1090, Vienna, Austria.
- Institute of Applied Physics, Vienna University of Technology, Getreidemarkt 9, 1060, Vienna, Austria.
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Schanzenstrasse 55, 4056, Basel, Switzerland.
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Schanzenstrasse 55, 4056, Basel, Switzerland.
| |
Collapse
|
29
|
Li IH, Shih JH, Yeh TY, Lin HC, Chen MH, Huang YS. Lysosomal Dysfunction and Autophagy Blockade Contribute to MDMA-Induced Neurotoxicity in SH-SY5Y Neuroblastoma Cells. Chem Res Toxicol 2020; 33:903-914. [PMID: 32186374 DOI: 10.1021/acs.chemrestox.9b00437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Methylenedioxymethamphetamine (MDMA) is a psychostimulant with high abuse potential and severe neurotoxicity. According to our previous study, MDMA promotes autophagosome accumulation and contributes to cell death in cultured cortical and serotonergic neurons. However, the detailed mechanism underlying autophagy dysfunction remains unclear. Lysosomes play an important role in autophagic degradation. The present study aimed to examine the role of lysosomal function in autophagic flux in neuronal cultures exposed to MDMA. We showed that MDMA induced enlarged vesicles that accumulate in SH-SY5Y neuroblastoma cells. In addition, we demonstrated that MDMA stimulated dynamin-dependent but clathrin-independent endocytosis, which might contribute to vacuole expansion. Morphological and Western blot analyses revealed that MDMA induced lysosomal swelling, whereas the activity of the lysosomal hydrolytic enzymes cathepsin B and cathepsin D was decreased in SH-SY5Y and cultured cortical neurons, which might lead to autophagosome accumulation and autophagic degradation blockage. Intriguingly, inactivation of cathepsins B and D led to cell death and autophagy-lysosomal dysregulation, which mimicked MDMA-induced neurotoxicity. Consequently, impairment of lysosomal proteolysis and blockage of autophagy degradation contributed to MDMA-induced neurotoxicity in neuronal cultures.
Collapse
Affiliation(s)
- I-Hsun Li
- Department of Pharmacy Practice, Tri-Service General Hospital, Taipei 114, Taiwan.,School of Pharmacy, National Defense Medical Center, Taipei 114, Taiwan
| | - Jui-Hu Shih
- Department of Pharmacy Practice, Tri-Service General Hospital, Taipei 114, Taiwan.,School of Pharmacy, National Defense Medical Center, Taipei 114, Taiwan
| | - Ting-Yin Yeh
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 114, Taiwan
| | - Hung-Che Lin
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Ming-Hua Chen
- Division of Neurology, Department of Medicine, Armed Forces Taoyuan General Hospital, Taoyuan 325, Taiwan.,Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yuahn-Sieh Huang
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 114, Taiwan
| |
Collapse
|
30
|
Leong HS, Philp M, Simone M, Witting PK, Fu S. Synthetic Cathinones Induce Cell Death in Dopaminergic SH-SY5Y Cells via Stimulating Mitochondrial Dysfunction. Int J Mol Sci 2020; 21:ijms21041370. [PMID: 32085614 PMCID: PMC7073199 DOI: 10.3390/ijms21041370] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/13/2020] [Accepted: 02/16/2020] [Indexed: 01/22/2023] Open
Abstract
Increasing reports of neurological and psychiatric complications due to psychostimulant synthetic cathinones (SCs) have recently raised public concern. However, the precise mechanism of SC toxicity is unclear. This paucity of understanding highlights the need to investigate the in-vitro toxicity and mechanistic pathways of three SCs: butylone, pentylone, and 3,4-Methylenedioxypyrovalerone (MDPV). Human neuronal cells of SH-SY5Y were cultured in supplemented DMEM/F12 media and differentiated to a neuronal phenotype using retinoic acid (10 μM) and 12-O-tetradecanoylphorbol-13-acetate (81 nM). Trypan blue and lactate dehydrogenase assays were utilized to assess the neurotoxicity potential and potency of these three SCs. To investigate the underlying neurotoxicity mechanisms, measurements included markers of oxidative stress, mitochondrial bioenergetics, and intracellular calcium (Ca2+), and cell death pathways were evaluated at two doses (EC15 and EC40), for each drug tested. Following 24 h of treatment, all three SCs exhibited a dose-dependent neurotoxicity, characterized by a significant (p < 0.0001 vs. control) production of reactive oxygen species, decreased mitochondrial bioenergetics, and increased intracellular Ca2+ concentrations. The activation of caspases 3 and 7 implicated the orchestration of mitochondrial-mediated neurotoxicity mechanisms for these SCs. Identifying novel therapeutic agents to enhance an altered mitochondrial function may help in the treatment of acute-neurological complications arising from the illicit use of these SCs.
Collapse
Affiliation(s)
- Huey Sze Leong
- Centre for Forensic Science, School of Mathematical and Physical Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (H.S.L.); (M.P.)
- Discipline of Pathology, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
| | - Morgan Philp
- Centre for Forensic Science, School of Mathematical and Physical Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (H.S.L.); (M.P.)
| | - Martin Simone
- Discipline of Pathology, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
| | - Paul Kenneth Witting
- Discipline of Pathology, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
- Correspondence: (P.K.W.); (S.F.); Tel.: +61-2-9114-0524 (P.K.W.); +61-2-9514-8207 (S.F.)
| | - Shanlin Fu
- Centre for Forensic Science, School of Mathematical and Physical Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (H.S.L.); (M.P.)
- Correspondence: (P.K.W.); (S.F.); Tel.: +61-2-9114-0524 (P.K.W.); +61-2-9514-8207 (S.F.)
| |
Collapse
|
31
|
Emerging club drugs: 5-(2-aminopropyl)benzofuran (5-APB) is more toxic than its isomer 6-(2-aminopropyl)benzofuran (6-APB) in hepatocyte cellular models. Arch Toxicol 2019; 94:609-629. [PMID: 31838565 DOI: 10.1007/s00204-019-02638-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/26/2019] [Indexed: 12/15/2022]
Abstract
New phenylethylamine derivatives are among the most commonly abused new psychoactive substances. They are synthesized and marketed in lieu of classical amphetaminic stimulants, with no previous safety testing. Our study aimed to determine the in vitro hepatotoxicity of two benzofurans [6-(2-aminopropyl)benzofuran (6-APB) and 5-(2-aminopropyl)benzofuran (5-APB)] that have been misused as 'legal highs'. Cellular viability was assessed through the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction assay, following 24-h drug exposure of human hepatoma HepaRG cells (EC50 2.62 mM 5-APB; 6.02 mM 6-APB), HepG2 cells (EC50 3.79 mM 5-APB; 8.18 mM 6-APB) and primary rat hepatocytes (EC50 964 μM 5-APB; 1.94 mM 6-APB). Co-incubation of primary hepatocytes, the most sensitive in vitro model, with CYP450 inhibitors revealed a role of metabolism, in particular by CYP3A4, in the toxic effects of both benzofurans. Also, 6-APB and 5-APB concentration-dependently enhanced oxidative stress (significantly increased reactive species and oxidized glutathione, and decreased reduced glutathione levels) and unsettled mitochondrial homeostasis, with disruption of mitochondrial membrane potential and decline of intracellular ATP. Evaluation of cell death mechanisms showed increased caspase-8, -9, and -3 activation, and nuclear morphological changes consistent with apoptosis; at concentrations higher than 2 mM, however, necrosis prevailed. Concentration-dependent formation of acidic vesicular organelles typical of autophagy was also observed for both drugs. Overall, 5-APB displayed higher hepatotoxicity than its 6-isomer. Our findings provide new insights into the potential hepatotoxicity of these so-called 'safe drugs' and highlight the putative risks associated with their use as psychostimulants.
Collapse
|
32
|
Soares J, Costa VM, Gaspar H, Santos S, de Lourdes Bastos M, Carvalho F, Capela JP. Structure-cytotoxicity relationship profile of 13 synthetic cathinones in differentiated human SH-SY5Y neuronal cells. Neurotoxicology 2019; 75:158-173. [DOI: 10.1016/j.neuro.2019.08.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 07/30/2019] [Accepted: 08/21/2019] [Indexed: 10/26/2022]
|
33
|
Gonçalves JL, Alves VL, Aguiar J, Teixeira HM, Câmara JS. Synthetic cathinones: an evolving class of new psychoactive substances. Crit Rev Toxicol 2019; 49:549-566. [PMID: 31747318 DOI: 10.1080/10408444.2019.1679087] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Synthetic cathinones (SCat) are amphetamine-like psychostimulants that emerged onto drug markets as "legal" alternatives to illicit drugs such as ecstasy, cocaine, and amphetamines. Usually they are sold as "bath salts," "plant food," or "research chemicals," and rapidly gained popularity amongst drugs users due to their potency, low cost, and availability. In addition, internet drug sales have been replacing the old way of supplying drugs of abuse, contributing to their rapid spread. Despite the legislative efforts to control SCat, new derivatives continue to emerge on the recreational drugs market and their abuse still represents a serious public health issue. To date, about 150 SCat have been identified on the clandestine drugs market, which are one of the largest groups of new psychoactive substances (NPS) monitored by the United Nations Office on Drugs and Crime and the European Monitoring Center for Drugs and Drug Addiction. Similar to the classical stimulants, SCat affect the levels of catecholamines in the central nervous system, which results in their psychological, behavioral and toxic effects. Generally, the effects of SCat greatly differ from drug to drug and relatively little information is available about their pharmacology. The present work provides a review on the development of SCat as substances of abuse, current patterns of abuse and their legal status, chemical classification, known mechanisms of action, and their toxicological effects.
Collapse
Affiliation(s)
- João L Gonçalves
- CQM - Centro de Química da Madeira, Universidade da Madeira, Funchal, Portugal
| | - Vera L Alves
- CQM - Centro de Química da Madeira, Universidade da Madeira, Funchal, Portugal
| | - Joselin Aguiar
- CQM - Centro de Química da Madeira, Universidade da Madeira, Funchal, Portugal
| | - Helena M Teixeira
- Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal.,Instituto Nacional de Medicina Legal e Ciências Forenses, IP, Delegação Centro, Coimbra, Portugal
| | - José S Câmara
- CQM - Centro de Química da Madeira, Universidade da Madeira, Funchal, Portugal.,Faculdade de Ciências Exactas e da Engenharia, Universidade da Madeira, Funchal, Portugal
| |
Collapse
|
34
|
Effect of sterilization methods on electrospun cellulose acetate butyrate nanofibers for SH-SY5Y cultivation. REACT FUNCT POLYM 2019. [DOI: 10.1016/j.reactfunctpolym.2019.104339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
35
|
The new psychoactive substance 3-methylmethcathinone (3-MMC or metaphedrone) induces oxidative stress, apoptosis, and autophagy in primary rat hepatocytes at human-relevant concentrations. Arch Toxicol 2019; 93:2617-2634. [PMID: 31468101 DOI: 10.1007/s00204-019-02539-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
Abstract
3-Methylmethcathinone (3-MMC or metaphedrone) has become one of the most popular recreational drugs worldwide after the ban of mephedrone, and was recently deemed responsible for several intoxications and deaths. This study aimed at assessing the hepatotoxicity of 3-MMC. For this purpose, Wistar rat hepatocytes were isolated by collagenase perfusion, cultured and exposed for 24 h at a concentration range varying from 31 nM to 10 mM 3-MMC. The modulatory effects of cytochrome P450 (CYP) inhibitors on 3-MMC hepatotoxicity were evaluated. 3-MMC-induced toxicity was perceived at the lysosome at lower concentrations (NOEC 312.5 µM), compared to mitochondria (NOEC 379.5 µM) and cytoplasmic membrane (NOEC 1.04 mM). Inhibition of CYP2D6 and CYP2E1 diminished 3-MMC cytotoxicity, yet for CYP2E1 inhibition this effect was only observed for concentrations up to 1.3 mM. A significant concentration-dependent increase of intracellular reactive species was observed from 10 μM 3-MMC on; a concentration-dependent decrease in antioxidant glutathione defences was also observed. At 10 μM, caspase-3, caspase-8, and caspase-9 activities were significantly elevated, corroborating the activation of both intrinsic and extrinsic apoptosis pathways. Nuclear morphology and formation of cytoplasmic acidic vacuoles suggest prevalence of necrosis and autophagy at concentrations higher than 10 μM. No significant alterations were observed in the mitochondrial membrane potential, but intracellular ATP significantly decreased at 100 μM. Our data point to a role of metabolism in the hepatotoxicity of 3-MMC, which seems to be triggered both by autophagic and apoptotic/necrotic mechanisms. This work is the first approach to better understand 3-MMC toxicology.
Collapse
|
36
|
Bernstein DL, Nayak SU, Oliver CF, Rawls SM, Rom S. Methylenedioxypyrovalerone (MDPV) impairs working memory and alters patterns of dopamine signaling in mesocorticolimbic substrates. Neurosci Res 2019; 155:56-62. [PMID: 31302200 DOI: 10.1016/j.neures.2019.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 06/30/2019] [Accepted: 07/10/2019] [Indexed: 02/06/2023]
Abstract
Knowledge remains limited about how chronic cathinone exposure impacts dopamine systems in brain reward circuits. In the present study, a binge-like MDPV exposure that impaired novel object recognition (NOR) dysregulated dopamine markers in mesocorticolimbic substrates of rats, with especially profound effects on D1 and D2 receptor's and VMAT gene expression. Our data suggested that dopamine receptivity was reduced in the NAc but increased in the PFC and dopamine-producing VTA. The MDPV-induced impairment of NOR was prevented by a D1 receptor antagonist, suggesting that chronic MDPV exposure produces site-specific dysregulation of dopamine markers in the mesocorticolimbic circuit and memory deficits in the NOR test that are influenced by D1 receptors.
Collapse
Affiliation(s)
- David L Bernstein
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Sunyl U Nayak
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Chicora F Oliver
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Scott M Rawls
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA; Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Slava Rom
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA; Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.
| |
Collapse
|
37
|
Sevoflurane induces cognitive impairment in young mice via autophagy. PLoS One 2019; 14:e0216372. [PMID: 31107909 PMCID: PMC6527218 DOI: 10.1371/journal.pone.0216372] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/18/2019] [Indexed: 11/19/2022] Open
Abstract
Background Anesthesia may induce neurotoxicity and neurocognitive impairment in young mice. However, the underlying mechanism remains largely to be determined. Meanwhile, autophagy is involved in brain development and contributes to neurodegenerative diseases. We, therefore, set out to determine the effects of sevoflurane on autophagy in the hippocampus of young mice and on cognitive function in the mice. Methods Six day-old mice received 3% sevoflurane, for two hours daily, on postnatal days (P) 6, 7 and 8. We then decapitated the mice and harvested the hippocampus of the young mice at P8. The level of LC3, the ratio of LC3-II to LC3-I, and SQSTM1/p62 level associated with the autophagy in the hippocampus of the mice were assessed by using Western blotting. We used different groups of mice for behavioral testing via the Morris Water Maze from P31 to P37. Results The anesthetic sevoflurane increased the level of LC3-II and ratio of LC3-II/LC3-I, decreased the p62 level in the hippocampus of the young mice, and induced cognitive impairment in the mice. 3-Methyladenine, the inhibitor of autophagy, attenuated the activation of autophagy and ameliorated the cognitive impairment induced by sevoflurane in the young mice. Conclusion These data showed that sevoflurane anesthesia might induce cognitive impairment in the young mice via activation of autophagy in the hippocampus of the young mice. These findings from the proof of concept studies have established a system and suggest the role of autophagy in anesthesia neurotoxicity and cognitive impairment in the young mice, pending further investigation.
Collapse
|
38
|
Molecular Toxicological Mechanisms of Synthetic Cathinones on C2C12 Myoblasts. Int J Mol Sci 2019; 20:ijms20071561. [PMID: 30925718 PMCID: PMC6479684 DOI: 10.3390/ijms20071561] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/22/2019] [Accepted: 03/22/2019] [Indexed: 12/22/2022] Open
Abstract
Synthetic cathinones are popular psychoactive substances that may cause skeletal muscle damage. In addition to indirect sympathomimetic myotoxicity, these substances could be directly myotoxic. Since studies in myocytes are currently lacking, the aim of the present study was to investigate potential toxicological effects by synthetic cathinones on C2C12 myoblasts (mouse skeletal muscle cell line). We exposed C2C12 myoblasts to 3-methylmethcathinone, 4-methylmethcathinone (mephedrone), 3,4-methylenedioxymethcathinone (methylone), 3,4-methylenedioxypyrovalerone (MDPV), alpha-pyrrolidinovalerophenone (α-PVP), and naphthylpyrovalerone (naphyrone) for 1 or 24 h before cell membrane integrity, ATP content, mitochondrial oxygen consumption, and mitochondrial superoxide production was measured. 3,4-Methylenedioxymethamphetamine (MDMA) was included as a reference compound. All investigated synthetic cathinones, as well as MDMA, impaired cell membrane integrity, depleted ATP levels, and increased mitochondrial superoxide concentrations in a concentration-dependent manner in the range of 50–2000 μM. The two pyrovalerone derivatives α-PVP and naphyrone, and MDMA, additionally impaired basal and maximal cellular respiration, suggesting mitochondrial dysfunction. Alpha-PVP inhibited complex I, naphyrone complex II, and MDMA complex I and III, whereas complex IV was not affected. We conclude that, in addition to sympathetic nervous system effects and strenuous muscle exercise, direct effects of some cathinones on skeletal muscle mitochondria may contribute to myotoxicity in susceptible synthetic cathinone drugs users.
Collapse
|
39
|
Leyrer-Jackson JM, Nagy EK, Olive MF. Cognitive deficits and neurotoxicity induced by synthetic cathinones: is there a role for neuroinflammation? Psychopharmacology (Berl) 2019; 236:1079-1095. [PMID: 30368582 PMCID: PMC6486871 DOI: 10.1007/s00213-018-5067-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/03/2018] [Indexed: 02/06/2023]
Abstract
RATIONALE The number of synthetic derivatives of cathinone, the primary psychoactive alkaloid found in Catha edulis (khat), has risen exponentially in the past decade. Synthetic cathinones (frequently referred to as "bath salts") produce adverse cognitive and behavioral sequelae, share similar pharmacological mechanisms of action with traditional psychostimulants, and may therefore trigger similar cellular events that give rise to neuroinflammation and neurotoxicity. OBJECTIVES In this review, we provide a brief overview of synthetic cathinones, followed by a summary of cognitive deficits in animals and humans that have been documented following acute or repeated exposure. We also summarize growing evidence from in vitro and in vivo studies for synthetic cathinone-induced neurotoxicity, and provide a working hypothetic model of potential cellular mechanisms. RESULTS Synthetic cathinones produce varying effects on markers of monoaminergic terminal function and can increase the formation of reactive oxygen and nitrogen species, induce apoptotic signaling, and cause neurodegeneration and cytotoxicity. We hypothesize that these effects result from biochemical events similar to those induced by traditional psychostimulants. However, empirical evidence for the ability of synthetic cathinones to induce neuroinflammatory processes is currently lacking. CONCLUSIONS Like their traditional psychostimulant counterparts, synthetic cathinones appear to induce neurocognitive dysfunction and cytotoxicity, which are dependent on drug type, dose, frequency, and time following exposure. However, additional studies on synthetic cathinone-induced neuroinflammation are clearly needed, as are investigations into the neurochemical and neuroimmune mechanisms underlying their neurotoxic effects. Such endeavors may lead to novel therapeutic avenues to promote recovery in habitual synthetic cathinone users.
Collapse
Affiliation(s)
| | | | - M. Foster Olive
- Correspondence to: M. Foster Olive, Ph.D. Department of Psychology, Arizona State University, 950 S. McAllister Ave. Tempe, AZ 85287 USA, Phone 1-480-727-9557, Fax 1-480-965-8544,
| |
Collapse
|
40
|
Methcathinone and 3-Fluoromethcathinone Stimulate Spontaneous Horizontal Locomotor Activity in Mice and Elevate Extracellular Dopamine and Serotonin Levels in the Mouse Striatum. Neurotox Res 2018; 35:594-605. [PMID: 30377956 PMCID: PMC6420425 DOI: 10.1007/s12640-018-9973-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/07/2018] [Accepted: 10/19/2018] [Indexed: 12/23/2022]
Abstract
Methcathinone (MC) and 3-fluoromethcathinone (3-FMC) are well-known members of the synthetic cathinone derivatives, the second most abused group of novel psychoactive substances (NPS). They are considered as methamphetamine-like cathinones, as they elicit their psychostimulatory effects via inhibition of monoamine uptake and enhanced release. The present study examines the effects of MC and 3-FMC on the spontaneous locomotor activity of mice and extracellular levels of dopamine and serotonin in the mouse striatum. Both MC and 3-FMC produced a dose-dependent increase of horizontal locomotor activity, but no significant changes in rearing behavior were observed. The locomotor stimulation induced by MC and 3-FMC is mediated by activation of dopaminergic neurotransmission, as selective D1-dopamine receptor antagonist, SCH 23390, abolished the effects of both drugs. In line with pharmacological data obtained by previous in vitro studies, MC and 3-FMC produced potent increases of extracellular dopamine and serotonin levels in the mouse striatum. Taken together, results presented within this study confirm previous findings and expand our knowledge on the pharmacology of MC and 3-FMC along with their behavioral effects.
Collapse
|
41
|
Silva B, Pereira JA, Cravo S, Araújo AM, Fernandes C, Pinto MMM, de Pinho PG, Remião F. Multi-milligram resolution and determination of absolute configuration of pentedrone and methylone enantiomers. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1100-1101:158-164. [PMID: 30336346 DOI: 10.1016/j.jchromb.2018.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/01/2018] [Accepted: 10/04/2018] [Indexed: 12/14/2022]
Abstract
The enantioresolution of pentedrone and methylone was carried out at a multi-milligram scale by liquid chromatography on a Chiralpak AS® stationary phase. The excellent enantioresolution using this column allowed to collect highly pure enantiomeric fractions, achieving enantiomeric ratios higher than 98%. An overall recovery of 72% was achieved for pentedrone enantiomers and 80% for methylone. Furthermore, the absolute configuration of the enantiomers of both cathinones was determined for the first time by electronic circular dichroism (ECD) spectroscopy, with the aid of theoretical calculations, as (+)‑(S) and (-)‑(R)-pentedrone, and (-)‑(S) and (+)‑(R)‑methylone.
Collapse
Affiliation(s)
- Bárbara Silva
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - José A Pereira
- ICBAS, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Avenida General Norton de Matos, 4450-208 Matosinhos, Portugal
| | - Sara Cravo
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Avenida General Norton de Matos, 4450-208 Matosinhos, Portugal
| | - Ana Margarida Araújo
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Carla Fernandes
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Avenida General Norton de Matos, 4450-208 Matosinhos, Portugal.
| | - Madalena M M Pinto
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Avenida General Norton de Matos, 4450-208 Matosinhos, Portugal
| | - Paula Guedes de Pinho
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Fernando Remião
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| |
Collapse
|
42
|
Abstract
INTRODUCTION The second largest group of new drugs monitored by the European Monitoring Centre for Drugs and Drug Addiction (EMCDDA) is synthetic cathinones. Substances that are controlled by the law are immediately replaced by new uncontrolled derivatives that cause constant and dynamic changes on the drug market. Some of the most recent synthetic cathinones that have appeared on the "legal highs" market are 3,4-methylenedioxy-α-pyrrolidinohexanophenone (3,4-MDPHP) and α-pyrrolidinohexanophenone (α-PHP). CASE HISTORY A 21-year-old woman in the 36th week of pregnancy presented with psychomotor agitation. Fetal demise was demonstrated and a caesarean delivery performed. METHODS The analyses were carried out by liquid chromatography with mass spectrometry (LC-MS/MS). The analytes were isolated from the biological material by liquid-liquid extraction with n-butyl chloride. RESULTS 3,4-MDPHP and α-PHP were detected and quantified in both the fetus' and the mothers blood, as well as in the mothers urine samples. The determined concentrations of 3,4-MDPHP and α-PHP were, 76 ng/mL and 12 ng/mL in the fetal blood sample, 16 ng/mL and traces in the mothers blood, and 697 mg/mL and 136 ng/mL in the mothers urine, respectively. DISCUSSION The presented case demonstrates that 3,4-MDPHP and α-PHP transfers from maternal blood to fetal blood. Blood concentrations of these compounds were higher in the fetus than in the mother. Based on the known effects of these substances and the patient's presentation and clinical course, it would seem that these substances contributed to the fetal death. CONCLUSIONS The detected substances transfer from maternal to fetal circulation, and synthetic cathinone blood concentration can be higher in the fetus than in the mother. This along with the fact immature metabolic ability makes a fetus more vulnerable to cathinones intoxication than adults.
Collapse
Affiliation(s)
- Piotr Adamowicz
- a Department of Forensic Toxicology , Institute of Forensic Research , Kraków , Poland
| | - Piotr Hydzik
- b Department of Clinical Toxicology , Jagiellonian University Medical College , Kraków , Poland
| |
Collapse
|
43
|
Wojcieszak J, Andrzejczak D, Kedzierska M, Milowska K, Zawilska JB. Cytotoxicity of α-Pyrrolidinophenones: an Impact of α-Aliphatic Side-chain Length and Changes in the Plasma Membrane Fluidity. Neurotox Res 2018; 34:613-626. [PMID: 29951896 PMCID: PMC6154177 DOI: 10.1007/s12640-018-9923-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/21/2018] [Accepted: 06/07/2018] [Indexed: 11/29/2022]
Abstract
Pyrovalerone derivatives (α-pyrrolidinophenones) form a branch of synthetic cathinones, a second most prominent group of novel psychoactive substances. Although the toxicity of 3,4-MDPV, a progenitor of the α-pyrrolidinophenones, is well described, little is known of the potential cytotoxicity of the new members of this group entering the recreational drug market each year. The present study assesses the cytotoxicity of members of the α-pyrrolidinophenone group, i.e., α-PVP, its longer side-chain derivatives PV8 and PV9, and their 4-fluoro- and 4-methoxy-analogs, against model cell lines for the nervous system (SH-SY5Y), liver (Hep G2) and upper airway epithelium (RPMI 2650), and cardiomyocytes (H9C2(2-1)). Additionally, an impact of pyrovalerones on the fluidity of the plasma membrane, as the potential mechanism of their cytotoxicity, was examined. The longer side-chain α-pyrrolidinophenones and their fluoro- and methoxy-analogs produce more pronounced maximal cytotoxicity, with regard to mitochondrial activity and cell membrane integrity, than the five-carbon α-PVP and its substituted derivatives. The report demonstrates, for the first time, that changes of fluidity of the interior part of plasma membrane contribute to the cytotoxicity of pyrovalerone derivatives, in addition to the previously reported mechanisms. Taking into consideration our previous findings that PV8 and PV9 produce weaker psychostimulatory effects than α-PVP, the higher cytotoxicity of the new generation of pyrovalerones can pose a serious threat to abusers, as it is possible that longer-chain compounds may be taken in higher doses to obtain similar levels of stimulation.
Collapse
Affiliation(s)
- Jakub Wojcieszak
- Department of Pharmacodynamics, Medical University of Łódź, Muszynskiego 1, 90-151, Łódź, Poland
| | - Dariusz Andrzejczak
- Department of Pharmacodynamics, Medical University of Łódź, Muszynskiego 1, 90-151, Łódź, Poland
| | - Marta Kedzierska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, 90-236, Łódź, Poland
| | - Katarzyna Milowska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, 90-236, Łódź, Poland
| | - Jolanta B Zawilska
- Department of Pharmacodynamics, Medical University of Łódź, Muszynskiego 1, 90-151, Łódź, Poland.
| |
Collapse
|
44
|
Coccini T, Vecchio S, Crevani M, De Simone U. Cytotoxic Effects of 3,4-Catechol-PV (One Major MDPV Metabolite) on Human Dopaminergic SH-SY5Y Cells. Neurotox Res 2018; 35:49-62. [DOI: 10.1007/s12640-018-9924-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/01/2018] [Accepted: 06/07/2018] [Indexed: 12/20/2022]
|
45
|
The Designer Drug 3-Fluoromethcathinone Induces Oxidative Stress and Activates Autophagy in HT22 Neuronal Cells. Neurotox Res 2018; 34:388-400. [PMID: 29656349 PMCID: PMC6154176 DOI: 10.1007/s12640-018-9898-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/27/2018] [Accepted: 03/29/2018] [Indexed: 12/24/2022]
Abstract
Synthetic cathinones are psychoactive substances, derivatives of a natural psychostimulant cathinone. Although many synthetic cathinones have lost their legal status in many countries, their abuse still continues worldwide. Recently, they have been reported to exert neurotoxic effects in vitro and in vivo. The molecular mechanisms of their action have not been fully elucidated. Recently, they have been linked to the induction of oxidative stress, autophagy, and apoptosis. The aim of this study was to investigate whether 3-fluoromethcathinone (3-FMC), a synthetic cathinone, is able to induce oxidative stress, autophagy, and apoptosis in HT22 immortalized mouse hippocampal cells. We found that treatment of HT22 cells with this compound results in a concentration-dependent increase in the intracellular production of reactive oxygen species. Moreover, 3-FMC induced concentration-dependent conversion of cytosolic LC3-I to membrane-bound LC3-II and formation of autophagic vacuoles. Additionally, the level of p62/SQSTM1 protein decreased after 3-FMC treatment, suggesting that accumulation of autophagic vacuoles resulted from activation rather than inhibition of autophagy. Our results also showed that 3-FMC at millimolar concentration is able to induce caspase-dependent apoptotic cell death in HT22 cells. Our findings suggest that abuse of 3-FMC may disturb neuronal homeostasis and impair functioning of the central nervous system.
Collapse
|
46
|
Gerecsei LI, Csillag A, Zachar G, Gévai L, Simon L, Dobolyi Á, Ádám Á. Gestational Exposure to the Synthetic Cathinone Methylenedioxypyrovalerone Results in Reduced Maternal Care and Behavioral Alterations in Mouse Pups. Front Neurosci 2018; 12:27. [PMID: 29459818 PMCID: PMC5807393 DOI: 10.3389/fnins.2018.00027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/12/2018] [Indexed: 11/13/2022] Open
Abstract
The member of synthetic cathinone family, methylenedioxypyrovalerone (MDPV), is a frequently used psychoactive drug of abuse. The objective of our study was to determine the effect of MDPV (administered from the 8th to the 14th day of gestation) on the behavior of neonatal and adolescent mice, as well as its effect on maternal care. We measured maternal care (pup retrieval test, nest building), locomotor activity (open field test), and motor coordination (grip strength test) of dams, whereas on pups we examined locomotor activity at postnatal day 7 and day 21 (open field test) and motor coordination on day 21 (grip strength test). On fresh-frozen brain samples of the dams we examined the expression of two important peptides implicated in the regulation of maternal behavior and lactation: tuberoinfundibular peptide 39 (TIP39) mRNA in the thalamic posterior intralaminar complex, and amylin mRNA in the medial preoptic nucleus. We detected decreased birth rate and survival of offspring, and reduced maternal care in the drug-treated animals, whereas there was no difference between the motility of treated and control mothers. Locomotor activity of the pups was increased in the MDPV treated group both at 7 and 21 days of age, while motor coordination was unaffected by MDPV treatment. TIP39 and amylin were detected in their typical location but failed to show a significant difference of expression between the drug-treated and control groups. The results suggest that chronic systemic administration of the cathinone agent MDPV to pregnant mice can reduce birth rate and maternal care, and it also enhances motility (without impairment of motor coordination) of the offspring.
Collapse
Affiliation(s)
- László I Gerecsei
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - András Csillag
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Gergely Zachar
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Lőrinc Gévai
- Laboratory of Sensorimotor Adaptation, Semmelweis University, Budapest, Hungary
| | - László Simon
- Laboratory of Sensorimotor Adaptation, Semmelweis University, Budapest, Hungary
| | - Árpád Dobolyi
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary.,MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary
| | - Ágota Ádám
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| |
Collapse
|