1
|
Asanuma M, Miyazaki I, Cadet JL. Differentially Expressed Nedd4-binding Protein Ndfip1 Protects Neurons Against Methamphetamine-induced Neurotoxicity. Neurotox Res 2025; 43:4. [PMID: 39808388 PMCID: PMC11732889 DOI: 10.1007/s12640-024-00725-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/30/2024] [Accepted: 12/13/2024] [Indexed: 01/30/2025]
Abstract
To identify factors involved in methamphetamine (METH) neurotoxicity, we comprehensively searched for genes which were differentially expressed in mouse striatum after METH administration using differential display (DD) reverse transcription-PCR method and sequent single-strand conformation polymorphism analysis, and found two DD cDNA fragments later identified as mRNA of Nedd4 (neural precursor cell expressed developmentally downregulated 4) WW domain-binding protein 5 (N4WBP5), later named Nedd4 family-interacting protein 1 (Ndfip1). It is an adaptor protein for the binding between Nedd4 of ubiquitin ligase (E3) and target substrate protein for ubiquitination. Northern blot analysis confirmed drastic increases in Ndfip1 mRNA in the striatum after METH injections, and in situ hybridization histochemistry showed that the mRNA expression was increased in the hippocampus and cerebellum at 2 h-2 days, in the cerebral cortex and striatum at 18 h-2 days after single METH administration. The knockdown of Ndfip1 expression with Ndfip1 siRNA significantly aggravated METH-induced neurotoxicity in the cultured monoaminergic neuronal cells. These results suggest that drastic increases in Ndfip1 mRNA is compensatory reaction to protect neurons against METH-induced neurotoxicity.
Collapse
Affiliation(s)
- Masato Asanuma
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
- Molecular Neuropsychiatry Section, Intramural Research Program, NIH/ NIDA, 21224, Baltimore, MD, U.S.A..
| | - Ikuko Miyazaki
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Section, Intramural Research Program, NIH/ NIDA, 21224, Baltimore, MD, U.S.A
| |
Collapse
|
2
|
Busceti CL, Bucci D, Damato A, De Lucia M, Venturini E, Ferrucci M, Lazzeri G, Puglisi-Allegra S, Scioli M, Carrizzo A, Nicoletti F, Vecchione C, Fornai F. Methamphetamine-Induced Blood Pressure Sensitization Correlates with Morphological Alterations within A1/C1 Catecholamine Neurons. Int J Mol Sci 2024; 25:10282. [PMID: 39408612 PMCID: PMC11476956 DOI: 10.3390/ijms251910282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Methamphetamine (METH) is a drug of abuse, which induces behavioral sensitization following repeated doses. Since METH alters blood pressure, in the present study we assessed whether systolic and diastolic blood pressure (SBP and DBP, respectively) are sensitized as well. In this context, we investigated whether alterations develop within A1/C1 neurons in the vasomotor center. C57Bl/6J male mice were administered METH (5 mg/kg, daily for 5 consecutive days). Blood pressure was measured by tail-cuff plethysmography. We found a sensitized response both to SBP and DBP, along with a significant decrease of catecholamine neurons within A1/C1 (both in the rostral and caudal ventrolateral medulla), while no changes were detected in glutamic acid decarboxylase. The decrease of catecholamine neurons was neither associated with the appearance of degeneration-related marker Fluoro-Jade B nor with altered expression of α-synuclein. Rather, it was associated with reduced free radicals and phospho-cJun and increased heat shock protein-70 and p62/sequestosome within A1/C1 cells. Blood pressure sensitization was not associated with altered arterial reactivity. These data indicate that reiterated METH administration may increase blood pressure persistently and may predispose to an increased cardiovascular response to METH. These data may be relevant to explain cardiovascular events following METH administration and stressful conditions.
Collapse
Affiliation(s)
- Carla Letizia Busceti
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
| | - Domenico Bucci
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
| | - Antonio Damato
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
| | - Massimiliano De Lucia
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
| | - Eleonora Venturini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
| | - Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (M.F.); (G.L.)
| | - Gloria Lazzeri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (M.F.); (G.L.)
| | - Stefano Puglisi-Allegra
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
| | - Mariarosaria Scioli
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
| | - Albino Carrizzo
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana” University of Salerno, 84081 Baronissi, Italy
| | - Ferdinando Nicoletti
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
- Department of Physiology and Pharmacology, University Sapienza, 00185 Roma, Italy
| | - Carmine Vecchione
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana” University of Salerno, 84081 Baronissi, Italy
| | - Francesco Fornai
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (M.F.); (G.L.)
| |
Collapse
|
3
|
Lazzeri G, Lenzi P, Busceti CL, Puglisi-Allegra S, Ferrucci M, Fornai F. Methamphetamine Increases Tubulo-Vesicular Areas While Dissipating Proteins from Vesicles Involved in Cell Clearance. Int J Mol Sci 2024; 25:9601. [PMID: 39273545 PMCID: PMC11395429 DOI: 10.3390/ijms25179601] [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: 08/01/2024] [Revised: 08/22/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Cytopathology induced by methamphetamine (METH) is reminiscent of degenerative disorders such as Parkinson's disease, and it is characterized by membrane organelles arranged in tubulo-vesicular structures. These areas, appearing as clusters of vesicles, have never been defined concerning the presence of specific organelles. Therefore, the present study aimed to identify the relative and absolute area of specific membrane-bound organelles following a moderate dose (100 µM) of METH administered to catecholamine-containing PC12 cells. Organelles and antigens were detected by immunofluorescence, and they were further quantified by plain electron microscopy and in situ stoichiometry. This analysis indicated an increase in autophagosomes and damaged mitochondria along with a decrease in lysosomes and healthy mitochondria. Following METH, a severe dissipation of hallmark proteins from their own vesicles was measured. In fact, the amounts of LC3 and p62 were reduced within autophagy vacuoles compared with the whole cytosol. Similarly, LAMP1 and Cathepsin-D within lysosomes were reduced. These findings suggest a loss of compartmentalization and confirm a decrease in the competence of cell clearing organelles during catecholamine degeneration. Such cell entropy is consistent with a loss of energy stores, which routinely govern appropriate subcellular compartmentalization.
Collapse
Affiliation(s)
- Gloria Lazzeri
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Paola Lenzi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Carla L Busceti
- IRCCS-Istituto di Ricovero e Cura a Carattere Scientifico, Neuromed, 86077 Pozzili, Italy
| | | | - Michela Ferrucci
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
- IRCCS-Istituto di Ricovero e Cura a Carattere Scientifico, Neuromed, 86077 Pozzili, Italy
| |
Collapse
|
4
|
Ferrucci M, Lenzi P, Lazzeri G, Busceti CL, Frati A, Puglisi-Allegra S, Fornai F. Combined light and electron microscopy (CLEM) to quantify methamphetamine-induced alpha-synuclein-related pathology. J Neural Transm (Vienna) 2024; 131:335-358. [PMID: 38367081 PMCID: PMC11016004 DOI: 10.1007/s00702-024-02741-x] [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: 11/20/2023] [Accepted: 01/09/2024] [Indexed: 02/19/2024]
Abstract
Methamphetamine (METH) produces a cytopathology, which is rather specific within catecholamine neurons both in vitro and ex vivo, in animal models and chronic METH abusers. This led some authors to postulate a sort of parallelism between METH cytopathology and cell damage in Parkinson's disease (PD). In fact, METH increases and aggregates alpha-syn proto-fibrils along with producing spreading of alpha-syn. Although alpha-syn is considered to be the major component of aggregates and inclusions developing within diseased catecholamine neurons including classic Lewy body (LB), at present, no study provided a quantitative assessment of this protein in situ, neither following METH nor in LB occurring in PD. Similarly, no study addressed the quantitative comparison between occurrence of alpha-syn and other key proteins and no investigation measured the protein compared with non-protein structure within catecholamine cytopathology. Therefore, the present study addresses these issues using an oversimplified model consisting of a catecholamine cell line where the novel approach of combined light and electron microscopy (CLEM) was used measuring the amount of alpha-syn, which is lower compared with p62 or poly-ubiquitin within pathological cell domains. The scenario provided by electron microscopy reveals unexpected findings, which are similar to those recently described in the pathology of PD featuring packing of autophagosome-like vesicles and key proteins shuttling autophagy substrates. Remarkably, small seed-like areas, densely packed with p62 molecules attached to poly-ubiquitin within wide vesicular domains occurred. The present data shed new light about quantitative morphometry of catecholamine cell damage in PD and within the addicted brain.
Collapse
Affiliation(s)
- Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Gloria Lazzeri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Carla L Busceti
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077, Pozzilli, Italy
| | - Alessandro Frati
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077, Pozzilli, Italy
- Neurosurgery Division, Human Neurosciences Department, Sapienza University, 00135, Rome, Italy
| | - Stefano Puglisi-Allegra
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077, Pozzilli, Italy
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077, Pozzilli, Italy.
| |
Collapse
|
5
|
Lenzi P, Lazzeri G, Ferrucci M, Scotto M, Frati A, Puglisi-Allegra S, Busceti CL, Fornai F. Is There a Place for Lewy Bodies before and beyond Alpha-Synuclein Accumulation? Provocative Issues in Need of Solid Explanations. Int J Mol Sci 2024; 25:3929. [PMID: 38612739 PMCID: PMC11011529 DOI: 10.3390/ijms25073929] [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: 03/07/2024] [Revised: 03/28/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
In the last two decades, alpha-synuclein (alpha-syn) assumed a prominent role as a major component and seeding structure of Lewy bodies (LBs). This concept is driving ongoing research on the pathophysiology of Parkinson's disease (PD). In line with this, alpha-syn is considered to be the guilty protein in the disease process, and it may be targeted through precision medicine to modify disease progression. Therefore, designing specific tools to block the aggregation and spreading of alpha-syn represents a major effort in the development of disease-modifying therapies in PD. The present article analyzes concrete evidence about the significance of alpha-syn within LBs. In this effort, some dogmas are challenged. This concerns the question of whether alpha-syn is more abundant compared with other proteins within LBs. Again, the occurrence of alpha-syn compared with non-protein constituents is scrutinized. Finally, the prominent role of alpha-syn in seeding LBs as the guilty structure causing PD is questioned. These revisited concepts may be helpful in the process of validating which proteins, organelles, and pathways are likely to be involved in the damage to meso-striatal dopamine neurons and other brain regions involved in PD.
Collapse
Affiliation(s)
- Paola Lenzi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (P.L.); (G.L.); (M.F.); (M.S.)
| | - Gloria Lazzeri
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (P.L.); (G.L.); (M.F.); (M.S.)
| | - Michela Ferrucci
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (P.L.); (G.L.); (M.F.); (M.S.)
| | - Marco Scotto
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (P.L.); (G.L.); (M.F.); (M.S.)
| | - Alessandro Frati
- IRCCS—Istituto di Ricovero e Cura a Carattere Scientifico, Neuromed, 86077 Pozzili, Italy or (A.F.); (S.P.-A.); (C.L.B.)
- Neurosurgery Division, Department of Human Neurosciences, Sapienza University, 00135 Roma, Italy
| | - Stefano Puglisi-Allegra
- IRCCS—Istituto di Ricovero e Cura a Carattere Scientifico, Neuromed, 86077 Pozzili, Italy or (A.F.); (S.P.-A.); (C.L.B.)
| | - Carla Letizia Busceti
- IRCCS—Istituto di Ricovero e Cura a Carattere Scientifico, Neuromed, 86077 Pozzili, Italy or (A.F.); (S.P.-A.); (C.L.B.)
| | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (P.L.); (G.L.); (M.F.); (M.S.)
- IRCCS—Istituto di Ricovero e Cura a Carattere Scientifico, Neuromed, 86077 Pozzili, Italy or (A.F.); (S.P.-A.); (C.L.B.)
| |
Collapse
|
6
|
Seyed Aliyan SM, Roohbakhsh A, Jafari Fakhrabad M, Salmasi Z, Moshiri M, Shahbazi N, Etemad L. Evaluating the Protective Effects of Thymoquinone on Methamphetamine-induced Toxicity in an In Vitro Model Based on Differentiated PC12 Cells. Altern Lab Anim 2024; 52:94-106. [PMID: 38445454 DOI: 10.1177/02611929241237409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Methamphetamine (Meth) is a highly addictive stimulant. Its potential neurotoxic effects are mediated through various mechanisms, including oxidative stress and the initiation of the apoptotic process. Thymoquinone (TQ), obtained from Nigella sativa seed oil, has extensive antioxidant and anti-apoptotic properties. This study aimed to investigate the potential protective effects of TQ against Meth-induced toxicity by using an in vitro model based on nerve growth factor-differentiated PC12 cells. Cell differentiation was assessed by detecting the presence of a neuronal marker with flow cytometry. The effects of Meth exposure were evaluated in the in vitro neuronal cell-based model via the determination of cell viability (in an MTT assay) and apoptosis (by annexin/propidium iodide staining). The generation of reactive oxygen species (ROS), as well as the levels of glutathione (GSH) and dopamine, were also determined. The model was used to determine the protective effects of 0.5, 1 and 2 μM TQ against Meth-induced toxicity (at 1 mM). The results showed that TQ reduced Meth-induced neurotoxicity, possibly through the inhibition of ROS generation and apoptosis, and by helping to maintain GSH and dopamine levels. Thus, the impact of TQ treatment on Meth-induced neurotoxicity could warrant further investigation.
Collapse
Affiliation(s)
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Jafari Fakhrabad
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahar Salmasi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Moshiri
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Clinical Toxicology, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niosha Shahbazi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Patrick MB, Omar N, Werner CT, Mitra S, Jarome TJ. The ubiquitin-proteasome system and learning-dependent synaptic plasticity - A 10 year update. Neurosci Biobehav Rev 2023; 152:105280. [PMID: 37315660 PMCID: PMC11323321 DOI: 10.1016/j.neubiorev.2023.105280] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/22/2023] [Accepted: 06/08/2023] [Indexed: 06/16/2023]
Abstract
Over 25 years ago, a seminal paper demonstrated that the ubiquitin-proteasome system (UPS) was involved in activity-dependent synaptic plasticity. Interest in this topic began to expand around 2008 following another seminal paper showing that UPS-mediated protein degradation controlled the "destabilization" of memories following retrieval, though we remained with only a basic understanding of how the UPS regulated activity- and learning-dependent synaptic plasticity. However, over the last 10 years there has been an explosion of papers on this topic that has significantly changed our understanding of how ubiquitin-proteasome signaling regulates synaptic plasticity and memory formation. Importantly, we now know that the UPS controls much more than protein degradation, is involved in plasticity underlying drugs of abuse and that there are significant sex differences in how ubiquitin-proteasome signaling is used for memory storage processes. Here, we aim to provide a critical 10-year update on the role of ubiquitin-proteasome signaling in synaptic plasticity and memory formation, including updated cellular models of how ubiquitin-proteasome activity could be regulating learning-dependent synaptic plasticity in the brain.
Collapse
Affiliation(s)
- Morgan B Patrick
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Nour Omar
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Craig T Werner
- Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA; National Center for Wellness and Recovery, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA.
| | - Swarup Mitra
- Department of Biomedical Sciences, Joan C Edwards School of Medicine, Marshall University, Huntington, WV, USA.
| | - Timothy J Jarome
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA; School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
8
|
Puglisi-Allegra S, Lazzeri G, Busceti CL, Giorgi FS, Biagioni F, Fornai F. Lithium engages autophagy for neuroprotection and neuroplasticity: translational evidence for therapy. Neurosci Biobehav Rev 2023; 148:105148. [PMID: 36996994 DOI: 10.1016/j.neubiorev.2023.105148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/23/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023]
Abstract
Here an overview is provided on therapeutic/neuroprotective effects of Lithium (Li+) in neurodegenerative and psychiatric disorders focusing on the conspicuous action of Li+ through autophagy. The effects on the autophagy machinery remain the key molecular mechanisms to explain the protective effects of Li+ for neurodegenerative diseases, offering potential therapeutic strategies for the treatment of neuropsychiatric disorders and emphasizes a crossroad linking autophagy, neurodegenerative disorders, and mood stabilization. Sensitization by psychostimulants points to several mechanisms involved in psychopathology, most also crucial in neurodegenerative disorders. Evidence shows the involvement of autophagy and metabotropic Glutamate receptors-5 (mGluR5) in neurodegeneration due to methamphetamine neurotoxicity as well as in neuroprotection, both in vitro and in vivo models. More recently, Li+ was shown to modulate autophagy through its action on mGluR5, thus pointing to an additional way of autophagy engagement by Li+ and to a substantial role of mGluR5 in neuroprotection related to neural e neuropsychiatry diseases. We propose Li+ engagement of autophagy through the canonical mechanisms of autophagy machinery and through the intermediary of mGluR5.
Collapse
|
9
|
Guo D, Huang X, Xiong T, Wang X, Zhang J, Wang Y, Liang J. Molecular mechanisms of programmed cell death in methamphetamine-induced neuronal damage. Front Pharmacol 2022; 13:980340. [PMID: 36059947 PMCID: PMC9428134 DOI: 10.3389/fphar.2022.980340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 07/28/2022] [Indexed: 12/02/2022] Open
Abstract
Methamphetamine, commonly referred to as METH, is a highly addictive psychostimulant and one of the most commonly misused drugs on the planet. Using METH continuously can increase your risk for drug addiction, along with other health complications like attention deficit disorder, memory loss, and cognitive decline. Neurotoxicity caused by METH is thought to play a significant role in the onset of these neurological complications. The molecular mechanisms responsible for METH-caused neuronal damage are discussed in this review. According to our analysis, METH is closely associated with programmed cell death (PCD) in the process that causes neuronal impairment, such as apoptosis, autophagy, necroptosis, pyroptosis, and ferroptosis. In reviewing this article, some insights are gained into how METH addiction is accompanied by cell death and may help to identify potential therapeutic targets for the neurological impairment caused by METH abuse.
Collapse
Affiliation(s)
- Dongming Guo
- Institute of Translational Medicine, Medical, Yangzhou University, Yangzhou, China
| | - Xinlei Huang
- Institute of Translational Medicine, Medical, Yangzhou University, Yangzhou, China
| | - Tianqing Xiong
- Institute of Translational Medicine, Medical, Yangzhou University, Yangzhou, China
| | - Xingyi Wang
- Institute of Translational Medicine, Medical, Yangzhou University, Yangzhou, China
| | - Jingwen Zhang
- Institute of Translational Medicine, Medical, Yangzhou University, Yangzhou, China
| | - Yingge Wang
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Jingyan Liang
- Institute of Translational Medicine, Medical, Yangzhou University, Yangzhou, China
- *Correspondence: Jingyan Liang,
| |
Collapse
|
10
|
Ferrucci M, Busceti CL, Lazzeri G, Biagioni F, Puglisi-Allegra S, Frati A, Lenzi P, Fornai F. Bacopa Protects against Neurotoxicity Induced by MPP+ and Methamphetamine. Molecules 2022; 27:molecules27165204. [PMID: 36014442 PMCID: PMC9414486 DOI: 10.3390/molecules27165204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/08/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
The neurotoxins methamphetamine (METH) and 1-methyl-4-phenylpyridinium (MPP+) damage catecholamine neurons. Although sharing the same mechanism to enter within these neurons, METH neurotoxicity mostly depends on oxidative species, while MPP+ toxicity depends on the inhibition of mitochondrial activity. This explains why only a few compounds protect against both neurotoxins. Identifying a final common pathway that is shared by these neurotoxins is key to prompting novel remedies for spontaneous neurodegeneration. In the present study we assessed whether natural extracts from Bacopa monnieri (BM) may provide a dual protection against METH- and MPP+-induced cell damage as measured by light and electron microscopy. The protection induced by BM against catecholamine cell death and degeneration was dose-dependently related to the suppression of reactive oxygen species (ROS) formation and mitochondrial alterations. These were measured by light and electron microscopy with MitoTracker Red and Green as well as by the ultrastructural morphometry of specific mitochondrial structures. In fact, BM suppresses the damage of mitochondrial crests and matrix dilution and increases the amount of healthy and total mitochondria. The present data provide evidence for a natural compound, which protects catecholamine cells independently by the type of experimental toxicity. This may be useful to counteract spontaneous degenerations of catecholamine cells.
Collapse
Affiliation(s)
- Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | | | - Gloria Lazzeri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | | | | | - Alessandro Frati
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
- Neurosurgery Division, Department of Human Neurosciences, Sapienza University, 00135 Rome, Italy
| | - Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
- Correspondence: or ; Tel.: +39-050-221-8667
| |
Collapse
|
11
|
Lenzi P, Biagioni F, Busceti CL, Lazzeri G, Polzella M, Frati A, Ferrucci M, Fornai F. Alterations of Mitochondrial Structure in Methamphetamine Toxicity. Int J Mol Sci 2022; 23:ijms23168926. [PMID: 36012188 PMCID: PMC9408775 DOI: 10.3390/ijms23168926] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 12/30/2022] Open
Abstract
Recent evidence shows that methamphetamine (METH) produces mitochondrial alterations that contribute to neurotoxicity. Nonetheless, most of these studies focus on mitochondrial activity, whereas mitochondrial morphology remains poorly investigated. In fact, morphological evidence about the fine structure of mitochondria during METH toxicity is not available. Thus, in the present study we analyzed dose-dependent mitochondrial structural alterations during METH exposure. Light and transmission electron microscopy were used, along with ultrastructural stoichiometry of catecholamine cells following various doses of METH. In the first part of the study cell death and cell degeneration were assessed and they were correlated with mitochondrial alterations observed using light microscopy. In the second part of the study, ultrastructural evidence of specific mitochondrial alterations of crests, inner and outer membranes and matrix were quantified, along with in situ alterations of mitochondrial proteins. Neurodegeneration induced by METH correlates significantly with specific mitochondrial damage, which allows definition of a scoring system for mitochondrial integrity. In turn, mitochondrial alterations are concomitant with a decrease in fission/mitophagy protein Fis1 and DRP1 and an increase in Pink1 and Parkin in situ, at the mitochondrial level. These findings provide structural evidence that mitochondria represent both direct and indirect targets of METH-induced toxicity
Collapse
Affiliation(s)
- Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Francesca Biagioni
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
| | - Carla L. Busceti
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
| | - Gloria Lazzeri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Maico Polzella
- Aliveda Laboratories, Viale Karol Wojtyla, 19, 56042 Crespina Lorenzana, Italy
| | - Alessandro Frati
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
- Neurosurgery Division, Department of Human Neurosciences, Sapienza University, 00135 Roma, Italy
| | - Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
- Correspondence: or ; Tel.: +39-050-2218667
| |
Collapse
|
12
|
Fucoxanthin alleviates methamphetamine-induced neurotoxicity possibly via the inhibition of interaction between Keap1 and Nrf2. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104713] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
13
|
Ferrucci M, Biagioni F, Busceti CL, Vidoni C, Castino R, Isidoro C, Ryskalin L, Frati A, Puglisi-Allegra S, Fornai F. Inhibition of Autophagy In Vivo Extends Methamphetamine Toxicity to Mesencephalic Cell Bodies. Pharmaceuticals (Basel) 2021; 14:ph14101003. [PMID: 34681227 PMCID: PMC8538796 DOI: 10.3390/ph14101003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 01/21/2023] Open
Abstract
Methamphetamine (METH) is a widely abused psychostimulant and a stress-inducing compound, which leads to neurotoxicity for nigrostriatal dopamine (DA) terminals in rodents and primates including humans. In vitro studies indicate that autophagy is a strong modulator of METH toxicity. In detail, suppressing autophagy increases METH toxicity, while stimulating autophagy prevents METH-induced toxicity in cell cultures. In the present study, the role of autophagy was investigated in vivo. In the whole brain, METH alone destroys meso-striatal DA axon terminals, while fairly sparing DA cell bodies within substantia nigra pars compacta (SNpc). No damage to either cell bodies or axons from ventral tegmental area (VTA) is currently documented. According to the hypothesis that ongoing autophagy prevents METH-induced DA toxicity, we tested whether systemic injection of autophagy inhibitors such as asparagine (ASN, 1000 mg/Kg) or glutamine (GLN, 1000 mg/Kg), may extend METH toxicity to DA cell bodies, both within SNpc and VTA, where autophagy was found to be inhibited. When METH (5 mg/Kg × 4, 2 h apart) was administered to C57Bl/6 mice following ASN or GLN, a frank loss of cell bodies takes place within SNpc and a loss of both axons and cell bodies of VTA neurons is documented. These data indicate that, ongoing autophagy protects DA neurons and determines the refractoriness of cell bodies to METH-induced toxicity.
Collapse
Affiliation(s)
- Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (M.F.); (L.R.)
| | - Francesca Biagioni
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Isernia, Italy; (F.B.); (C.L.B.); (A.F.); (S.P.-A.)
| | - Carla L. Busceti
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Isernia, Italy; (F.B.); (C.L.B.); (A.F.); (S.P.-A.)
| | - Chiara Vidoni
- Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (C.V.); (R.C.); (C.I.)
| | - Roberta Castino
- Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (C.V.); (R.C.); (C.I.)
| | - Ciro Isidoro
- Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (C.V.); (R.C.); (C.I.)
| | - Larisa Ryskalin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (M.F.); (L.R.)
| | - Alessandro Frati
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Isernia, Italy; (F.B.); (C.L.B.); (A.F.); (S.P.-A.)
- Neurosurgery Division, Human Neurosciences Department, Sapienza University, 00135 Rome, Italy
| | - Stefano Puglisi-Allegra
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Isernia, Italy; (F.B.); (C.L.B.); (A.F.); (S.P.-A.)
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (M.F.); (L.R.)
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Isernia, Italy; (F.B.); (C.L.B.); (A.F.); (S.P.-A.)
- Correspondence: or ; Tel.: +39-050-2218601
| |
Collapse
|
14
|
Ryskalin L, Biagioni F, Busceti CL, Polzella M, Lenzi P, Frati A, Ferrucci M, Fornai F. Lactoferrin Protects against Methamphetamine Toxicity by Modulating Autophagy and Mitochondrial Status. Nutrients 2021; 13:nu13103356. [PMID: 34684361 PMCID: PMC8537867 DOI: 10.3390/nu13103356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/17/2021] [Accepted: 09/23/2021] [Indexed: 01/18/2023] Open
Abstract
Lactoferrin (LF) was used at first as a vehicle to deliver non-soluble active compounds to the body, including the central nervous system (CNS). Nonetheless, it soon became evident that, apart from acting as a vehicle, LF itself owns active effects in the CNS. In the present study, the effects of LF are assessed both in baseline conditions, as well as to counteract methamphetamine (METH)-induced neurodegeneration by assessing cell viability, cell phenotype, mitochondrial status, and specific autophagy steps. In detail, cell integrity in baseline conditions and following METH administration was carried out by using H&E staining, Trypan blue, Fluoro Jade B, and WST-1. Western blot and immuno-fluorescence were used to assess the expression of the neurofilament marker βIII-tubulin. Mitochondria were stained using Mito Tracker Red and Green and were further detailed and quantified by using transmission electron microscopy. Autophagy markers were analyzed through immuno-fluorescence and electron microscopy. LF counteracts METH-induced degeneration. In detail, LF significantly attenuates the amount of cell loss and mitochondrial alterations produced by METH; and mitigates the dissipation of autophagy-related proteins from the autophagy compartment, which is massively induced by METH. These findings indicate a protective role of LF in the molecular mechanisms of neurodegeneration.
Collapse
Affiliation(s)
- Larisa Ryskalin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (L.R.); (P.L.); (M.F.)
| | - Francesca Biagioni
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (A.F.)
| | - Carla L. Busceti
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (A.F.)
| | - Maico Polzella
- Aliveda Laboratories, Viale Karol Wojtyla, 19, 56042 Crespina Lorenzana, Italy;
| | - Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (L.R.); (P.L.); (M.F.)
| | - Alessandro Frati
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (A.F.)
- Neurosurgery Division, Human Neurosciences Department, Sapienza University, 00135 Rome, Italy
| | - Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (L.R.); (P.L.); (M.F.)
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (L.R.); (P.L.); (M.F.)
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (A.F.)
- Correspondence:
| |
Collapse
|
15
|
Costa G, Caputi FF, Serra M, Simola N, Rullo L, Stamatakos S, Sanna F, Germain M, Martinoli MG, Candeletti S, Morelli M, Romualdi P. Activation of Antioxidant and Proteolytic Pathways in the Nigrostriatal Dopaminergic System After 3,4-Methylenedioxymethamphetamine Administration: Sex-Related Differences. Front Pharmacol 2021; 12:713486. [PMID: 34512343 PMCID: PMC8430399 DOI: 10.3389/fphar.2021.713486] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/12/2021] [Indexed: 12/29/2022] Open
Abstract
3,4-Methylenedioxymethamphetamine (MDMA, “ecstasy”) is an amphetamine-related drug that may damage the dopaminergic nigrostriatal system. To investigate the mechanisms that sustain this toxic effect and ascertain their sex-dependence, we evaluated in the nigrostriatal system of MDMA-treated (4 × 20 mg/kg, 2 h apart) male and female mice the activity of superoxide dismutase (SOD), the gene expression of SOD type 1 and 2, together with SOD1/2 co-localization with tyrosine hydroxylase (TH)-positive neurons. In the same mice and brain areas, activity of glutathione peroxidase (GPx) and of β2/β5 subunits of the ubiquitin-proteasome system (UPS) were also evaluated. After MDMA, SOD1 increased in striatal TH-positive terminals, but not nigral neurons, of males and females, while SOD2 increased in striatal TH-positive terminals and nigral neurons of males only. Moreover, after MDMA, SOD1 gene expression increased in the midbrain of males and females, whereas SOD2 increased only in males. Finally, MDMA increased the SOD activity in the midbrain of females, without affecting GPx activity, decreased the β2/β5 activities in the striatum of males and the β2 activity in the midbrain of females. These results suggest that the mechanisms of MDMA-induced neurotoxic effects are sex-dependent and dopaminergic neurons of males could be more sensitive to SOD2- and UPS-mediated toxic effects.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Francesca Felicia Caputi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Marcello Serra
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Laura Rullo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Serena Stamatakos
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Fabrizio Sanna
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Marc Germain
- Department of Medical Biology, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada.,CERMO-FC UQAM, Québec, QC, Canada
| | - Maria-Grazia Martinoli
- Department of Medical Biology, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada.,Department of Psychiatry and Neuroscience, Université Laval and CHU Research Center, Québec, QC, Canada
| | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy.,National Research Council of Italy, Neuroscience Institute, Cagliari, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| |
Collapse
|
16
|
Shin EJ, Jeong JH, Hwang Y, Sharma N, Dang DK, Nguyen BT, Nah SY, Jang CG, Bing G, Nabeshima T, Kim HC. Methamphetamine-induced dopaminergic neurotoxicity as a model of Parkinson's disease. Arch Pharm Res 2021; 44:668-688. [PMID: 34286473 DOI: 10.1007/s12272-021-01341-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/06/2021] [Indexed: 12/01/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with a high prevalence, approximately 1 % in the elderly population. Numerous studies have demonstrated that methamphetamine (MA) intoxication caused the neurological deficits and nigrostriatal damage seen in Parkinsonian conditions, and subsequent rodent studies have found that neurotoxic binge administration of MA reproduced PD-like features, in terms of its symptomatology and pathology. Several anti-Parkinsonian medications have been shown to attenuate the motor impairments and dopaminergic damage induced by MA. In addition, it has been recognized that mitochondrial dysfunction, oxidative stress, pro-apoptosis, proteasomal/autophagic impairment, and neuroinflammation play important roles in inducing MA neurotoxicity. Importantly, MA neurotoxicity has been shown to share a common mechanism of dopaminergic toxicity with that of PD pathogenesis. This review describes the major findings on the neuropathological features and underlying neurotoxic mechanisms induced by MA and compares them with Parkinsonian pathogenesis. Taken together, it is suggested that neurotoxic binge-type administration of MA in rodents is a valid animal model for PD that may provide knowledge on the neuropathogenesis of PD.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, 06974, Seoul, Republic of Korea
| | - Yeonggwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.,Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, 06974, Seoul, Republic of Korea
| | - Duy-Khanh Dang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.,Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, 900000, Can Tho City, Vietnam
| | - Bao-Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory, Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, 05029, Seoul, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, 16419, Suwon, Republic of Korea
| | - Guoying Bing
- Department of Neuroscience, College of Medicine, University of Kentucky, KY, 40536, Lexington, USA
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Graduate School of Health Science, Fujita Health University, 470-1192, Toyoake, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea. .,Neuropsychopharmacology & Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.
| |
Collapse
|
17
|
Lazzeri G, Busceti CL, Biagioni F, Fabrizi C, Morucci G, Giorgi FS, Ferrucci M, Lenzi P, Puglisi-Allegra S, Fornai F. Norepinephrine Protects against Methamphetamine Toxicity through β2-Adrenergic Receptors Promoting LC3 Compartmentalization. Int J Mol Sci 2021; 22:7232. [PMID: 34281286 PMCID: PMC8269332 DOI: 10.3390/ijms22137232] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/01/2021] [Accepted: 07/03/2021] [Indexed: 01/18/2023] Open
Abstract
Norepinephrine (NE) neurons and extracellular NE exert some protective effects against a variety of insults, including methamphetamine (Meth)-induced cell damage. The intimate mechanism of protection remains difficult to be analyzed in vivo. In fact, this may occur directly on target neurons or as the indirect consequence of NE-induced alterations in the activity of trans-synaptic loops. Therefore, to elude neuronal networks, which may contribute to these effects in vivo, the present study investigates whether NE still protects when directly applied to Meth-treated PC12 cells. Meth was selected based on its detrimental effects along various specific brain areas. The study shows that NE directly protects in vitro against Meth-induced cell damage. The present study indicates that such an effect fully depends on the activation of plasma membrane β2-adrenergic receptors (ARs). Evidence indicates that β2-ARs activation restores autophagy, which is impaired by Meth administration. This occurs via restoration of the autophagy flux and, as assessed by ultrastructural morphometry, by preventing the dissipation of microtubule-associated protein 1 light chain 3 (LC3) from autophagy vacuoles to the cytosol, which is produced instead during Meth toxicity. These findings may have an impact in a variety of degenerative conditions characterized by NE deficiency along with autophagy impairment.
Collapse
Affiliation(s)
- Gloria Lazzeri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy; (G.L.); (G.M.); (F.S.G.); (M.F.); (P.L.)
| | - Carla L. Busceti
- I.R.C.C.S. Neuromed, via Atinense 18, 86077 Pozzilli, Italy; (C.L.B.); (F.B.); (S.P.-A.)
| | - Francesca Biagioni
- I.R.C.C.S. Neuromed, via Atinense 18, 86077 Pozzilli, Italy; (C.L.B.); (F.B.); (S.P.-A.)
| | - Cinzia Fabrizi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, via A. Borelli 50, 00161 Rome, Italy;
| | - Gabriele Morucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy; (G.L.); (G.M.); (F.S.G.); (M.F.); (P.L.)
| | - Filippo S. Giorgi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy; (G.L.); (G.M.); (F.S.G.); (M.F.); (P.L.)
| | - Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy; (G.L.); (G.M.); (F.S.G.); (M.F.); (P.L.)
| | - Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy; (G.L.); (G.M.); (F.S.G.); (M.F.); (P.L.)
| | | | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy; (G.L.); (G.M.); (F.S.G.); (M.F.); (P.L.)
- I.R.C.C.S. Neuromed, via Atinense 18, 86077 Pozzilli, Italy; (C.L.B.); (F.B.); (S.P.-A.)
| |
Collapse
|
18
|
Limanaqi F, Busceti CL, Celli R, Biagioni F, Fornai F. Autophagy as a gateway for the effects of methamphetamine: From neurotransmitter release and synaptic plasticity to psychiatric and neurodegenerative disorders. Prog Neurobiol 2021; 204:102112. [PMID: 34171442 DOI: 10.1016/j.pneurobio.2021.102112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/27/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
As a major eukaryotic cell clearing machinery, autophagy grants cell proteostasis, which is key for neurotransmitter release, synaptic plasticity, and neuronal survival. In line with this, besides neuropathological events, autophagy dysfunctions are bound to synaptic alterations that occur in mental disorders, and early on, in neurodegenerative diseases. This is also the case of methamphetamine (METH) abuse, which leads to psychiatric disturbances and neurotoxicity. While consistently altering the autophagy machinery, METH produces behavioral and neurotoxic effects through molecular and biochemical events that can be recapitulated by autophagy blockade. These consist of altered physiological dopamine (DA) release, abnormal stimulation of DA and glutamate receptors, as well as oxidative, excitotoxic, and neuroinflammatory events. Recent molecular insights suggest that METH early impairs the autophagy machinery, though its functional significance remains to be investigated. Here we discuss evidence suggesting that alterations of DA transmission and autophagy are intermingled within a chain of events underlying behavioral alterations and neurodegenerative phenomena produced by METH. Understanding how METH alters the autophagy machinery is expected to provide novel insights into the neurobiology of METH addiction sharing some features with psychiatric disorders and parkinsonism.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55, 56126, Pisa, PI, Italy
| | | | - Roberta Celli
- IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, IS, Italy
| | | | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55, 56126, Pisa, PI, Italy; IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, IS, Italy.
| |
Collapse
|
19
|
Limanaqi F, Biagioni F, Salvetti A, Puglisi-Allegra S, Lenzi P, Fornai F. Morphology, clearing efficacy, and mTOR dependency of the organelle autophagoproteasome. Eur J Histochem 2021; 65. [PMID: 34060734 PMCID: PMC8200839 DOI: 10.4081/ejh.2021.3220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/01/2021] [Indexed: 01/18/2023] Open
Abstract
The interplay between autophagy (ATG) and ubiquitin proteasome (UP) cell-clearing systems was recently evidenced at biochemical and morphological levels, where subunits belonging to both pathways co-localize within a novel organelle named autophagoproteasome (APP). We previously documented that APP occurs at baseline conditions, while it is hindered by neurotoxicant administration. This is bound to the activity of the mechanistic target of rapamycin (mTOR), since APP is stimulated by mTOR inhibition, which in turn, is correlated with cell protection. In this brief report, we provide novel morphological and biochemical evidence on APP, suggesting the presence of active UP subunits within ATG vacuoles. Although a stream of interpretation considers such a merging as a catabolic pathway to clear inactive UP subunits, our data further indicate that UP-ATG merging may rather provide an empowered catalytic organelle.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa.
| | | | | | | | - Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa.
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa; IRCCS Neuromed, Pozzilli (IS).
| |
Collapse
|
20
|
Neuroprotective Effects of Curcumin in Methamphetamine-Induced Toxicity. Molecules 2021; 26:molecules26092493. [PMID: 33923340 PMCID: PMC8123176 DOI: 10.3390/molecules26092493] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 01/18/2023] Open
Abstract
Curcumin (CUR), a natural polyphenol extracted from rhizome of the Curcuma longa L, has received great attention for its multiple potential health benefits as well as disease prevention. For instance, CUR protects against toxic agents acting on the human body, including the nervous system. In detail, CUR possesses, among others, strong effects as an autophagy activator. The present study indicates that CUR counteracts methamphetamine (METH) toxicity. Such a drug of abuse is toxic by disturbing the autophagy machinery. We profited from an unbiased, low variable cell context by using rat pheochromocytoma PC12 cell line. In such a system, a strong protection was exerted by CUR against METH toxicity. This was associated with increased autophagy flux, merging of autophagosomes with lysosomes and replenishment of autophagy vacuoles with LC3, which instead is moved out from the vacuoles by METH. This is expected to enable the autophagy machinery. In fact, while in METH-treated cells the autophagy substrates α-synuclein accumulates in the cytosol, CUR speeds up α-synuclein clearance. Under the effects of CUR LC3 penetrate in autophagy vacuoles to commit them to cell clearance and promotes the autophagy flux. The present data provide evidence that CUR counteracts the neurotoxic effects induced by METH by promoting autophagy.
Collapse
|
21
|
Wu M, Su H, Zhao M. The Role of α-Synuclein in Methamphetamine-Induced Neurotoxicity. Neurotox Res 2021; 39:1007-1021. [PMID: 33555547 DOI: 10.1007/s12640-021-00332-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 12/19/2020] [Accepted: 01/06/2021] [Indexed: 12/28/2022]
Abstract
Methamphetamine (METH), a highly addictive psychostimulant, is the second most widely used illicit drug. METH produces damage dopamine neurons and apoptosis via multiple inter-regulating mechanisms, including dopamine overload, hyperthermia, oxidative stress, mitochondria dysfunction, endoplasmic reticulum stress, protein degradation system dysfunction, and neuroinflammation. Increasing evidence suggests that chronic METH abuse is associated with neurodegenerative changes in the human brain and an increased risk of Parkinson's disease (PD). METH use and PD may share some common steps in causing neurotoxicity. Accumulation of α-synuclein, a presynaptic protein, is the pathological hallmark of PD. Intriguingly, α-synuclein upregulation and aggregation are also found in dopaminergic neurons in the substantia nigra in chronic METH users. This suggests α-synuclein may play a role in METH-induced neurotoxicity. The mechanism of α-synuclein cytotoxicity in PD has attracted considerable attention; however, how α-synuclein affects METH-induced neurotoxicity has not been reviewed. In this review, we summarize the relationship between METH use and PD, interdependent mechanisms that are involved in METH-induced neurotoxicity and the significance of α-synuclein upregulation in response to METH use. The identification of α-synuclein overexpression and aggregation as a contributor to METH-induced neurotoxicity may provide a novel therapeutic target for the treatment of the deleterious effect of this drug and drug addiction.
Collapse
Affiliation(s)
- Manqing Wu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hang Su
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Zhao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China.
- Shanghai Clinical Research Center for Mental Health, Shanghai, China.
- CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
22
|
Asser A, Kõks S, Soomets U, Terasmaa A, Sauk M, Eltermaa M, Piip P, Ubhayasekera K, Bergquist J, Taba P. Acute effects of methcathinone and manganese in mice: A dose response study. Heliyon 2019; 5:e02475. [PMID: 31687570 PMCID: PMC6819833 DOI: 10.1016/j.heliyon.2019.e02475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 06/15/2019] [Accepted: 09/11/2019] [Indexed: 11/24/2022] Open
Abstract
An intravenously injectable illicit drug made by mixing pseudoephedrine, potassium permanganate, vinegar and water, yielding methcathinone (Mcat) and manganese (Mn), induces an extrapyramidal syndrome with parkinsonism, dystonia, gait and balance disorders similar to manganism. Although the cause of the syndrome is largely attributed to Mn, the interaction of the drug's individual components is not known and the role of Mcat is possibly underestimated. Aim of the present study was to analyze dose-dependent behavioral effects of the mixture and its two main active components Mcat and Mn in an acute setting and determine the lethal doses of each substance. Three groups of C57BL/6 mice were injected intraperitoneally with (1) the drug mixture containing 10, 25, 50, 100 or 150 mg of Mcat and respectively 1.6, 3.8, 6.9, 17.1 and 22.6 mg of Mn per kilogram of body weight; (2) 10, 25, 50, 100, 150, 200 or 300 mg of racemic Mcat/kg of body weight; (3) MnCl2 10, 25 or 50 mg/kg of body weight. Locomotor activity of the animals, various signs and time of death were recorded. Lower doses (10 and 25 mg/kg) of Mcat had a clear motor activity stimulating effect and this was clearly dose-dependent. High doses of Mcat produced epileptic seizures in 74% of the animals and became lethal with the highest doses. Similarly, the mixture had a clear dose-dependent stimulating effect and the higher doses became lethal. The LD50 of the pseudoephedrine mixture was 110.2 mg of Mcat/kg and for pure Mcat 201.7 mg/kg. Mn did not prove to be lethal in doses up to 50 mg/kg, but had a strong dose dependent inhibitory effect on the animals’ behavior. Our data reveal that both Mn and Mcat have a significant role in the toxicity of the mixture.
Collapse
Affiliation(s)
- Andres Asser
- Department of Neurology and Neurosurgery, University of Tartu, Puusepa 8, Tartu, 51014, Estonia
| | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, University of Western Australia, Perth, WA, Australia
| | - Ursel Soomets
- Department of Biochemistry, University of Tartu, Ravila 19, Tartu, 50411, Estonia
| | - Anton Terasmaa
- Department of Physiology, University of Tartu, Ravila 19, Tartu, 50411, Estonia
| | - Martin Sauk
- Institute of Molecular and Cell Biology, University of Tartu, Riia 23, Tartu, 51010, Estonia
| | - Mall Eltermaa
- Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, Tartu, 50411, Estonia
| | - Piret Piip
- Department of Neurology and Neurosurgery, University of Tartu, Puusepa 8, Tartu, 51014, Estonia
| | - Kumari Ubhayasekera
- Department of Chemistry, Biomedical Center, Uppsala University, SE-751 24, Uppsala, Sweden
| | - Jonas Bergquist
- Department of Chemistry, Biomedical Center, Uppsala University, SE-751 24, Uppsala, Sweden
| | - Pille Taba
- Department of Neurology and Neurosurgery, University of Tartu, Puusepa 8, Tartu, 51014, Estonia
| |
Collapse
|
23
|
Biagioni F, Ferese R, Limanaqi F, Madonna M, Lenzi P, Gambardella S, Fornai F. Methamphetamine persistently increases alpha-synuclein and suppresses gene promoter methylation within striatal neurons. Brain Res 2019; 1719:157-175. [DOI: 10.1016/j.brainres.2019.05.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/24/2019] [Accepted: 05/27/2019] [Indexed: 12/20/2022]
|
24
|
mTOR Modulates Methamphetamine-Induced Toxicity through Cell Clearing Systems. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6124745. [PMID: 30647813 PMCID: PMC6311854 DOI: 10.1155/2018/6124745] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/31/2018] [Indexed: 11/17/2022]
Abstract
Methamphetamine (METH) is abused worldwide, and it represents a threat for public health. METH exposure induces a variety of detrimental effects. In fact, METH produces a number of oxidative species, which lead to lipid peroxidation, protein misfolding, and nuclear damage. Cell clearing pathways such as ubiquitin-proteasome (UP) and autophagy (ATG) are involved in METH-induced oxidative damage. Although these pathways were traditionally considered to operate as separate metabolic systems, recent studies demonstrate their interconnection at the functional and biochemical level. Very recently, the convergence between UP and ATG was evidenced within a single organelle named autophagoproteasome (APP), which is suppressed by mTOR activation. In the present research study, the occurrence of APP during METH toxicity was analyzed. In fact, coimmunoprecipitation indicates a binding between LC3 and P20S particles, which also recruit p62 and alpha-synuclein. The amount of METH-induced toxicity correlates with APP levels. Specific markers for ATG and UP, such as LC3 and P20S in the cytosol, and within METH-induced vacuoles, were measured at different doses and time intervals following METH administration either alone or combined with mTOR modulators. Western blotting, coimmunoprecipitation, light microscopy, confocal microscopy, plain transmission electron microscopy, and immunogold staining were used to document the effects of mTOR modulation on METH toxicity and the merging of UP with ATG markers within APPs. METH-induced cell death is prevented by mTOR inhibition, while it is worsened by mTOR activation, which correlates with the amount of autophagoproteasomes. The present data, which apply to METH toxicity, are also relevant to provide a novel insight into cell clearing pathways to counteract several kinds of oxidative damage.
Collapse
|
25
|
Activation of the interferon type I response rather than autophagy contributes to myogenesis inhibition in congenital DM1 myoblasts. Cell Death Dis 2018; 9:1071. [PMID: 30341284 PMCID: PMC6195593 DOI: 10.1038/s41419-018-1080-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 12/16/2022]
Abstract
Congenital myotonic dystrophy type 1 (CDM1) is characterized by severe symptoms that affect patients from birth, with 40% mortality in the neonatal period and impaired skeletal muscle development. In this paper, we examined the relationship between autophagy and abnormal myogenic differentiation of CDM1 myoblasts. We investigated these pathological features at both ultrastructural and molecular levels, utilizing two CDM1 foetal myoblasts, CDM13 and CDM15, with 1800 and 3200 repeats, respectively. The congenital nature of these CDM1 myoblasts was confirmed by the high methylation level at the DMPK locus. Our results indicated that abnormal autophagy was independent of myogenic differentiation, as CDM13 myoblasts differentiated as well as control myoblasts but underwent autophagy like CDM15, displaying impaired differentiation. miRNA expression profiles revealed that CDM15 myoblasts failed to upregulate the complex network of myo-miRNAs under MYOD and MEF2A control, while this network was upregulated in CDM13 myoblasts. Interestingly, the abnormal differentiation of CDM15 myoblasts was associated with cellular stress accompanied by the induction of the interferon type 1 pathway (innate immune response). Indeed, inhibition of the interferon (IFN) type I pathway restores myogenic differentiation of CDM15 myoblasts, suggesting that the inappropriate activation of the innate immune response might contribute to impaired myogenic differentiation and severe muscle symptoms observed in some CDM1 patients. These findings open up the possibility of new therapeutic approaches to treat CDM1.
Collapse
|
26
|
Flack A, Persons AL, Kousik SM, Celeste Napier T, Moszczynska A. Self-administration of methamphetamine alters gut biomarkers of toxicity. Eur J Neurosci 2018; 46:1918-1932. [PMID: 28661099 DOI: 10.1111/ejn.13630] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 12/13/2022]
Abstract
Methamphetamine (METH) is a highly abused psychostimulant that is associated with an increased risk for developing Parkinson's disease (PD). This enhanced vulnerability likely relates to the toxic effects of METH that overlap with PD pathology, for example, aberrant functioning of α-synuclein and parkin. In PD, peripheral factors are thought to contribute to central nervous system (CNS) degeneration. For example, α-synuclein levels in the enteric nervous system (ENS) are elevated, and this precedes the onset of motor symptoms. It remains unclear whether neurons of the ENS, particularly catecholaminergic neurons, exhibit signs of METH-induced toxicity as seen in the CNS. The aim of this study was to determine whether self-administered METH altered the levels of α-synuclein, parkin, tyrosine hydroxylase (TH), and dopamine-β-hydroxylase (DβH) in the myenteric plexus of the distal colon ENS. Young adult male Sprague-Dawley rats self-administered METH for 3 h per day for 14 days and controls were saline-yoked. Distal colon tissue was collected at 1, 14, or 56 days after the last operant session. Levels of α-synuclein were increased, while levels of parkin, TH, and DβH were decreased in the myenteric plexus in the METH-exposed rats at 1 day following the last operant session and returned to the control levels after 14 or 56 days of forced abstinence. The changes were not confined to neurofilament-positive neurons. These results suggest that colon biomarkers may provide early indications of METH-induced neurotoxicity, particularly in young chronic METH users who may be more susceptible to progression to PD later in life.
Collapse
Affiliation(s)
- Amanda Flack
- Department of Pharmaceutical Sciences, Wayne State University, Eugene Applebaum College of Pharmaceutical Sciences, Detroit, MI, 48201, USA
| | - Amanda L Persons
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, USA.,Department of Pharmacology, Rush University Medial Center, Chicago, IL, USA
| | - Sharanya M Kousik
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, USA.,Department of Pharmacology, Rush University Medial Center, Chicago, IL, USA
| | - T Celeste Napier
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, USA.,Department of Pharmacology, Rush University Medial Center, Chicago, IL, USA.,Department of Psychiatry, Rush University Medical Center, Chicago, IL, USA
| | - Anna Moszczynska
- Department of Pharmaceutical Sciences, Wayne State University, Eugene Applebaum College of Pharmaceutical Sciences, Detroit, MI, 48201, USA
| |
Collapse
|
27
|
Ferrucci M, Biagioni F, Ryskalin L, Limanaqi F, Gambardella S, Frati A, Fornai F. Ambiguous Effects of Autophagy Activation Following Hypoperfusion/Ischemia. Int J Mol Sci 2018; 19:ijms19092756. [PMID: 30217100 PMCID: PMC6163197 DOI: 10.3390/ijms19092756] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 01/07/2023] Open
Abstract
Autophagy primarily works to counteract nutrient deprivation that is strongly engaged during starvation and hypoxia, which happens in hypoperfusion. Nonetheless, autophagy is slightly active even in baseline conditions, when it is useful to remove aged proteins and organelles. This is critical when the mitochondria and/or proteins are damaged by toxic stimuli. In the present review, we discuss to that extent the recruitment of autophagy is beneficial in counteracting brain hypoperfusion or, vice-versa, its overactivity may per se be detrimental for cell survival. While analyzing these opposite effects, it turns out that the autophagy activity is likely not to be simply good or bad for cell survival, but its role varies depending on the timing and amount of autophagy activation. This calls for the need for an appropriate autophagy tuning to guarantee a beneficial effect on cell survival. Therefore, the present article draws a theoretical pattern of autophagy activation, which is hypothesized to define the appropriate timing and intensity, which should mirrors the duration and severity of brain hypoperfusion. The need for a fine tuning of the autophagy activation may explain why confounding outcomes occur when autophagy is studied using a rather simplistic approach.
Collapse
Affiliation(s)
- Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | | | - Larisa Ryskalin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | | | | | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
- IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli (IS), Italy.
| |
Collapse
|
28
|
Ryskalin L, Limanaqi F, Frati A, Busceti CL, Fornai F. mTOR-Related Brain Dysfunctions in Neuropsychiatric Disorders. Int J Mol Sci 2018; 19:ijms19082226. [PMID: 30061532 PMCID: PMC6121884 DOI: 10.3390/ijms19082226] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) is an ubiquitously expressed serine-threonine kinase, which senses and integrates several intracellular and environmental cues to orchestrate major processes such as cell growth and metabolism. Altered mTOR signalling is associated with brain malformation and neurological disorders. Emerging evidence indicates that even subtle defects in the mTOR pathway may produce severe effects, which are evident as neurological and psychiatric disorders. On the other hand, administration of mTOR inhibitors may be beneficial for a variety of neuropsychiatric alterations encompassing neurodegeneration, brain tumors, brain ischemia, epilepsy, autism, mood disorders, drugs of abuse, and schizophrenia. mTOR has been widely implicated in synaptic plasticity and autophagy activation. This review addresses the role of mTOR-dependent autophagy dysfunction in a variety of neuropsychiatric disorders, to focus mainly on psychiatric syndromes including schizophrenia and drug addiction. For instance, amphetamines-induced addiction fairly overlaps with some neuropsychiatric disorders including neurodegeneration and schizophrenia. For this reason, in the present review, a special emphasis is placed on the role of mTOR on methamphetamine-induced brain alterations.
Collapse
Affiliation(s)
- Larisa Ryskalin
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Fiona Limanaqi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | | | | | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Isernia, Italy.
| |
Collapse
|
29
|
Epigenetic Effects Induced by Methamphetamine and Methamphetamine-Dependent Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4982453. [PMID: 30140365 PMCID: PMC6081569 DOI: 10.1155/2018/4982453] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/10/2018] [Indexed: 12/21/2022]
Abstract
Methamphetamine is a widely abused drug, which possesses neurotoxic activity and powerful addictive effects. Understanding methamphetamine toxicity is key beyond the field of drug abuse since it allows getting an insight into the molecular mechanisms which operate in a variety of neuropsychiatric disorders. In fact, key alterations produced by methamphetamine involve dopamine neurotransmission in a way, which is reminiscent of spontaneous neurodegeneration and psychiatric schizophrenia. Thus, understanding the molecular mechanisms operated by methamphetamine represents a wide window to understand both the addicted brain and a variety of neuropsychiatric disorders. This overlapping, which is already present when looking at the molecular and cellular events promoted immediately after methamphetamine intake, becomes impressive when plastic changes induced in the brain of methamphetamine-addicted patients are considered. Thus, the present manuscript is an attempt to encompass all the molecular events starting at the presynaptic dopamine terminals to reach the nucleus of postsynaptic neurons to explain how specific neurotransmitters and signaling cascades produce persistent genetic modifications, which shift neuronal phenotype and induce behavioral alterations. A special emphasis is posed on disclosing those early and delayed molecular events, which translate an altered neurotransmitter function into epigenetic events, which are derived from the translation of postsynaptic noncanonical signaling into altered gene regulation. All epigenetic effects are considered in light of their persistent changes induced in the postsynaptic neurons including sensitization and desensitization, priming, and shift of neuronal phenotype.
Collapse
|
30
|
Wang Y, Gu YH, Liang LY, Liu M, Jiang B, Zhu MJ, Wang X, Shi L. Concurrence of autophagy with apoptosis in alveolar epithelial cells contributes to chronic pulmonary toxicity induced by methamphetamine. Cell Prolif 2018; 51:e12476. [PMID: 29956395 DOI: 10.1111/cpr.12476] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/21/2018] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES Methamphetamine (MA) abuse evokes pulmonary toxicity. The aim of our study is to investigate if autophagy is induced by MA and if autophagy-initiated apoptosis in alveolar epithelial cells is involved in MA-induced chronic pulmonary toxicity. MATERIALS AND METHODS The rats in Control group and MA group were tested by Doppler and HE staining. The alveolar epithelial cells were treated with MA, following by western blot, RT-PCR and immunofluorescence assay. RESULTS Chronic exposure to MA resulted in lower growth ratio of weight and in higher heart rate and peak blood flow velocity of the main pulmonary artery of rats. MA induced infiltration of inflammatory cells in lungs, more compact lung parenchyma, thickened alveolar septum and reduction in the number of alveolar sacs. In alveolar epithelial cells, the autophagy marker LC3 and per cent of cells containing LC3-positive autophagosome were significantly increased. MA dose dependently suppressed the phosphorylation of mTOR to inactivate mTOR, elicited autophagy regulatory proteins LC3 and Beclin-1, accelerated the transformation from LC3 I to LC3 II and initiated apoptosis by decreasing Bcl-2 and increasing Bax, Bax/Bcl-2 and cleaved Caspase 3. The above results suggest that sustained autophagy was induced by long-term exposure to MA and that the increased Beclin-1 autophagy initiated apoptosis in alveolar epithelial cells. CONCLUSIONS Concurrence of autophagy with apoptosis in alveolar epithelial cells contributes to chronic pulmonary toxicity induced by MA.
Collapse
Affiliation(s)
- Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yu-Han Gu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Li-Ye Liang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ming Liu
- Department of Drug Control, China Criminal Police University, Shenyang, China
| | - Bin Jiang
- Department of Cardiovascular Ultrasound, The First Hospital, China Medical University, Shenyang, China
| | - Mei-Jia Zhu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Xin Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Lin Shi
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| |
Collapse
|
31
|
Ryskalin L, Busceti CL, Limanaqi F, Biagioni F, Gambardella S, Fornai F. A Focus on the Beneficial Effects of Alpha Synuclein and a Re-Appraisal of Synucleinopathies. Curr Protein Pept Sci 2018; 19:598-611. [PMID: 29150919 PMCID: PMC5925871 DOI: 10.2174/1389203718666171117110028] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/03/2017] [Accepted: 11/13/2017] [Indexed: 01/01/2023]
Abstract
Alpha synuclein (α-syn) belongs to a class of proteins which are commonly considered to play a detrimental role in neuronal survival. This assumption is based on the occurrence of a severe neuronal degeneration in patients carrying a multiplication of the α-syn gene (SNCA) and in a variety of experimental models, where overexpression of α-syn leads to cell death and neurological impairment. In these conditions, a higher amount of normally structured α-syn produces a damage, which is even worse compared with that produced by α-syn owning an abnormal structure (as occurring following point gene mutations). In line with this, knocking out the expression of α-syn is reported to protect from specific neurotoxins such as 1-methyl, 4-phenyl 1,2,3,6-tetrahydropyridine (MPTP). In the present review we briefly discuss these well-known detrimental effects but we focus on findings showing that, in specific conditions α-syn is beneficial for cell survival. This occurs during methamphetamine intoxication which is counteracted by endogenous α-syn. Similarly, the dysfunction of the chaperone cysteine-string protein- alpha leads to cell pathology which is counteracted by over-expressing α-syn. In line with this, an increased expression of α-syn protects against oxidative damage produced by dopamine. Remarkably, when the lack of α-syn is combined with a depletion of β- and γ- synucleins, alterations in brain structure and function occur. This review tries to balance the evidence showing a beneficial effect with the bulk of data reporting a detrimental effect of endogenous α-syn. The specific role of α-syn as a chaperone protein is discussed to explain such a dual effect.
Collapse
Affiliation(s)
- Larisa Ryskalin
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126Pisa, Italy
| | - Carla L. Busceti
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Pozzilli, Isernia, Italy
| | - Fiona Limanaqi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126Pisa, Italy
| | | | | | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126Pisa, Italy
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Pozzilli, Isernia, Italy
| |
Collapse
|
32
|
Xu X, Huang E, Tai Y, Zhao X, Chen X, Chen C, Chen R, Liu C, Lin Z, Wang H, Xie WB. Nupr1 Modulates Methamphetamine-Induced Dopaminergic Neuronal Apoptosis and Autophagy through CHOP-Trib3-Mediated Endoplasmic Reticulum Stress Signaling Pathway. Front Mol Neurosci 2017; 10:203. [PMID: 28694771 PMCID: PMC5483452 DOI: 10.3389/fnmol.2017.00203] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/08/2017] [Indexed: 12/13/2022] Open
Abstract
Methamphetamine (METH) is an illegal and widely abused psychoactive stimulant. METH exposure causes detrimental effects on multiple organ systems, primarily the nervous system, especially dopaminergic pathways, in both laboratory animals and humans. In this study, we hypothesized that Nuclear protein 1 (Nupr1/com1/p8) is involved in METH-induced neuronal apoptosis and autophagy through endoplasmic reticulum (ER) stress signaling pathway. To test this hypothesis, we measured the expression levels of Nupr1, ER stress protein markers CHOP and Trib3, apoptosis-related protein markers cleaved-caspase3 and PARP, as well as autophagy-related protein markers LC3 and Beclin-1 in brain tissues of adult male Sprague-Dawley (SD) rats, rat primary cultured neurons and the rat adrenal pheochromocytoma cells (PC12 cells) after METH exposure. We also determined the effects of METH exposure on the expression of these proteins after silencing Nupr1, CHOP, or Trib3 expression with synthetic small hairpin RNA (shRNA) or siRNA in vitro, and after silencing Nupr1 in the striatum of rats by injecting lentivirus containing shRNA sequence targeting Nupr1 gene to rat striatum. The results showed that METH exposure increased Nupr1 expression that was accompanied with increased expression of ER stress protein markers CHOP and Trib3, and also led to apoptosis and autophagy in rat primary neurons and in PC12 cells after 24 h exposure (3.0 mM), and in the prefrontal cortex and striatum of rats after repeated intraperitoneal injections (15 mg/kg × 8 injections at 12 h intervals). Silencing of Nupr1 expression partly reduced METH-induced apoptosis and autophagy in vitro and in vivo. These results suggest that Nupr1 plays an essential role in METH-caused neuronal apoptosis and autophagy at relatively higher doses and may be a potential therapeutic target in high-dose METH-induced neurotoxicity.
Collapse
Affiliation(s)
- Xiang Xu
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China.,School of Forensic Medicine, Wannan Medical CollegeWuhu, China
| | - Enping Huang
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China
| | - Yunchun Tai
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China
| | - Xu Zhao
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China
| | - Xuebing Chen
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China
| | - Chuanxiang Chen
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China
| | - Rui Chen
- Department of Forensic Medicine, Guangdong Medical UniversityDongguan, China
| | - Chao Liu
- Guangzhou Forensic Science InstituteGuangzhou, China
| | - Zhoumeng Lin
- Institute of Computational Comparative Medicine and Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State UniversityManhattan, KS, United States
| | - Huijun Wang
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China
| | - Wei-Bing Xie
- School of Forensic Medicine, Southern Medical UniversityGuangzhou, China
| |
Collapse
|
33
|
Shin EJ, Tran HQ, Nguyen PT, Jeong JH, Nah SY, Jang CG, Nabeshima T, Kim HC. Role of Mitochondria in Methamphetamine-Induced Dopaminergic Neurotoxicity: Involvement in Oxidative Stress, Neuroinflammation, and Pro-apoptosis-A Review. Neurochem Res 2017; 43:66-78. [PMID: 28589520 DOI: 10.1007/s11064-017-2318-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/25/2017] [Accepted: 05/30/2017] [Indexed: 01/08/2023]
Abstract
Methamphetamine (MA), an amphetamine-type psychostimulant, is associated with dopaminergic toxicity and has a high abuse potential. Numerous in vivo and in vitro studies have suggested that impaired mitochondria are critical in dopaminergic toxicity induced by MA. Mitochondria are important energy-producing organelles with dynamic nature. Evidence indicated that exposure to MA can disturb mitochondrial energetic metabolism by inhibiting the Krebs cycle and electron transport chain. Alterations in mitochondrial dynamic processes, including mitochondrial biogenesis, mitophagy, and fusion/fission, have recently been shown to contribute to dopaminergic toxicity induced by MA. Furthermore, it was demonstrated that MA-induced mitochondrial impairment enhances susceptibility to oxidative stress, pro-apoptosis, and neuroinflammation in a positive feedback loop. Protein kinase Cδ has emerged as a potential mediator between mitochondrial impairment and oxidative stress, pro-apoptosis, or neuroinflammation in MA neurotoxicity. Understanding the role and underlying mechanism of mitochondrial impairment could provide a molecular target to prevent or alleviate dopaminergic toxicity induced by MA.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Hai-Quyen Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Phuong-Tram Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Toyoake, 470-1192, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
34
|
Roohbakhsh A, Shirani K, Karimi G. Methamphetamine-induced toxicity: The role of autophagy? Chem Biol Interact 2016; 260:163-167. [PMID: 27746146 DOI: 10.1016/j.cbi.2016.10.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 10/04/2016] [Accepted: 10/12/2016] [Indexed: 12/11/2022]
Abstract
Methamphetamine (METH) is a highly potent and addictive drug with major medical, psychiatric, cognitive, socioeconomic, and legal consequences. It is well absorbed following different routes of administration and distributed throughout the body. METH is known as psychomotor stimulant with potent physiological outcomes on peripheral and central nervous systems, resulting in physical and psychological disorders. Autophagy is a highly conserved and regulated catabolic pathway which is critical for maintaining cellular energy homeostasis and regulating cell growth. The mechanism of autophagy has attracted considerable attention in the last few years because of its recognition as a vital arbiter of death/survival decisions in cells and as a critical defense mechanism in undesirable physiological conditions. The purpose of the current article was to review available evidence to find a relationship between METH toxicity and mechanisms associated with autophagy in different organs.
Collapse
Affiliation(s)
- Ali Roohbakhsh
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Neurocognitive Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kobra Shirani
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Neurocognitive Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
35
|
Chaturvedi SK, Siddiqi MK, Alam P, Khan RH. Protein misfolding and aggregation: Mechanism, factors and detection. Process Biochem 2016. [DOI: 10.1016/j.procbio.2016.05.015] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
36
|
Lenzi P, Lazzeri G, Biagioni F, Busceti CL, Gambardella S, Salvetti A, Fornai F. The Autophagoproteasome a Novel Cell Clearing Organelle in Baseline and Stimulated Conditions. Front Neuroanat 2016; 10:78. [PMID: 27493626 PMCID: PMC4955296 DOI: 10.3389/fnana.2016.00078] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 07/05/2016] [Indexed: 12/24/2022] Open
Abstract
Protein clearing pathways named autophagy (ATG) and ubiquitin proteasome (UP) control homeostasis within eukaryotic cells, while their dysfunction produces neurodegeneration. These pathways are viewed as distinct biochemical cascades occurring within specific cytosolic compartments owing pathway-specific enzymatic activity. Recent data strongly challenged the concept of two morphologically distinct and functionally segregated compartments. In fact, preliminary evidence suggests the convergence of these pathways to form a novel organelle named autophagoproteasome. This is characterized in the present study by using a cell line where, mTOR activity is upregulated and autophagy is suppressed. This was reversed dose-dependently by administering the mTOR inhibitor rapamycin. Thus, we could study autophagoproteasomes when autophagy was either suppressed or stimulated. The occurrence of autophagoproteasome was shown also in non-human cell lines. Ultrastructural morphometry, based on the stochiometric binding of immunogold particles allowed the quantitative evaluation of ATG and UP component within autophagoproteasomes. The number of autophagoproteasomes increases following mTOR inhibition. Similarly, mTOR inhibition produces overexpression of both LC3 and P20S particles. This is confirmed by the fact that the ratio of free vs. autophagosome-bound LC3 is similar to that measured for P20S, both in baseline conditions and following mTOR inhibition. Remarkably, within autophagoproteasomes there is a slight prevalence of ATG compared with UP components for low rapamycin doses, whereas for higher rapamycin doses UP increases more than ATG. While LC3 is widely present within cytosol, UP is strongly polarized within autophagoproteasomes. These fine details were evident at electron microscopy but could not be deciphered by using confocal microscopy. Despite its morphological novelty autophagoproteasomes appear in the natural site where clearing pathways (once believed to be anatomically segregated) co-exist and they are likely to interact at molecular level. In fact, LC3 and P20S co-immunoprecipitate, suggesting a specific binding and functional interplay, which may be altered by inhibiting mTOR. In summary, ATG and UP often represent two facets of a single organelle, in which unexpected amount of enzymatic activity should be available. Thus, autophagoproteasome may represent a sophisticated ultimate clearing apparatus.
Collapse
Affiliation(s)
- Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa Pisa, Italy
| | - Gloria Lazzeri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa Pisa, Italy
| | - Francesca Biagioni
- Istituti di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.), Neuromed Pozzilli, Italy
| | - Carla L Busceti
- Istituti di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.), Neuromed Pozzilli, Italy
| | - Stefano Gambardella
- Istituti di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.), Neuromed Pozzilli, Italy
| | - Alessandra Salvetti
- Department of Clinical and Experimental Medicine, University of Pisa Pisa, Italy
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of PisaPisa, Italy; Istituti di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.), NeuromedPozzilli, Italy
| |
Collapse
|
37
|
Effects of DDIT4 in Methamphetamine-Induced Autophagy and Apoptosis in Dopaminergic Neurons. Mol Neurobiol 2016; 54:1642-1660. [PMID: 26873849 DOI: 10.1007/s12035-015-9637-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 12/15/2015] [Indexed: 01/14/2023]
Abstract
Methamphetamine (METH) is an illicit psychoactive drug that can cause a variety of detrimental effects to the nervous system, especially dopaminergic pathways. We hypothesized that DNA damage-inducible transcript 4 (DDIT4) is involved in METH-induced dopaminergic neuronal autophagy and apoptosis. To test the hypothesis, we determined changes of DDIT4 protein expression and the level of autophagy in rat catecholaminergic PC12 cells and human dopaminergic SH-SY5Y cells, and in the hippocampus, prefrontal cortex, and striatum of Sprague Dawley rats exposed to METH. We also examined the effects of silencing DDIT4 expression on METH-induced dopaminergic neuronal autophagy using fluorescence microscopy and electron microscopy. Flow cytometry and Western blot were used to determine apoptosis and the expression of apoptotic markers (cleaved caspase-3 and cleaved PARP) after blocking DDIT4 expression in PC12 cells and SH-SY5Y cells with synthetic siRNA, as well as in the striatum of rats by injecting LV-shDDIT4 lentivirus using a stereotaxic positioning system. Our results showed that METH exposure increased DDIT4 expression that was accompanied with increased autophagy and apoptosis in PC12 cells (3 mM) and SH-SY5Y cells (2 mM), and in the hippocampus, prefrontal cortex, and striatum of rats. Inhibition of DDIT4 expression reduced METH-induced autophagy and apoptosis in vitro and in vivo. However, DDIT4-related effects were not observed at a low concentration of METH (1 μM). These results suggest that DDIT4 plays an essential role in METH-induced dopaminergic neuronal autophagy and apoptosis at higher doses and may be a potential gene target for therapeutics in high-dose METH-induced neurotoxicity.
Collapse
|
38
|
Wang SY, Chen L, Xue Y, Xia YJ. Substance P prevents 1-methyl-4-phenylpyridinium-induced cytotoxicity through inhibition of apoptosis via neurokinin-1 receptors in MES23.5 cells. Mol Med Rep 2015; 12:8085-92. [PMID: 26497672 DOI: 10.3892/mmr.2015.4464] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 08/25/2015] [Indexed: 11/06/2022] Open
Abstract
[Sar9, Met(O2)11] termed Substance P (SP), is an effective and selective agonist for the neurokinin‑1 (NK‑1) receptors, which are synthetic peptides, similar in structure to SP. SP is an important neurotransmitter or neuromodulator mediated by neurokinin receptors, namely the SP receptor in the central nervous system. The excitatory effects induced by SP may be selectively inhibited by a neurokinin‑1 receptor antagonist, such as SR140333B. It has been proposed that Parkinson's disease (PD) is primarily caused by the loss of trophic peptidergic neurotransmitter, possibly SP, which may lead to the degeneration of neurons. In previous studies, 1‑methyl‑4‑phenylpyridinium (MPP+) has been frequently utilized to establish animal or cell models of PD. In the present study, to further investigate the effects of SP in PD, MPP+ was employed to investigate the promising anti‑apoptotic effects of SP, and examine the underlying mechanisms of the pathology in the MES23.5 dopaminergic cell line. The results indicated that MPP+‑triggered apoptosis was prevented by treatment with SP. SP treatment also decreased the MPP+‑triggered Ca2+ influx, caspase‑3 re‑activity, reactive oxygen species production and mitochondrial membrane potential decrease. Treatment with MPP+ also induced phosphorylation of c‑Jun N‑terminal kinase and p38 mitogen‑activated protein kinase. In addition, treatment with SP inhibited the MPP+‑triggered neurotoxicity in MES23.5 cells. However, no changes were observed in SR140333B+SP+MPP+‑treated MES23.5 cell lines. In conclusion, SP could protect the cells from MPP+‑induced cytotoxicity by inhibiting the apoptosis via NK-1 receptors.
Collapse
Affiliation(s)
- Shuang-Yan Wang
- Department of Physiology, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Lei Chen
- Department of Physiology, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Yan Xue
- Department of Physiology, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Yu-Jun Xia
- Department of Anatomy, Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
39
|
Miller JD, Ganat YM, Kishinevsky S, Bowman RL, Liu B, Tu EY, Mandal P, Vera E, Shim JW, Kriks S, Taldone T, Fusaki N, Tomishima MJ, Krainc D, Milner TA, Rossi DJ, Studer L. Human iPSC-based modeling of late-onset disease via progerin-induced aging. Cell Stem Cell 2013; 13:691-705. [PMID: 24315443 PMCID: PMC4153390 DOI: 10.1016/j.stem.2013.11.006] [Citation(s) in RCA: 568] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 09/27/2013] [Accepted: 11/05/2013] [Indexed: 12/15/2022]
Abstract
Reprogramming somatic cells to induced pluripotent stem cells (iPSCs) resets their identity back to an embryonic age and, thus, presents a significant hurdle for modeling late-onset disorders. In this study, we describe a strategy for inducing aging-related features in human iPSC-derived lineages and apply it to the modeling of Parkinson's disease (PD). Our approach involves expression of progerin, a truncated form of lamin A associated with premature aging. We found that expression of progerin in iPSC-derived fibroblasts and neurons induces multiple aging-related markers and characteristics, including dopamine-specific phenotypes such as neuromelanin accumulation. Induced aging in PD iPSC-derived dopamine neurons revealed disease phenotypes that require both aging and genetic susceptibility, such as pronounced dendrite degeneration, progressive loss of tyrosine hydroxylase (TH) expression, and enlarged mitochondria or Lewy-body-precursor inclusions. Thus, our study suggests that progerin-induced aging can be used to reveal late-onset age-related disease features in hiPSC-based disease models.
Collapse
Affiliation(s)
- Justine D. Miller
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
- Gerstner Sloan-Kettering Graduate School, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
| | - Yosif M. Ganat
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
| | - Sarah Kishinevsky
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
| | - Robert L. Bowman
- Gerstner Sloan-Kettering Graduate School, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
- Cancer Biology and Genetics Program, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
| | - Becky Liu
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
| | - Edmund Y. Tu
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
| | - Pankaj Mandal
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Elsa Vera
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
| | - Jae-won Shim
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
| | - Sonja Kriks
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
| | - Tony Taldone
- Molecular Pharmacology & Chemistry, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
| | - Noemi Fusaki
- DNAVEC Corporation, Tsukuba, Ibaraki 300-2611, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Mark J. Tomishima
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
| | - Dimitri Krainc
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Massachusetts General Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Teresa A. Milner
- Brain and Mind Research Institute, Weill Cornell Medical College, 407 East 61st Street, New York, NY 10065, USA
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Ave, New York, NY 10065, USA
| | - Derrick J. Rossi
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, 1275 York Ave, New York, NY 10065, USA
| |
Collapse
|
40
|
Rendón WO, Martínez-Alonso E, Tomás M, Martínez-Martínez N, Martínez-Menárguez JA. Golgi fragmentation is Rab and SNARE dependent in cellular models of Parkinson’s disease. Histochem Cell Biol 2012; 139:671-84. [DOI: 10.1007/s00418-012-1059-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2012] [Indexed: 10/27/2022]
|
41
|
Dimatelis JJ, Russell VA, Stein DJ, Daniels WM. Effects of maternal separation and methamphetamine exposure on protein expression in the nucleus accumbens shell and core. Metab Brain Dis 2012; 27:363-75. [PMID: 22451087 DOI: 10.1007/s11011-012-9295-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 03/15/2012] [Indexed: 12/14/2022]
Abstract
Early life adversity has been suggested to predispose an individual to later drug abuse. The core and shell sub-regions of the nucleus accumbens are differentially affected by both stressors and methamphetamine. This study aimed to characterize and quantify methamphetamine-induced protein expression in the shell and core of the nucleus accumbens in animals exposed to maternal separation during early development. Isobaric tagging (iTRAQ) which enables simultaneous identification and quantification of peptides with tandem mass spectrometry (MS/MS) was used. We found that maternal separation altered more proteins involved in structure and redox regulation in the shell than in the core of the nucleus accumbens, and that maternal separation and methamphetamine had differential effects on signaling proteins in the shell and core. Compared to maternal separation or methamphetamine alone, the maternal separation/methamphetamine combination altered more proteins involved in energy metabolism, redox regulatory processes and neurotrophic proteins. Methamphetamine treatment of rats subjected to maternal separation caused a reduction of cytoskeletal proteins in the shell and altered cytoskeletal, signaling, energy metabolism and redox proteins in the core. Comparison of maternal separation/methamphetamine to methamphetamine alone resulted in decreased cytoskeletal proteins in both the shell and core and increased neurotrophic proteins in the core. This study confirms that both early life stress and methamphetamine differentially affect the shell and core of the nucleus accumbens and demonstrates that the combination of early life adversity and later methamphetamine use results in more proteins being affected in the nucleus accumbens than either treatment alone.
Collapse
Affiliation(s)
- J J Dimatelis
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, 7925 Cape Town, South Africa.
| | | | | | | |
Collapse
|
42
|
Lin M, Chandramani-Shivalingappa P, Jin H, Ghosh A, Anantharam V, Ali S, Kanthasamy AG, Kanthasamy A. Methamphetamine-induced neurotoxicity linked to ubiquitin-proteasome system dysfunction and autophagy-related changes that can be modulated by protein kinase C delta in dopaminergic neuronal cells. Neuroscience 2012; 210:308-32. [PMID: 22445524 DOI: 10.1016/j.neuroscience.2012.03.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 02/09/2012] [Accepted: 03/04/2012] [Indexed: 01/07/2023]
Abstract
A compromised protein degradation machinery has been implicated in methamphetamine (MA)-induced neurodegeneration. However, the signaling mechanisms that induce autophagy and ubiquitin-proteasome system (UPS) dysfunction are not well understood. The present study investigates the contributions of protein kinase C delta (PKCδ)-mediated signaling events in MA-induced autophagy, UPS dysfunction, and cell death. Using an in vitro mesencephalic dopaminergic cell culture model, we demonstrate that MA-induced early induction of autophagy is associated with reduction in proteasomal function and concomitant dissipation of mitochondrial membrane potential (MMP), followed by significantly increased PKCδ activation, caspase-3 activation, accumulation of ubiquitin-positive aggregates and microtubule-associated light chain-3 (LC3-II) levels. Interestingly, siRNA-mediated knockdown of PKCδ or overexpression of cleavage-resistant mutant of PKCδ dramatically reduced MA-induced autophagy, proteasomal function, and associated accumulation of ubiquitinated protein aggregates, which closely paralleled cell survival. Importantly, when autophagy was inhibited either pharmacologically (3-MA) or genetically (siRNA-mediated silencing of LC3), the dopaminergic cells became sensitized to MA-induced apoptosis through caspase-3 activation. Conversely, overexpression of LC3 partially protected against MA-induced apoptotic cell death, suggesting a neuroprotective role for autophagy in MA-induced neurotoxicity. Notably, rat striatal tissue isolated from MA-treated rats also exhibited elevated LC3-II, ubiquitinated protein levels, and PKCδ cleavage. Taken together, our data demonstrate that MA-induced autophagy serves as an adaptive strategy for inhibiting mitochondria-mediated apoptotic cell death and degradation of aggregated proteins. Our results also suggest that the sustained activation of PKCδ leads to UPS dysfunction, resulting in the activation of caspase-3-mediated apoptotic cell death in the nigrostriatal dopaminergic system.
Collapse
Affiliation(s)
- M Lin
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Sae-Ung K, Uéda K, Govitrapong P, Phansuwan-Pujito P. Melatonin reduces the expression of alpha-synuclein in the dopamine containing neuronal regions of amphetamine-treated postnatal rats. J Pineal Res 2012; 52:128-37. [PMID: 21851386 DOI: 10.1111/j.1600-079x.2011.00927.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Alpha-synuclein (α-syn) is a neuronal protein that is involved in various degenerative disorders such as Parkinson's disease. It is found in the presynaptic terminals and perinuclear zones of many brain regions. Amphetamine (AMPH), a psychostimulant drug abused progressively more commonly in recent years, has been known to induce neurotoxicity in the central dopaminergic pathway, which is associated with increased oxidative stress. Recently, AMPH has been shown to significantly increase the level of α-syn in dopaminergic neuroblastoma cell cultures. Melatonin is recognized as an antioxidant for the nervous system. This study tested whether melatonin can attenuate the effect of AMPH on the expression of α-syn in the dopaminergic pathway of the neonatal rat. Four-day old postnatal rats (P4) were injected subcutaneously with either AMPH (increasing dose, 5-10 mg/kg daily) alone or AMPH with melatonin (2 mg/kg) daily at 10:00 AM for 7 consecutive days. As determined using Western blot, the level of α-syn was significantly increased in the substantia nigra, dorsal striatum, nucleus accumbens, and prefrontal cortex of the AMPH-treated group, while melatonin treatment either prior to AMPH or alone decreased the accumulation of the protein to 77%, 96%, 78%, and 77% of the control value, respectively. Furthermore, an immunofluorescent study showed that the α-syn-immunoreactivity increased noticeably in the nuclei of cell bodies and nerve terminals of the AMPH-treated group. Again, melatonin lowered this immunoreactivity. These results indicate that melatonin has a direct or indirect effect in reducing the expression of α-syn in the postnatal rat. The exact mechanism of this mitigation should be further investigated.
Collapse
Affiliation(s)
- Kwankanit Sae-Ung
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Wattana, Bangkok, Thailand
| | | | | | | |
Collapse
|
44
|
Kanthasamy K, Gordon R, Jin H, Anantharam V, Ali S, Kanthasamy AG, Kanthasamy A. Neuroprotective effect of resveratrol against methamphetamine-induced dopaminergic apoptotic cell death in a cell culture model of neurotoxicity. Curr Neuropharmacol 2011; 9:49-53. [PMID: 21886561 PMCID: PMC3137200 DOI: 10.2174/157015911795017353] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 04/17/2010] [Accepted: 05/26/2010] [Indexed: 12/15/2022] Open
Abstract
A growing body of evidence suggests that oxidative stress-mediated cell death signaling mechanisms may exert neurotoxic effects of methamphetamine (MA)-induced dopaminergic neuronal loss. However, the means by which oxidative stress induced by MA causes neurodegeneration remains unclear. In recent years, resveratrol has garnered considerable attention owing to its antioxidant, anti-inflammatory, anti-aging, and neuroprotective properties. In the present study, we sought to investigate the neuroprotective effects of resveratrol against apoptotic cell death in a mesencephalic dopaminergic neuronal cell culture model of MA neurotoxicity. MA treatment in the N27 dopaminergic neuronal cell model produced a time-dependent activation of the apoptotic cascade involving caspase-3 and DNA fragmentation. We found that the caspase-3 activation preceded DNA fragmentation. Notably, treatment with resveratrol almost completely attenuated MA-induced caspase-3 activity, but only partially reduced apoptotic cell death. We conclude that the neuroprotective effect of resveratrol is at least in part mediated by suppression of caspase-3 dependent cell death pathways. Collectively, our results demonstrate that resveratrol can attenuate MA-induced apoptotic cell death and suggest that resveratrol or its analogs may have therapeutic benefits in mitigating MA-induced dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Kavin Kanthasamy
- Dept. of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011-1250
| | | | | | | | | | | | | |
Collapse
|
45
|
Scatena A, Fornai F, Trincavelli ML, Taliani S, Daniele S, Pugliesi I, Cosconati S, Martini C, Da Settimo F. 3-(Fur-2-yl)-10-(2-phenylethyl)-[1,2,4]triazino[4,3-a]benzimidazol-4(10H)-one, a novel adenosine receptor antagonist with A(2A)-mediated neuroprotective effects. ACS Chem Neurosci 2011; 2:526-35. [PMID: 22860174 DOI: 10.1021/cn200036s] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 06/10/2011] [Indexed: 12/20/2022] Open
Abstract
In this study, compound FTBI (3-(2-furyl)-10-(2-phenylethyl)[1,2,4]triazino[4,3-a]benzimidazol-4(10H)-one) was selected from a small library of triazinobenzimidazole derivatives as a potent A(2A) adenosine receptor (AR) antagonist and tested for its neuroprotective effects against two different kinds of dopaminergic neurotoxins, 1-methyl-4-phenylpyridinium (MPP+) and methamphetamine (METH), in rat PC12 and in human neuroblastoma SH-SY5Y cell lines. FTBI, in a concentration range corresponding to its affinity for A(2A) AR subtype, significantly increased the number of viable PC12 cells after their exposure to METH and, to a similar extent, to MPP+, as demonstrated in both trypan blue exclusion assay and in cytological staining. These neuroprotective effects were also observed with a classical A(2A) AR antagonist, ZM241385, and appeared to be completely counteracted by the AR agonist, NECA, supporting A(2A) ARs are directly involved in FTBI-mediated effects. Similarly, in human SH-SY5Y cells, FTBI was able to prevent cell toxicity induced by MPP+ and METH, showing that this A(2A) AR antagonist has a neuroprotective effect independently by the specific cell model. Altogether these results demonstrate that the A(2A) AR blockade mediates cell protection against neurotoxicity induced by dopaminergic neurotoxins in dopamine containing cells, supporting the potential use of A(2A) AR antagonists in dopaminergic degenerative diseases including Parkinson's disease.
Collapse
Affiliation(s)
- Alessia Scatena
- Dipartimento di Morfologia Umana e Biologia Applicata, Università di Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Francesco Fornai
- Dipartimento di Morfologia Umana e Biologia Applicata, Università di Pisa, Via Roma 55, 56126 Pisa, Italy
- Neurobiologia dei disturbi del movimento, IRCCS INM Neuromed, Via Atinense 18, 86077 Pozzilli, Isernia, Italy
| | - Maria Letizia Trincavelli
- Dipartimento di Psichiatria, Neurobiologia, Farmacologia e Biotecnologie, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Sabrina Taliani
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Simona Daniele
- Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
| | - Isabella Pugliesi
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Sandro Cosconati
- Dipartimento di Scienze Ambientali, Seconda Università di Napoli, Via Vivaldi 43, 81100 Caserta, Italy
| | - Claudia Martini
- Dipartimento di Psichiatria, Neurobiologia, Farmacologia e Biotecnologie, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Federico Da Settimo
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| |
Collapse
|
46
|
Tulloch I, Ghazaryan N, Mexhitaj I, Ordonez D, Angulo JA. Role of neurokinin-1 and dopamine receptors on the striatal methamphetamine-induced proliferation of new cells in mice. Brain Res 2011; 1399:33-9. [PMID: 21652034 PMCID: PMC3117948 DOI: 10.1016/j.brainres.2011.05.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 05/07/2011] [Accepted: 05/10/2011] [Indexed: 01/03/2023]
Abstract
A neurotoxic dose of methamphetamine (METH) induces the loss of some striatal neurons. Interestingly, the METH-induced apoptosis in the striatum is immediately followed by the generation of new cells (cytogenesis). In the present study, we investigated the role of the neurokinin-1, dopamine D1 and D2 receptors on the METH-induced cytogenesis. To that end, male mice were given a single injection (30 mg/kg, ip) or a binge of METH (10mg/kg, 4× at two-hour intervals, ip). BrdU (100mg/kg, ip) was given 36 h after the last injection of METH. Newly generated cells were detected by immunohistochemistry and cell counts were performed using unbiased computerized stereology. Either single or binge exposure to METH resulted in the generation of new cells. The single optimized dose was used for subsequent mechanistic studies. Pretreatment with the dopamine D1 receptor antagonist SCH23390 (0.1mg/kg, ip) 30 min prior to METH abrogated the METH-induced striatal cytogenesis. Pretreatment with the dopamine D2 receptor antagonist raclopride (1mg/kg, ip) failed to affect this phenomenon. Finally, pretreatment with the neurokinin-1 receptor antagonist WIN 51,708 (5mg/kg, ip) 30 min prior to METH abrogated the METH-induced cytogenesis. In conclusion, neurokinin-1 and dopamine D1 receptors are required for the METH-induced striatal cytogenesis while the D2 receptor is without effect.
Collapse
Affiliation(s)
- Ingrid Tulloch
- Hunter College, The City University of New York, New York NY
- The Graduate Center, The City University of New York, New York NY
| | - Nane Ghazaryan
- Hunter College, The City University of New York, New York NY
| | - Ina Mexhitaj
- Hunter College, The City University of New York, New York NY
| | - Dalila Ordonez
- Hunter College, The City University of New York, New York NY
| | - Jesus A. Angulo
- Hunter College, The City University of New York, New York NY
- The Graduate Center, The City University of New York, New York NY
| |
Collapse
|
47
|
Lipski J, Nistico R, Berretta N, Guatteo E, Bernardi G, Mercuri NB. L-DOPA: a scapegoat for accelerated neurodegeneration in Parkinson's disease? Prog Neurobiol 2011; 94:389-407. [PMID: 21723913 DOI: 10.1016/j.pneurobio.2011.06.005] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 06/17/2011] [Accepted: 06/17/2011] [Indexed: 12/21/2022]
Abstract
There is consensus that amelioration of the motor symptoms of Parkinson's disease is most effective with L-DOPA (levodopa). However, this necessary therapeutic step is biased by an enduring belief that L-DOPA is toxic to the remaining substantia nigra dopaminergic neurons by itself, or by specific metabolites such as dopamine. The concept of L-DOPA toxicity originated from pre-clinical studies conducted mainly in cell culture, demonstrating that L-DOPA or its derivatives damage dopaminergic neurons due to oxidative stress and other mechanisms. However, the in vitro data remain controversial as some studies showed neuroprotective, rather than toxic action of the drug. The relevance of this debate needs to be considered in the context of the studies conducted on animals and in clinical trials that do not provide convincing evidence for L-DOPA toxicity in vivo. This review presents the current views on the pathophysiology of Parkinson's disease, focusing on mitochondrial dysfunction and oxidative/proteolytic stress, the factors that can be affected by L-DOPA or its metabolites. We then critically discuss the evidence supporting the two opposing views on the effects of L-DOPA in vitro, as well as the animal and human data. We also address the problem of inadequate experimental models used in these studies. L-DOPA remains the symptomatic 'hero' of Parkinson's disease. Whether it contributes to degeneration of nigral dopaminergic neurons, or is a 'scapegoat' for explaining undesirable or unexpected effects of the treatment, remains a hotly debated topic.
Collapse
Affiliation(s)
- Janusz Lipski
- Department of Physiology and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd., Auckland 1142, New Zealand.
| | | | | | | | | | | |
Collapse
|
48
|
Funk KE, Mrak RE, Kuret J. Granulovacuolar degeneration (GVD) bodies of Alzheimer's disease (AD) resemble late-stage autophagic organelles. Neuropathol Appl Neurobiol 2011; 37:295-306. [PMID: 20946470 DOI: 10.1111/j.1365-2990.2010.01135.x] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
AIMS Granulovacuolar degeneration involves the accumulation of large, double membrane-bound bodies within certain neurones during the course of Alzheimer's disease (AD) and other adult-onset dementias. Because of the two-layer membrane morphology, it has been proposed that the bodies are related to autophagic organelles. The aim of this study was to test this hypothesis, and determine the approximate stage at which the pathway stalls in AD. METHODS Spatial colocalization of autophagic and endocytic markers with casein kinase 1 delta, a marker for granulovacuolar degeneration (GVD) bodies, was evaluated in hippocampal sections prepared from post mortem Braak stage IV and V AD cases using double-label confocal fluorescence microscopy. RESULTS GVD bodies colocalized weakly with early-stage autophagy markers LC3 and p62, but strongly with late-stage marker lysosome-associated membrane protein 1 (LAMP1), which decorated their surrounding membranes. GVD bodies also colocalized strongly with charged multivesicular body protein 2B (CHMP2B), which colocalized with the core granule, but less strongly with lysosomal marker cathepsin D. CONCLUSIONS The resultant immunohistochemical signature suggests that granulovacuolar degeneration bodies (GVBs) do contain late-stage autophagic markers, and accumulate at the nexus of autophagic and endocytic pathways. The data further suggest that failure to complete autolysosome formation may be an important correlate of GVB accumulation.
Collapse
Affiliation(s)
- K E Funk
- Department of Molecular and Cellular Biochemistry, Center for Molecular Neurobiology, The Ohio State University, Columbus, USA
| | | | | |
Collapse
|
49
|
Esteves AR, Arduíno DM, Silva DFF, Oliveira CR, Cardoso SM. Mitochondrial Dysfunction: The Road to Alpha-Synuclein Oligomerization in PD. PARKINSON'S DISEASE 2011; 2011:693761. [PMID: 21318163 PMCID: PMC3026982 DOI: 10.4061/2011/693761] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 12/21/2010] [Accepted: 12/27/2010] [Indexed: 12/21/2022]
Abstract
While the etiology of Parkinson's disease remains largely elusive, there is accumulating evidence suggesting that mitochondrial dysfunction occurs prior to the onset of symptoms in Parkinson's disease. Mitochondria are remarkably primed to play a vital role in neuronal cell survival since they are key regulators of energy metabolism (as ATP producers), of intracellular calcium homeostasis, of NAD(+)/NADH ratio, and of endogenous reactive oxygen species production and programmed cell death. In this paper, we focus on mitochondrial dysfunction-mediated alpha-synuclein aggregation. We highlight some of the findings that provide proof of evidence for a mitochondrial metabolism control in Parkinson's disease, namely, mitochondrial regulation of microtubule-dependent cellular traffic and autophagic lysosomal pathway. The knowledge that microtubule alterations may lead to autophagic deficiency and may compromise the cellular degradation mechanisms that culminate in the progressive accumulation of aberrant protein aggregates shields new insights to the way we address Parkinson's disease. In line with this knowledge, an innovative window for new therapeutic strategies aimed to restore microtubule network may be unlocked.
Collapse
Affiliation(s)
- A. R. Esteves
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra, 3004 Coimbra, Portugal
| | - D. M. Arduíno
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra, 3004 Coimbra, Portugal
| | - D. F. F. Silva
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra, 3004 Coimbra, Portugal
| | - C. R. Oliveira
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra, 3004 Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, 3000 Coimbra, Portugal
| | - S. M. Cardoso
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra, 3004 Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, 3000 Coimbra, Portugal
| |
Collapse
|
50
|
Burguillos MA, Hajji N, Englund E, Persson A, Cenci AM, Machado A, Cano J, Joseph B, Venero JL. Apoptosis-inducing factor mediates dopaminergic cell death in response to LPS-induced inflammatory stimulus: evidence in Parkinson's disease patients. Neurobiol Dis 2011; 41:177-88. [PMID: 20850531 DOI: 10.1016/j.nbd.2010.09.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Revised: 09/06/2010] [Accepted: 09/09/2010] [Indexed: 10/19/2022] Open
Abstract
We show that intranigral lipopolysaccharide (LPS) injection, which provokes specific degeneration of DA neurons, induced caspase-3 activation in the rat ventral mesencephalon, which was mostly associated with glial cells. In contrast, nigral DA neurons exhibited AIF nuclear translocation in response to LPS. A significant decrease of the Bcl-2/Bax ratio in nigral tissue after LPS injection was observed. We next developed an in vitro co-culture system with the microglial BV2 and the DA neuronal MN9D murine cell lines. The silencing of caspase-3 or AIF by small interfering RNAs exclusively in the DA MN9D cells demonstrated the key role of AIF in the LPS-induced death of DA cells. In vivo chemical inhibition of caspases and poly(ADP-ribose)polymerase-1, an upstream regulator of AIF release and calpain, proved the central role of the AIF-dependent pathway in LPS-induced nigral DA cell death. We also observed nuclear translocation of AIF in the ventral mesencephalon of Parkinson's disease subjects.
Collapse
Affiliation(s)
- M A Burguillos
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|