1
|
Mayer C, Riera-Ponsati L, Kauppinen S, Klitgaard H, Erler JT, Hansen SN. Targeting the NRF2 pathway for disease modification in neurodegenerative diseases: mechanisms and therapeutic implications. Front Pharmacol 2024; 15:1437939. [PMID: 39119604 PMCID: PMC11306042 DOI: 10.3389/fphar.2024.1437939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
Neurodegenerative diseases constitute a global health issue and a major economic burden. They significantly impair both cognitive and motor functions, and their prevalence is expected to rise due to ageing societies and continuous population growth. Conventional therapies provide symptomatic relief, nevertheless, disease-modifying treatments that reduce or halt neuron death and malfunction are still largely unavailable. Amongst the common hallmarks of neurodegenerative diseases are protein aggregation, oxidative stress, neuroinflammation and mitochondrial dysfunction. Transcription factor nuclear factor-erythroid 2-related factor 2 (NRF2) constitutes a central regulator of cellular defense mechanisms, including the regulation of antioxidant, anti-inflammatory and mitochondrial pathways, making it a highly attractive therapeutic target for disease modification in neurodegenerative disorders. Here, we describe the role of NRF2 in the common hallmarks of neurodegeneration, review the current pharmacological interventions and their challenges in activating the NRF2 pathway, and present alternative therapeutic approaches for disease modification.
Collapse
Affiliation(s)
| | - Lluís Riera-Ponsati
- NEUmiRNA Therapeutics, Copenhagen, Denmark
- Center for RNA Medicine, Aalborg University, Copenhagen, Denmark
| | - Sakari Kauppinen
- NEUmiRNA Therapeutics, Copenhagen, Denmark
- Center for RNA Medicine, Aalborg University, Copenhagen, Denmark
| | | | | | | |
Collapse
|
2
|
Messana I, Manconi B, Cabras T, Boroumand M, Sanna MT, Iavarone F, Olianas A, Desiderio C, Rossetti DV, Vincenzoni F, Contini C, Guadalupi G, Fiorita A, Faa G, Castagnola M. The Post-Translational Modifications of Human Salivary Peptides and Proteins Evidenced by Top-Down Platforms. Int J Mol Sci 2023; 24:12776. [PMID: 37628956 PMCID: PMC10454625 DOI: 10.3390/ijms241612776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
In this review, we extensively describe the main post-translational modifications that give rise to the multiple proteoforms characterized to date in the human salivary proteome and their potential role. Most of the data reported were obtained by our group in over twenty-five years of research carried out on human saliva mainly by applying a top-down strategy. In the beginning, we describe the products generated by proteolytic cleavages, which can occur before and after secretion. In this section, the most relevant families of salivary proteins are also described. Next, we report the current information concerning the human salivary phospho-proteome and the limited news available on sulfo-proteomes. Three sections are dedicated to the description of glycation and enzymatic glycosylation. Citrullination and N- and C-terminal post-translational modifications (PTMs) and miscellaneous other modifications are described in the last two sections. Results highlighting the variation in the level of some proteoforms in local or systemic pathologies are also reviewed throughout the sections of the manuscript to underline the impact and relevance of this information for the development of new diagnostic biomarkers useful in clinical practice.
Collapse
Affiliation(s)
- Irene Messana
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00168 Rome, Italy; (I.M.); (C.D.); (D.V.R.)
| | - Barbara Manconi
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Tiziana Cabras
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | | | - Maria Teresa Sanna
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Federica Iavarone
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.I.); (F.V.)
- Fondazione Policlinico Universitario A. Gemelli Fondazione IRCCS, 00168 Rome, Italy;
| | - Alessandra Olianas
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Claudia Desiderio
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00168 Rome, Italy; (I.M.); (C.D.); (D.V.R.)
| | - Diana Valeria Rossetti
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00168 Rome, Italy; (I.M.); (C.D.); (D.V.R.)
| | - Federica Vincenzoni
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.I.); (F.V.)
- Fondazione Policlinico Universitario A. Gemelli Fondazione IRCCS, 00168 Rome, Italy;
| | - Cristina Contini
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Giulia Guadalupi
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Antonella Fiorita
- Fondazione Policlinico Universitario A. Gemelli Fondazione IRCCS, 00168 Rome, Italy;
- Dipartimento di Scienze dell’Invecchiamento, Neurologiche, Ortopediche e della Testa e del Collo, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Gavino Faa
- Unit of Pathology, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy;
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Massimo Castagnola
- Proteomics Laboratory, European Center for Brain Research, (IRCCS) Santa Lucia Foundation, 00168 Rome, Italy;
| |
Collapse
|
3
|
Usui N, Togawa S, Sumi T, Kobayashi Y, Koyama Y, Nakamura Y, Kondo M, Shinoda K, Kobayashi H, Shimada S. Si-Based Hydrogen-Producing Nanoagent Protects Fetuses From Miscarriage Caused by Mother-to-Child Transmission. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 3:665506. [PMID: 35047922 PMCID: PMC8757766 DOI: 10.3389/fmedt.2021.665506] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/06/2021] [Indexed: 12/24/2022] Open
Abstract
Mother-to-child transmission of viruses and bacteria increases the risk of miscarriage and various diseases in children. Such transmissions can result in infections and diseases in infants or the induction of an inflammatory immune response through the placenta. Recently, we developed a silicon (Si)-based hydrogen-producing nanoagent (Si-based agent) that continuously and effectively produces hydrogen in the body. Since medical hydrogen has antioxidative, anti-inflammatory, antiallergic, and antiapoptotic effects, we investigated the effects of our Si-based agent on mother-to-child transmission, with a focus on the rate of miscarriage. In pregnant mice fed a diet containing the Si-based agent, lipopolysaccharide (LPS)-induced miscarriage due to mother-to-child transmission was reduced and inflammation and neutrophil infiltration in the placenta were suppressed. We also found that the Si-based agent suppressed IL-6 expression in the placenta and induced the expression of antioxidant and antiapoptotic genes, such as Hmox1 and Ptgs2. The observed anti-inflammatory effects of the Si-based agent suggest that it may be an effective preventative or therapeutic drug for miscarriage or threatened miscarriage during pregnancy by suppressing maternal inflammation caused by bacterial and viral infections.
Collapse
Affiliation(s)
- Noriyoshi Usui
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan.,United Graduate School of Child Development, Osaka University, Suita, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, Suita, Japan.,Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| | - Shogo Togawa
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan.,Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Takuya Sumi
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yuki Kobayashi
- Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Japan
| | - Yoshihisa Koyama
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan.,Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| | - Yukiko Nakamura
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan.,Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| | - Makoto Kondo
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan.,Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| | - Koh Shinoda
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hikaru Kobayashi
- Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Japan
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan.,United Graduate School of Child Development, Osaka University, Suita, Japan.,Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| |
Collapse
|
4
|
Zhang Z, Hao L, Shi M, Yu Z, Shao S, Yuan Y, Zhang Z, Hölscher C. Neuroprotective Effects of a GLP-2 Analogue in the MPTP Parkinson's Disease Mouse Model. JOURNAL OF PARKINSONS DISEASE 2021; 11:529-543. [PMID: 33523018 DOI: 10.3233/jpd-202318] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Glucagon-like peptide 2 (GLP-2) is a peptide hormone derived from the proglucagon gene expressed in the intestines, pancreas and brain. Some previous studies showed that GLP-2 improved aging and Alzheimer's disease related memory impairments. Parkinson's disease (PD) is a progressive neurodegenerative disorder, and to date, there is no particular medicine reversed PD symptoms effectively. OBJECTIVE The aim of this study was to evaluate neuroprotective effects of a GLP-2 analogue in the 1-Methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) PD mouse model. METHODS In the present study, the protease resistant Gly(2)-GLP-2 (50 nmol/kg ip.) analogue has been tested for 14 days by behavioral assessment, transmission electron microscope, immunofluorescence histochemistry, enzyme-linked immunosorbent assay and western blot in an acute PD mouse model induced by MPTP. For comparison, the incretin receptor dual agonist DA5-CH was tested in a separate group. RESULTS The GLP-2 analogue treatment improved the locomotor and exploratory activity of mice, and improved bradykinesia and movement imbalance of mice. Gly(2)-GLP-2 treatment also protected dopaminergic neurons and restored tyrosine hydroxylase expression levels in the substantia nigra. Gly(2)-GLP-2 furthermore reduced the inflammation response as seen in lower microglia activation, and decreased NLRP3 and interleukin-1β pro-inflammatory cytokine expression levels. In addition, the GLP-2 analogue improved MPTP-induced mitochondrial dysfunction in the substantia nigra. The protective effects were comparable to those of the dual agonist DA5-CH. CONCLUSION The present results demonstrate that Gly(2)-GLP-2 can attenuate NLRP3 inflammasome-mediated inflammation and mitochondrial damage in the substantia nigra induced by MPTP, and Gly(2)-GLP-2 shows neuroprotective effects in this PD animal model.
Collapse
Affiliation(s)
- Zijuan Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China.,Basic Medical Collenge, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Li Hao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China.,Basic Medical Collenge, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Ming Shi
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Ziyang Yu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Simai Shao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Ye Yuan
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Zhenqiang Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Christian Hölscher
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| |
Collapse
|
5
|
Zhang L, Zhang L, Li L, Hölscher C. Semaglutide is Neuroprotective and Reduces α-Synuclein Levels in the Chronic MPTP Mouse Model of Parkinson's Disease. JOURNAL OF PARKINSONS DISEASE 2020; 9:157-171. [PMID: 30741689 DOI: 10.3233/jpd-181503] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is a progressive neurological motor control disorder. A key feature is the loss of midbrain dopaminergic neurons and the accumulation of aggregated alpha-synuclein (α-syn). No current treatment is on the market that slows or halts disease progression. Previous studies have shown that glucagon-like peptide-1 (GLP-1) receptor agonists have neuroprotective effects in animal models of PD. In addition, in a phase II clinical trial, the GLP-1 receptor agonist exendin-4 has shown good protective effects in PD patients. In the present study, we have investigated the neuroprotective effects of the GLP-1 analogues semaglutide (25 nmol/kg ip. once every two days for 30 days) and liraglutide (25 nmol/kg ip. once daily for 30 days) in the chronic MPTP mouse model of PD. Both drugs are currently on the market as a treatment for Type II diabetes. Our results show that both semaglutide and liraglutide improved MPTP-induced motor impairments. In addition, both drugs rescued the decrease of tyrosine hydroxylase (TH) levels, reduced the accumulation of α-syn, alleviated the chronic inflammation response in the brain, reduced lipid peroxidation, and inhibited the mitochondrial mitophagy signaling pathway, and furthermore increased expression of the key growth factor GDNF that protects dopaminergic neurons in the substantia nigra (SN) and striatum. Moreover, the long- acting GLP-1 analogue semaglutide was more potent compared with once daily liraglutide in most parameters measured in this study. Our results demonstrate that semaglutide may be a promising treatment for PD. A clinical trial testing semaglutide in PD patients will start shortly.
Collapse
Affiliation(s)
- Liping Zhang
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - LingYu Zhang
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Lin Li
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Christian Hölscher
- Department of Second Hospital Neurology, Shanxi medical University, Taiyuan, Shanxi, PR China.,Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| |
Collapse
|
6
|
Renoprotective and neuroprotective effects of enteric hydrogen generation from Si-based agent. Sci Rep 2020; 10:5859. [PMID: 32246095 PMCID: PMC7125117 DOI: 10.1038/s41598-020-62755-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 03/18/2020] [Indexed: 01/08/2023] Open
Abstract
We have developed Si-based agent which can generate a large amount of hydrogen. Si-based agent continues generating hydrogen for more than 24 h by the reaction with water under conditions similar to those in bowels, i.e., pH8.3 and 36 °C, and generates ~400 mL hydrogen. To investigate beneficial effects for diseases associated with oxidative stress, Si-based agent is administered to remnant kidney rats and Parkinson's disease mice. Rats are fed with control or Si-based agent-containing diet for 8 weeks. Si-based agent is found to greatly suppress the development of renal failure and the parameters of oxidative stress. Treatment with Si-based agent in a mouse model of hemi-Parkinson's disease induced by 6-hydroxydopamine attenuated degeneration of dopaminergic neurons and prevented impairment of motor balance and coordination. These findings indicate that the Si-based agent shows renoprotective and neuroprotective effects presumably via suppression of oxidative stress by generation of hydrogen.
Collapse
|
7
|
Hölscher C. Brain insulin resistance: role in neurodegenerative disease and potential for targeting. Expert Opin Investig Drugs 2020; 29:333-348. [PMID: 32175781 DOI: 10.1080/13543784.2020.1738383] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: This review evaluates the novel strategy of treating Alzheimer's and Parkinson's disease (AD and PD) withdrugs that initially have been developed to treat type 2 diabetes. As insulin signalling has been found to be de-sensitized in the brains of patients, drugs that can re-sensitize insulin signalling have been tested to evaluate if this strategy can alter disease progression.Areas covered: The review will give an overview of preclinical and clinical tests in AD and PD of drugs activating insulin receptors, glucagon-like peptide -1 (GLP-1) receptors, and glucose-dependent insulinotropic polypeptide (GIP) receptors.Expert opinion: Insulin, GLP-1 and GIP receptor agonists have shown good effects in preclinical studies. First clinical trials in MCI/AD patients have shown that insulin can improve on key pathological symptoms of AD such as memory impairment, brain activity, neuronal energy utilization, and inflammation markers. A GLP-1 receptor agonist has shown disease-modifying effects in PD patients, and first pilot studies have shown encouraging effects of a GLP-1 receptor agonist in AD patients. Novel dual GLP-1/GIP receptor agonists that cross the blood brain barrier show superior neuroprotective effects compared to single GLP-1 or GIP receptor agonists, and show great promise as novel treatments of AD and PD.
Collapse
Affiliation(s)
- Christian Hölscher
- Second Hospital, Neurology Department, Shanxi Medical University, Taiyuan, Shanxi, PR China.,Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, Henan, PR China
| |
Collapse
|
8
|
Zhang ZQ, Hölscher C. GIP has neuroprotective effects in Alzheimer and Parkinson's disease models. Peptides 2020; 125:170184. [PMID: 31705913 DOI: 10.1016/j.peptides.2019.170184] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/17/2022]
Abstract
Glucose-dependent Insulinotropic polypeptide (GIP) is a peptide hormone of the incretin family. It has growth factor properties and can re-activate energy utilization. In progressive neurodegenerative disorders such as Alzheimer's and Parkinson's disease, energy utilization is much reduced, and GIP has the potential to reverse this. Furthermore, GIP can reduce the inflammation response in the brain and reduce levels of pro-inflammatory cytokines. Tests in animal models of Alzheimer's and Parkinson's disease show good neuroprotective effects. In Parkinson's disease models, motor activity is normalized, dopaminergic neurons are protected, synapse numbers and dopamine levels are maintained. Levels of growth factors that are essential for neuronal and synaptic function are increased and alpha-synuclein levels are reduced. The chronic inflammation response and mitochondrial damage is reduced. In Alzheimer's disease models, memory is rescued, synapse numbers and synaptic plasticity in the hippocampus is normalized, amyloid plaque load and the chronic inflammation is reduced. Similar protective effects have been previously reported with analogues of glucagon-like peptide 1 (GLP-1), the sister incretin hormone. First clinical trials show good protective effects in both diseases. Recently, novel dual GLP-1/GIP receptor agonists have been developed. The ability to cross the blood-brain barrier (BBB) is key to their neuroprotective effects. We have developed two dual GLP-1/GIP receptor agonist that have cell penetrating sequences added for better BBB penetration. In direct comparisons, these dual agonists show improved neuroprotection in a mouse model of Parkinson's disease. Therefore, such novel multiple receptor agonists hold great promise as potential treatments for Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Zhen Qiang Zhang
- Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Christian Hölscher
- Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, Henan Province, China.
| |
Collapse
|
9
|
Obata T. Glutaminergic tonic action potentiate MPP+-induced hydroxyl radical production in rat striatum. Neurosci Lett 2019; 705:51-53. [DOI: 10.1016/j.neulet.2019.02.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/03/2019] [Accepted: 02/09/2019] [Indexed: 11/15/2022]
|
10
|
Van Laar VS, Otero PA, Hastings TG, Berman SB. Potential Role of Mic60/Mitofilin in Parkinson's Disease. Front Neurosci 2019; 12:898. [PMID: 30740041 PMCID: PMC6357844 DOI: 10.3389/fnins.2018.00898] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/16/2018] [Indexed: 12/21/2022] Open
Abstract
There are currently no treatments that hinder or halt the inexorable progression of Parkinson's disease (PD). While the etiology of PD remains elusive, evidence suggests that early dysfunction of mitochondrial respiration and homeostasis play a major role in PD pathogenesis. The mitochondrial structural protein Mic60, also known as mitofilin, is critical for maintaining mitochondrial architecture and function. Loss of Mic60 is associated with detrimental effects on mitochondrial homeostasis. Growing evidence now implicates Mic60 in the pathogenesis of PD. In this review, we discuss the data supporting a role of Mic60 and mitochondrial dysfunction in PD. We will also consider the potential of Mic60 as a therapeutic target for treating neurological disorders.
Collapse
Affiliation(s)
- Victor S Van Laar
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States
| | - P Anthony Otero
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States.,Division of Neuropathology, Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Cellular and Molecular Pathology (CMP) Program, Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Teresa G Hastings
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sarah B Berman
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States.,Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
11
|
Zhang L, Zhang L, Li L, Hölscher C. Neuroprotective effects of the novel GLP-1 long acting analogue semaglutide in the MPTP Parkinson's disease mouse model. Neuropeptides 2018; 71:70-80. [PMID: 30017231 DOI: 10.1016/j.npep.2018.07.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 12/25/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, and there is no recognised therapy to cure it. Recently, it has been shown that treatments to improve insulin resistance in type 2 diabetes (T2DM) may be useful for PD patients. In previous studies, the glucagon-like peptide-1 (GLP-1) receptor agonist liraglutide showed good neuroprotective effects in animal models of PD. In addition, the GLP-1 mimetic exendin-4 has shown good protective effects in PD patients in a phase II clinical trial. Here, we report the protective effects of semaglutide (25 nmol/kg ip. once-daily for 7 days), a new long-acting GLP-1 analogue, in the MPTP mouse model of PD. Moreover, we compared the neuroprotective effect of semaglutide with liraglutide given at the same dose. Our work shows that both semaglutide and liraglutide improved 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced motor impairments. In addition, both GLP-1 analogues rescued the decrease of tyrosine hydroxylase (TH) levels, alleviated the inflammation response, reduced lipid peroxidation, inhibited the apoptosis pathway, and also increased autophagy- related protein expression, to protect dopaminergic neurons in the substantia nigra and striatum. Moreover, the long-acting GLP-1 analogue semaglutide was superior to liraglutide in most parameters measured in this study. Our results demonstrate that the new long- acting GLP-1 analogue semaglutide may be a promising treatment for PD.
Collapse
Affiliation(s)
- Liping Zhang
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Lingyu Zhang
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Lin Li
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, PR China.
| | - Christian Hölscher
- Second hospital Neurology Dept., Shanxi medical University, Taiyuan, Shanxi, PR China; Biomedical and Life Science, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| |
Collapse
|
12
|
Mészáros AT, Szilágyi ÁL, Juhász L, Tuboly E, Érces D, Varga G, Hartmann P. Mitochondria As Sources and Targets of Methane. Front Med (Lausanne) 2017; 4:195. [PMID: 29181377 PMCID: PMC5693848 DOI: 10.3389/fmed.2017.00195] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/25/2017] [Indexed: 12/14/2022] Open
Abstract
This review summarizes the current knowledge on the role of mitochondria in the context of hypoxic cell biology, while providing evidence of how these mechanisms are modulated by methane (CH4). Recent studies have unambiguously confirmed CH4 bioactivity in various in vitro and in vivo experimental models and established the possibility that CH4 can affect many aspects of mitochondrial physiology. To date, no specific binding of CH4 to any enzymes or receptors have been reported, and it is probable that many of its effects are related to physico-chemical properties of the non-polar molecule. (i) Mitochondria themselves can be sources of endogenous CH4 generation under oxido-reductive stress conditions; chemical inhibition of the mitochondrial electron transport chain with site-specific inhibitors leads to increased formation of CH4 in eukaryote cells, in plants, and in animals. (ii) Conventionally believed as physiologically inert, studies cited in this review demonstrate that exogenous CH4 modulates key events of inflammation. The anti-apoptotic effects of exogenously administered CH4 are also recognized, and these properties also suggest that CH4-mediated intracellular signaling is closely associated with mitochondria. (iii) Mitochondrial substrate oxidation is coupled with the reduction of molecular oxygen, thus providing energy for cellular metabolism. Interestingly, recent in vivo studies have shown improved basal respiration and modulated mitochondrial oxidative phosphorylation by exogenous CH4. Overall, these data suggest that CH4 liberation and effectiveness in eukaryotes are both linked to hypoxic events and redox regulation and support the notion that CH4 has therapeutic roles in mammalian pathophysiologies.
Collapse
Affiliation(s)
| | | | - László Juhász
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| | - Eszter Tuboly
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| | - Dániel Érces
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| | - Gabriella Varga
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| | - Petra Hartmann
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| |
Collapse
|
13
|
Yuan Z, Li D, Feng P, Xue G, Ji C, Li G, Hölscher C. A novel GLP-1/GIP dual agonist is more effective than liraglutide in reducing inflammation and enhancing GDNF release in the MPTP mouse model of Parkinson's disease. Eur J Pharmacol 2017; 812:82-90. [DOI: 10.1016/j.ejphar.2017.06.029] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/23/2017] [Accepted: 06/26/2017] [Indexed: 12/13/2022]
|
14
|
Wimalasena K. Current Status, Gaps, and Weaknesses of the Mechanism of Selective Dopaminergic Toxicity of MPTP/MPP +. ADVANCES IN MOLECULAR TOXICOLOGY 2017. [DOI: 10.1016/b978-0-12-812522-9.00003-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
15
|
Lin Z, Guichun Z, Lifeng L, Chen C, Xuecheng C, Jinfang C. Protective effect of α-lipoic acid against antimycin A cytotoxicity in MC3T3-E1 osteoblastic cells. Cell Stress Chaperones 2017; 22:5-13. [PMID: 27796798 PMCID: PMC5225054 DOI: 10.1007/s12192-016-0735-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 08/20/2016] [Accepted: 09/10/2016] [Indexed: 11/29/2022] Open
Abstract
Oxidative stress represents a major cause of cellular damage and death in the process of osteoporosis. Antimycin A (AMA) has been shown to stimulate mitochondrial superoxide anions and reactive oxygen species (ROS). α-Lipoic acid (α-LA) is a naturally occurring essential coenzyme in mitochondrial multienzyme complexes and acts as a key player in mitochondrial energy production. However, whether α-LA affects the cytotoxicity of AMA in osteoblastic cells is unknown. In this study, we investigated the protective effects of α-LA against AMA-induced cytotoxicity using the MC3T3-E1 osteoblast-like cell line. Our results indicated that α-LA treatment attenuated AMA-induced cytotoxicity and LDH release in a dose-dependent manner. Notably, a significant recovery effect of α-LA on mineralization inhibited by AMA was found. Our results also demonstrated that treatment with 50 μM AMA leads to a reduction of mitochondrial membrane potential (MMP) and the complex IV dysfunction, which was inhibited by pretreatment with α-LA in a dose-dependent manner. In addition, treatment with α-LA significantly reduced the generation of ROS and mitochondrial superoxide production induced by AMA. In addition, our result suggests that PI3K/Akt and CREB pathways are related to the protective effect of α-LA. Importantly, Hoechst 33258 staining results indicated that pretreatment with α-LA prevented AMA-induced apoptosis. Mechanistically, we found that α-LA prevents MC3T3-E1 cells from apoptosis through attenuating cytochrome C release and reducing the level of cleaved caspase-3.
Collapse
Affiliation(s)
- Zou Lin
- Department of Traumatic Orthopedic Surgery, The General Hospital of Ji'nan Military Command, 25 Shifan St, Jinan, Shandong Province, 250031, China
| | - Zhang Guichun
- Department of Traumatic Orthopedic Surgery, The General Hospital of Ji'nan Military Command, 25 Shifan St, Jinan, Shandong Province, 250031, China
| | - Liu Lifeng
- Department of Traumatic Orthopedic Surgery, The General Hospital of Ji'nan Military Command, 25 Shifan St, Jinan, Shandong Province, 250031, China
| | - Chen Chen
- Department of Traumatic Orthopedic Surgery, The General Hospital of Ji'nan Military Command, 25 Shifan St, Jinan, Shandong Province, 250031, China
| | - Cao Xuecheng
- Department of Traumatic Orthopedic Surgery, The General Hospital of Ji'nan Military Command, 25 Shifan St, Jinan, Shandong Province, 250031, China.
| | - Cai Jinfang
- Department of Traumatic Orthopedic Surgery, The General Hospital of Ji'nan Military Command, 25 Shifan St, Jinan, Shandong Province, 250031, China
| |
Collapse
|
16
|
Zailaie MZ. Epidermal hydrogen peroxide is not increased in lesional and non-lesional skin of vitiligo. Arch Dermatol Res 2016; 309:31-42. [PMID: 27783153 DOI: 10.1007/s00403-016-1695-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/26/2016] [Accepted: 10/02/2016] [Indexed: 11/28/2022]
Abstract
It is widely believed that the loss of the epidermal melanocytes in vitiligo is basically due to excessive oxidative stress. Previous research work described abnormal elevation of the absolute concentration of the epidermal hydrogen peroxide (H2O2) in lesional and non-lesional skin of vitiligo. Based on this finding, our primary research objective was to use this feature as a screening marker in individuals at a great risk of developing vitiligo. Ninety-six patients of non-segmental vitiligo (NSV) of varying durations, skin phototypes, and treatment modalities (psoralen UVA-, narrow band UVB-treated) were recruited for this study. Raman spectroscopic measurements, using an external probehead, of the lesional and non-lesional skin were obtained, and the resulting spectra were analyzed using the Opus software package of the MultiRam spectrometer and the intensity of the peak at 875 cm-1 that represents the absolute concentration of H2O2 was calculated. Contrary to previous reports, in patients of skin phototype IV, the absolute concentrations of H2O2 in non-lesional and lesional NSV of all groups were non-significantly decreased compared to normal control. In patients of NSV of skin phototype V, the decrease in the absolute concentrations of H2O2 was not significant in the untreated group, and a slight non-significant increase in the NBUVB-treated group was noted. However, in the PUVA-treated group, the non-lesional skin demonstrated significant increase in the absolute concentration of H2O2, whereas the lesional skin showed only a slight non-significant increase compared to normal control. In NSV patients of skin phototype VI who were previously treated with PUVA, the non-lesional skin showed a slight non-significant increase in the absolute concentration of H2O2; however, the lesional skin showed a marked significant decrease compared to normal control and the non-lesional skin. Thereof, one can conclude that the epidermal H2O2 is not increased in NSV as previously thought and may not be responsible for the oxidative stress that leads to the melanocytes destruction, the hallmark of vitiligo pathogenesis.
Collapse
Affiliation(s)
- Mohammad Z Zailaie
- The Clinical and Experimental Dermatology Unit, King Fahad Medical Research Center, King Abdulaziz University Medical Center, Jeddah, 21589, Saudi Arabia.
| |
Collapse
|
17
|
Chaves RS, Kazi AI, Silva CM, Almeida MF, Lima RS, Carrettiero DC, Demasi M, Ferrari MFR. Presence of insoluble Tau following rotenone exposure ameliorates basic pathways associated with neurodegeneration. IBRO Rep 2016; 1:32-45. [PMID: 30135926 PMCID: PMC6084878 DOI: 10.1016/j.ibror.2016.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/23/2016] [Accepted: 09/24/2016] [Indexed: 01/24/2023] Open
Abstract
Protein aggregation is an important feature of neurodegenerative disorders. In Alzheimer's disease (AD) protein aggregates are composed of hyperphosphorylated Tau and amyloid beta peptide (Aβ). Despite the involvement and identification of the molecular composition of these aggregates, their role in AD pathophysiology is not fully understood. However, depositions of these insoluble aggregates are typically reported as pathogenic and toxic for cell homeostasis. New evidences suggest that the deposition of these aggregates is a protective mechanism that preserves cell from toxic insults associated with the early stages of neurodegenerative diseases. To better understand the biological role of the protein aggregation with regard its effects in cellular homeostasis, the present study investigated the role of insoluble Tau and Tau aggregates on crucial cellular parameters such as redox homeostasis, proteasome activity and autophagy in hippocampal cell cultures and hippocampus of aged Lewis rats using a rotenone-induced aggregation model. Neurons were exposed to rotenone in different concentrations and exposure times aiming to determine the interval required for Tau aggregation. Our experimental design allowed us to demonstrate that rotenone exposure induces Tau hyperphosphorylation and aggregation in a concentration and time-dependent manner. Oxidative stress triggered by rotenone exposure was observed with the absence of Tau aggregates and was reduced or absent when Tau aggregates were present. This reduction of oxidative stress along with the presence of insoluble Tau was independent of alterations in antioxidant enzymes activities or cell death. In addition, rotenone induced oxidative stress was mainly associated with decrease in proteasome activity and autophagy flux. Conversely, when insoluble Tau appeared, autophagy turns to be overactivated while proteasome activity remained low. Our studies significantly advance the understanding that Tau aggregation might exert protective cellular effects, at least briefly, when neurons are facing neurodegeneration stimulus. We believe that our data add more complexity for the understanding of protein aggregation role in AD etiology.
Collapse
Affiliation(s)
- Rodrigo S Chaves
- Department of Genetics and Evolutionary Biology - Institute for Biosciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Amajad I Kazi
- Department of Genetics and Evolutionary Biology - Institute for Biosciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Carolliny M Silva
- Department of Genetics and Evolutionary Biology - Institute for Biosciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Michael F Almeida
- Department of Genetics and Evolutionary Biology - Institute for Biosciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Raquel S Lima
- Department of Genetics and Evolutionary Biology - Institute for Biosciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | | | - Marilene Demasi
- Laboratory of Biochemistry and Biophysics - Butantan Institute, Sao Paulo, SP, Brazil
| | - Merari F R Ferrari
- Department of Genetics and Evolutionary Biology - Institute for Biosciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
18
|
Li Y, Liu W, Li L, Hölscher C. Neuroprotective effects of a GIP analogue in the MPTP Parkinson's disease mouse model. Neuropharmacology 2016; 101:255-63. [DOI: 10.1016/j.neuropharm.2015.10.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 09/06/2015] [Accepted: 10/02/2015] [Indexed: 12/25/2022]
|
19
|
Liu W, Li Y, Jalewa J, Saunders-Wood T, Li L, Hölscher C. Neuroprotective effects of an oxyntomodulin analogue in the MPTP mouse model of Parkinson's disease. Eur J Pharmacol 2015; 765:284-90. [DOI: 10.1016/j.ejphar.2015.08.038] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/18/2015] [Accepted: 08/19/2015] [Indexed: 11/26/2022]
|
20
|
Yoon SY, Oh YJ. Glucose Levels in Culture Medium Determine Cell Death Mode in MPP(+)-treated Dopaminergic Neuronal Cells. Exp Neurobiol 2015; 24:197-205. [PMID: 26412968 PMCID: PMC4580746 DOI: 10.5607/en.2015.24.3.197] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 08/16/2015] [Accepted: 08/17/2015] [Indexed: 11/19/2022] Open
Abstract
We previously demonstrated that 1-methyl-4-phenylpyridinium (MPP(+)) causes caspase-independent, non-apoptotic death of dopaminergic (DA) neuronal cells. Here, we specifically examined whether change of glucose concentration in culture medium may play a role for determining cell death modes of DA neurons following MPP(+) treatment. By incubating MN9D cells in medium containing varying concentrations of glucose (5~35 mM), we found that cells underwent a distinct cell death as determined by morphological and biochemical criteria. At 5~10 mM glucose concentration (low glucose levels), MPP(+) induced typical of the apoptotic dell death accompanied with caspase activation and DNA fragmentation as well as cell shrinkage. In contrast, MN9D cells cultivated in medium containing more than 17.5 mM (high glucose levels) did not demonstrate any of these changes. Subsequently, we observed that MPP(+) at low glucose levels but not high glucose levels led to ROS generation and subsequent JNK activation. Therefore, MPP(+)-induced cell death only at low glucose levels was significantly ameliorated following co-treatment with ROS scavenger, caspase inhibitor or JNK inhibitor. We basically confirmed the quite similar pattern of cell death in primary cultures of DA neurons. Taken together, our results suggest that a biochemically distinct cell death mode is recruited by MPP(+) depending on extracellular glucose levels.
Collapse
Affiliation(s)
- So-Young Yoon
- Department of Systems Biology, Yonsei University College of Life Science and Biotechnology, Seoul 03722, Korea
| | - Young J Oh
- Department of Systems Biology, Yonsei University College of Life Science and Biotechnology, Seoul 03722, Korea
| |
Collapse
|
21
|
Liu W, Jalewa J, Sharma M, Li G, Li L, Hölscher C. Neuroprotective effects of lixisenatide and liraglutide in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. Neuroscience 2015; 303:42-50. [DOI: 10.1016/j.neuroscience.2015.06.054] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 06/16/2015] [Accepted: 06/24/2015] [Indexed: 12/25/2022]
|
22
|
The novel mitochondrial iron chelator 5-((methylamino)methyl)-8-hydroxyquinoline protects against mitochondrial-induced oxidative damage and neuronal death. Biochem Biophys Res Commun 2015; 463:787-92. [DOI: 10.1016/j.bbrc.2015.06.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/02/2015] [Indexed: 11/21/2022]
|
23
|
Delgado-Cortés MJ, Espinosa-Oliva AM, Sarmiento M, Argüelles S, Herrera AJ, Mauriño R, Villarán RF, Venero JL, Machado A, de Pablos RM. Synergistic Deleterious Effect of Chronic Stress and Sodium Azide in the Mouse Hippocampus. Chem Res Toxicol 2015; 28:651-61. [DOI: 10.1021/tx5004408] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- María José Delgado-Cortés
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Sevilla, 41012-Sevilla, Spain
- Instituto de Biomedicina
de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad
de Sevilla, 41013-Sevilla, Spain
| | - Ana M. Espinosa-Oliva
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Sevilla, 41012-Sevilla, Spain
- Instituto de Biomedicina
de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad
de Sevilla, 41013-Sevilla, Spain
| | - Manuel Sarmiento
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Sevilla, 41012-Sevilla, Spain
- Instituto de Biomedicina
de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad
de Sevilla, 41013-Sevilla, Spain
| | - Sandro Argüelles
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Sevilla, 41012-Sevilla, Spain
- Instituto de Biomedicina
de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad
de Sevilla, 41013-Sevilla, Spain
| | - Antonio J. Herrera
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Sevilla, 41012-Sevilla, Spain
- Instituto de Biomedicina
de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad
de Sevilla, 41013-Sevilla, Spain
| | - Raquel Mauriño
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Sevilla, 41012-Sevilla, Spain
- Instituto de Biomedicina
de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad
de Sevilla, 41013-Sevilla, Spain
| | - Ruth F. Villarán
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Sevilla, 41012-Sevilla, Spain
- Instituto de Biomedicina
de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad
de Sevilla, 41013-Sevilla, Spain
| | - José L. Venero
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Sevilla, 41012-Sevilla, Spain
- Instituto de Biomedicina
de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad
de Sevilla, 41013-Sevilla, Spain
| | - Alberto Machado
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Sevilla, 41012-Sevilla, Spain
- Instituto de Biomedicina
de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad
de Sevilla, 41013-Sevilla, Spain
| | - Rocío M. de Pablos
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Sevilla, 41012-Sevilla, Spain
- Instituto de Biomedicina
de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad
de Sevilla, 41013-Sevilla, Spain
| |
Collapse
|
24
|
Choi EM, Lee YS. Paeoniflorin isolated from Paeonia lactiflora attenuates osteoblast cytotoxicity induced by antimycin A. Food Funct 2014; 4:1332-8. [PMID: 23824342 DOI: 10.1039/c3fo60147a] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The protective effects of paeoniflorin isolated from Paeonia lactiflora against pharmacological inhibition of the respiratory chain were studied using osteoblastic MC3T3-E1 cells. Here we show that paeoniflorin decreases cell death induced by antimycin A, an inhibitor of mitochondrial complex III. Paeoniflorin restored antimycin A-induced inactivation of phosphoinositide 3-kinase (PI3K) and thioredoxin reductase, suggesting that PI3K and thioredoxin reductase may be involved in paeoniflorin-induced cytoprotective responses. We also examined the effect of paeoniflorin on mitochondrial dysfunction and oxidative stress induced by antimycin A. Paeoniflorin inhibited mitochondrial membrane potential dissipation, ATP loss, inactivation of complexes I and IV, cytochrome c release, and cardiolipin oxidation induced by antimycin A. In addition, paeoniflorin prevented antimycin A-induced ROS release and nitrotyrosine increase. These results imply that paeoniflorin protects osteoblasts from antimycin A-induced cell death via improved mitochondrial function.
Collapse
Affiliation(s)
- Eun Mi Choi
- Department of Food & Nutrition, Kyung Hee University, 1, Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Republic of Korea.
| | | |
Collapse
|
25
|
Zhelev Z, Bakalova R, Aoki I, Lazarova D, Saga T. Imaging of superoxide generation in the dopaminergic area of the brain in Parkinson's disease, using mito-TEMPO. ACS Chem Neurosci 2013; 4:1439-45. [PMID: 24024751 PMCID: PMC3837371 DOI: 10.1021/cn400159h] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
We report a new methodology for direct visualization of superoxide production in the dopaminergic area of the brain in Parkinson's disease, based on the redox cycle of mito-TEMPO, a blood-brain barrier-, cell-, and mitochondria-penetrating nitroxide derivative with superoxide scavenging properties and T1 magnetic resonance imaging (MRI) contrast. The experiments were conducted on healthy and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice. In healthy mice, the nitroxide-enhanced MRI signal was weak and short-lived (half-life ∼ 40 s; duration ∼ 80 s). The profile of the histograms indicated a high reducing activity of normal brain tissues against mito-TEMPO. In MPTP-treated mice, the nitroxide-enhanced MRI signal was strong and long-lived (half-life > 20 min; duration > 20 min), especially in the dopaminergic area of the brain. The histograms indicated a high oxidative activity in dopaminergic tissues of MPTP-treated mice. The results show directly, on intact mammals, that superoxide is a major inducer and/or mediator of neurodegenerative damage in Parkinson's disease. The high oxidative status of brain tissue in Parkinson's disease was also confirmed on isolated tissue specimens, using total reducing capacity assay and ROS/RNS assay.
Collapse
Affiliation(s)
- Zhivko Zhelev
- Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- Medical Faculty, Trakia University, 11 Armeiska Str., Stara Zagora 6000, Bulgaria
- Institute of Biophysics
and Biomedical Engineering, Bulgarian Academy
of Sciences, 23 Avad.
G. Bonchev Str., Sofia 1000, Bulgaria
| | - Rumiana Bakalova
- Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- Medical Faculty, Sofia University, 1 Koziak Str., Sofia 1407, Bulgaria
| | - Ichio Aoki
- Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | | | - Tsuneo Saga
- Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| |
Collapse
|
26
|
Tuboly E, Szabó A, Erős G, Mohácsi Á, Szabó G, Tengölics R, Rákhely G, Boros M. Determination of endogenous methane formation by photoacoustic spectroscopy. J Breath Res 2013; 7:046004. [PMID: 24185326 DOI: 10.1088/1752-7155/7/4/046004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
27
|
Suh KS, Choi EM, Lee YS, Kim YS. Protective effect of albiflorin against oxidative-stress-mediated toxicity in osteoblast-like MC3T3-E1 cells. Fitoterapia 2013; 89:33-41. [DOI: 10.1016/j.fitote.2013.05.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/16/2013] [Accepted: 05/19/2013] [Indexed: 01/10/2023]
|
28
|
Madathil KS, Karuppagounder SS, Haobam R, Varghese M, Rajamma U, Mohanakumar KP. Nitric oxide synthase inhibitors protect against rotenone-induced, oxidative stress mediated parkinsonism in rats. Neurochem Int 2013; 62:674-83. [PMID: 23353925 DOI: 10.1016/j.neuint.2013.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Revised: 12/24/2012] [Accepted: 01/08/2013] [Indexed: 12/14/2022]
Abstract
Rotenone is known to cause progressive dopaminergic neuronal loss in rodents, but it remains unclear how this mitochondrial complex-I inhibitor mediates neurodegeneration specific to substantia nigra pars compacta (SNpc). One of the proposed mechanisms is increased free radical generation owing to mitochondrial electron transport chain dysfunction following complex-I inhibition. The present study examined the role of nitric oxide (NO) and hydroxyl radicals (OH) in mediating rotenone-induced dopaminergic neurotoxicity. Indications of NO involvement are evidenced by inducible nitric oxide synthase (NOS) over-expression, and increased NADPH-diaphorase staining in SNpc neurons 96h following rotenone administration. Treatment of these animals with specific neuronal NOS inhibitor, 7-nitroindazole (7-NI) and non-specific NOS inhibitor, N-ω-nitro-l-argenine methyl ester (l-NAME) caused reversal of rotenone-induced striatal dopamine depletion, and attenuation of the neurotoxin-induced decrease in the number of tyrosine hydroxylase immunoreactive neurons in SNpc, as well as in apomorphine and amphetamine-induced unilateral rotations. Interestingly, the study also demonstrated the contribution of OH in mediating rotenone nigral toxicity since there appeared a significant generation of the reactive oxygen species in vivo 24h following rotenone administration, a copious loss of reduced and oxidized glutathione, and increased superoxide dismutase and catalase activities in the cytosolic fractions of the ipsilateral SNpc area on the 5th day. An OH scavenging capacity of 7-NI and l-NAME in a Fenton-like reaction, as well as complete reversal of the rotenone-induced increases in the antioxidant enzyme activities, and the loss in reduced and oxidized glutathione contents in the SNpc supported OH involvement in rotenone-induced dopaminergic neurotoxicity. While these results strongly suggest the contribution of both OH and NO, resulting in acute oxidative stress culminating in dopaminergic neurodegeneration caused by rotenone, the course of events indicated generation of OH as the primary event in the neurotoxic processes.
Collapse
Affiliation(s)
- K S Madathil
- Division of Cell Biology & Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, India
| | | | | | | | | | | |
Collapse
|
29
|
Mechanism of oxidative stress in neurodegeneration. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:428010. [PMID: 22685618 PMCID: PMC3362933 DOI: 10.1155/2012/428010] [Citation(s) in RCA: 595] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 03/14/2012] [Indexed: 12/11/2022]
Abstract
Biological tissues require oxygen to meet their energetic demands. However, the consumption of oxygen also results in the generation of free radicals that may have damaging effects on cells. The brain is particularly vulnerable to the effects of reactive oxygen species due to its high demand for oxygen, and its abundance of highly peroxidisable substrates. Oxidative stress is caused by an imbalance in the redox state of the cell, either by overproduction of reactive oxygen species, or by dysfunction of the antioxidant systems. Oxidative stress has been detected in a range of neurodegenerative disease, and emerging evidence from in vitro and in vivo disease models suggests that oxidative stress may play a role in disease pathogenesis. However, the promise of antioxidants as novel therapies for neurodegenerative diseases has not been borne out in clinical studies. In this review, we critically assess the hypothesis that oxidative stress is a crucial player in common neurodegenerative disease and discuss the source of free radicals in such diseases. Furthermore, we examine the issues surrounding the failure to translate this hypothesis into an effective clinical treatment.
Collapse
|
30
|
Choi EM. Luteolin protects osteoblastic MC3T3-E1 cells from antimycin A-induced cytotoxicity through the improved mitochondrial function and activation of PI3K/Akt/CREB. Toxicol In Vitro 2011; 25:1671-9. [DOI: 10.1016/j.tiv.2011.07.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 06/29/2011] [Accepted: 07/07/2011] [Indexed: 10/18/2022]
|
31
|
Choi EM. Protective effect of diazoxide against antimycin A-induced mitochondrial dysfunction in osteoblastic MC3T3-E1 cells. Toxicol In Vitro 2011; 25:1603-8. [DOI: 10.1016/j.tiv.2011.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 05/30/2011] [Accepted: 06/07/2011] [Indexed: 10/18/2022]
|
32
|
Choi EM. Glabridin protects osteoblastic MC3T3-E1 cells against antimycin A induced cytotoxicity. Chem Biol Interact 2011; 193:71-8. [DOI: 10.1016/j.cbi.2011.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 05/09/2011] [Accepted: 05/13/2011] [Indexed: 12/21/2022]
|
33
|
Choi EM. Honokiol protects osteoblastic MC3T3-E1 cells against antimycin A-induced cytotoxicity. Inflamm Res 2011; 60:1005-12. [DOI: 10.1007/s00011-011-0360-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2011] [Accepted: 06/27/2011] [Indexed: 01/17/2023] Open
|
34
|
Choi EM. Kaempferol protects MC3T3-E1 cells through antioxidant effect and regulation of mitochondrial function. Food Chem Toxicol 2011; 49:1800-5. [PMID: 21565246 DOI: 10.1016/j.fct.2011.04.031] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 04/20/2011] [Accepted: 04/23/2011] [Indexed: 12/22/2022]
Abstract
Kaempferol, a natural flavonoid present in fruits, vegetables, and teas, provides beneficial effects for human health. In this study, we investigated the protective effects of kaempferol on antimycin A (AMA)-induced toxicity in osteoblast-like MC3T3-E1 cells. Exposure of MC3T3-E1 cells to AMA caused significant cell viability loss, as well as mitochondrial membrane potential dissipation, complex IV inactivation, intracellular calcium ([Ca²⁺](i)) elevation, and reactive oxygen species (ROS) production. Pretreatment with kaempferol prior to AMA exposure significantly reduced AMA-induced cell damage by preventing mitochondrial membrane potential dissipation, complex IV inactivation, [Ca²⁺](i) elevation, and ROS production. Kaempferol also induced the activation of PI3K (phosphoinositide 3-kinase), Akt (protein kinase B), and CREB (cAMP-response element-binding protein) inhibited by AMA, which result demonstrates that kaempferol utilizes the PI3K/Akt/CREB pathway to augment metabolic activity inhibited by AMA. All these data indicate that kaempferol may reduce or prevent osteoblasts degeneration in osteoporosis or other degenerative disorders.
Collapse
Affiliation(s)
- Eun Mi Choi
- Department of Food and Nutrition, Education Graduate School, Kyung Hee University, Seoul 130-701, South Korea.
| |
Collapse
|
35
|
Existence of a threshold for hydroxyl radical generation independent of hypoxia in rat striatum during carbon monoxide poisoning. Arch Toxicol 2011; 85:1091-9. [DOI: 10.1007/s00204-010-0637-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Accepted: 12/16/2010] [Indexed: 10/18/2022]
|
36
|
Genetic, functional and evolutionary characterization of scox, the Drosophila melanogaster ortholog of the human SCO1 gene. Mitochondrion 2010; 10:433-48. [DOI: 10.1016/j.mito.2010.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 02/26/2010] [Accepted: 04/02/2010] [Indexed: 12/15/2022]
|
37
|
Smaili S, Hirata H, Ureshino R, Monteforte PT, Morales AP, Muler ML, Terashima J, Oseki K, Rosenstock TR, Lopes GS, Bincoletto C. Calcium and cell death signaling in neurodegeneration and aging. AN ACAD BRAS CIENC 2010; 81:467-75. [PMID: 19722016 DOI: 10.1590/s0001-37652009000300011] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Accepted: 02/16/2009] [Indexed: 01/12/2023] Open
Abstract
Transient increase in cytosolic (Cac2+) and mitochondrial Ca2+ (Ca m2+) are essential elements in the control of many physiological processes. However, sustained increases in Ca c2+ and Ca m2+ may contribute to oxidative stress and cell death. Several events are related to the increase in Ca m2+, including regulation and activation of a number of Ca2+ dependent enzymes, such as phospholipases, proteases and nucleases. Mitochondria and endoplasmic reticulum (ER) play pivotal roles in the maintenance of intracellular Ca2+ homeostasis and regulation of cell death. Several lines of evidence have shown that, in the presence of some apoptotic stimuli, the activation of mitochondrial processes may lead to the release of cytochrome c followed by the activation of caspases, nuclear fragmentation and apoptotic cell death. The aim of this review was to show how changes in calcium signaling can be related to the apoptotic cell death induction. Calcium homeostasis was also shown to be an important mechanism involved in neurodegenerative and aging processes.
Collapse
Affiliation(s)
- Soraya Smaili
- Departamento de Farmacologia, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brasil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Zhang X, Monroe ME, Chen B, Chin MH, Heibeck TH, Schepmoes AA, Yang F, Petritis BO, Camp DG, Pounds JG, Jacobs JM, Smith DJ, Bigelow DJ, Smith RD, Qian WJ. Endogenous 3,4-dihydroxyphenylalanine and dopaquinone modifications on protein tyrosine: links to mitochondrially derived oxidative stress via hydroxyl radical. Mol Cell Proteomics 2010; 9:1199-208. [PMID: 20124354 DOI: 10.1074/mcp.m900321-mcp200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Oxidative modifications of protein tyrosines have been implicated in multiple human diseases. Among these modifications, elevations in levels of 3,4-dihydroxyphenylalanine (DOPA), a major product of hydroxyl radical addition to tyrosine, has been observed in a number of pathologies. Here we report the first proteome survey of endogenous site-specific modifications, i.e. DOPA and its further oxidation product dopaquinone in mouse brain and heart tissues. Results from LC-MS/MS analyses included 50 and 14 DOPA-modified tyrosine sites identified from brain and heart, respectively, whereas only a few nitrotyrosine-containing peptides, a more commonly studied marker of oxidative stress, were detectable, suggesting the much higher abundance for DOPA modification as compared with tyrosine nitration. Moreover, 20 and 12 dopaquinone-modified peptides were observed from brain and heart, respectively; nearly one-fourth of these peptides were also observed with DOPA modification on the same sites. For both tissues, these modifications are preferentially found in mitochondrial proteins with metal binding properties, consistent with metal-catalyzed hydroxyl radical formation from mitochondrial superoxide and hydrogen peroxide. These modifications also link to a number of mitochondrially associated and other signaling pathways. Furthermore, many of the modification sites were common sites of previously reported tyrosine phosphorylation, suggesting potential disruption of signaling pathways. Collectively, the results suggest that these modifications are linked with mitochondrially derived oxidative stress and may serve as sensitive markers for disease pathologies.
Collapse
Affiliation(s)
- Xu Zhang
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Masoud A, Kiran R, Sandhir R. Impaired mitochondrial functions in organophosphate induced delayed neuropathy in rats. Cell Mol Neurobiol 2009; 29:1245-55. [PMID: 19517227 DOI: 10.1007/s10571-009-9420-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2009] [Accepted: 05/22/2009] [Indexed: 12/15/2022]
Abstract
Acute exposure to organophosphates induces a delayed neurodegenerative condition known as organophosphate-induced delayed neuropathy (OPIDN). The mechanism of OPIDN has not been fully understood as it does not involve cholinergic crisis. The present study has been designed to evaluate the role of mitochondrial dysfunctions in the development of OPIDN. OPIDN was induced in rats by administering acute dose of monocrotophos (MCP, 20 mg/kg body weight, orally) or dichlorvos (DDVP, 200 mg/kg body weight, subcutaneously), 15-20 min after treatment with antidotes [atropine (20 mg/kg body weight) and 2-PAM (100 mg/kg body weight) intraperitoneally]. MDA levels were observed to be higher and thiol content was lower in mitochondria from brain regions of OP exposed animals. This was accompanied by decreased activities of the mitochondrial enzymes; NADH dehydrogenase, succinate dehydrogenase, and cytochrome oxidase. In addition, mitochondrial functions assessed by MTT reduction also confirmed mitochondrial dysfunctions following development of OPIDN. The spatial long-term memory evaluated using elevated plus-maze test was observed to be deficit in OPIDN. The results suggest impaired mitochondrial functions as a mechanism involved in the development of organophosphate induced delayed neuropathy.
Collapse
Affiliation(s)
- Anwar Masoud
- Department of Biochemistry, Basic Medical Science Building, Panjab University, Chandigarh, 160014, India
| | | | | |
Collapse
|
40
|
Chowanadisai W, Bauerly KA, Tchaparian E, Wong A, Cortopassi GA, Rucker RB. Pyrroloquinoline quinone stimulates mitochondrial biogenesis through cAMP response element-binding protein phosphorylation and increased PGC-1alpha expression. J Biol Chem 2009; 285:142-52. [PMID: 19861415 DOI: 10.1074/jbc.m109.030130] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Bioactive compounds reported to stimulate mitochondrial biogenesis are linked to many health benefits such increased longevity, improved energy utilization, and protection from reactive oxygen species. Previously studies have shown that mice and rats fed diets lacking in pyrroloquinoline quinone (PQQ) have reduced mitochondrial content. Therefore, we hypothesized that PQQ can induce mitochondrial biogenesis in mouse hepatocytes. Exposure of mouse Hepa1-6 cells to 10-30 microm PQQ for 24-48 h resulted in increased citrate synthase and cytochrome c oxidase activity, Mitotracker staining, mitochondrial DNA content, and cellular oxygen respiration. The induction of this process occurred through the activation of cAMP response element-binding protein (CREB) and peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha), a pathway known to regulate mitochondrial biogenesis. PQQ exposure stimulated phosphorylation of CREB at serine 133, activated the promoter of PGC-1alpha, and increased PGC-1alpha mRNA and protein expression. PQQ did not stimulate mitochondrial biogenesis after small interfering RNA-mediated reduction in either PGC-1alpha or CREB expression. Consistent with activation of the PGC-1alpha pathway, PQQ increased nuclear respiratory factor activation (NRF-1 and NRF-2) and Tfam, TFB1M, and TFB2M mRNA expression. Moreover, PQQ protected cells from mitochondrial inhibition by rotenone, 3-nitropropionic acid, antimycin A, and sodium azide. The ability of PQQ to stimulate mitochondrial biogenesis accounts in part for action of this compound and suggests that PQQ may be beneficial in diseases associated with mitochondrial dysfunction.
Collapse
|
41
|
Cruciferous nutraceutical 3H-1,2-dithiole-3-thione protects human primary astrocytes against neurocytotoxicity elicited by MPTP, MPP(+), 6-OHDA, HNE and acrolein. Neurochem Res 2009; 34:1924-34. [PMID: 19408115 DOI: 10.1007/s11064-009-9978-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2009] [Accepted: 04/16/2009] [Indexed: 12/31/2022]
Abstract
Astrocytes possess important roles in maintaining normal brain function and providing trophic support to the neurons. They also suffer a range of toxic insults, being a chief target of prooxidants such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), 1-methyl-4-phenylpyridinium (MPP(+)), 6-hydroxydopamine (6-OHDA), 4-hydroxy-2-nonenal (HNE), and acrolein. Recently, we have observed that the cellular antioxidants and phase 2 enzymes can be upregulated by 3H-1,2-dithiole-3-thione (D3T), a nutraceutical found in cruciferous vegetables, against many prooxidants in human neuroblastoma cell lines (SH-SY5Y). However, the regulation of the above cellular factors by D3T in astrocytes and their role in ameliorating the neurotoxic effects of the above neurotoxins have not been investigated. In this study, we show that incubation of human primary astrocytes with micromolar concentrations (5-100 microM) of D3T for 24 h resulted in significant increases in the levels of reduced glutathione (GSH), glutathione reductase (GR), and the phase 2 enzyme NAD(P)H:quinone oxidoreductase 1 (NQO1). D3T treatment also caused time-dependent increases in mRNA expression of the gamma-glutamylcysteine ligase catalytic subunit (GCLC), GR, and of NQO1 in these cells. Pretreatment of astrocytes with D3T was found to afford remarkable protection against the neurocytotoxicity elicited by MPTP, MPP(+), 6-OHDA, HNE and acrolein. Taken together, this study demonstrates for the first time that in human astrocytes, the cruciferous nutraceutical D3T potently induces the cellular GSH system and the phase 2 enzyme NQO1, which is accompanied by dramatically increased resistance of these cells to the damage induced by various neurotoxicants. The results of this study may have important implications for the development of novel neuroprotective strategies.
Collapse
|
42
|
Di Giovanni G, Esposito E, Di Matteo V. In vivo microdialysis in Parkinson's research. JOURNAL OF NEURAL TRANSMISSION. SUPPLEMENTUM 2009:223-43. [PMID: 20411781 DOI: 10.1007/978-3-211-92660-4_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that is primarily characterized by the degeneration of dopamine (DA) neurons in the nigrostriatal system, which in turn produces profound neurochemical changes within the basal ganglia, representing the neural substrate for parkinsonian motor symptoms. The pathogenesis of the disease is still not completely understood, but environmental and genetic factors are thought to play important roles. Research into the pathogenesis and the development of new therapeutic intervention strategies that will slow or stop the progression of the disease in human has rapidly advanced by the use of neurotoxins that specifically target DA neurons. Over the years, a broad variety of experimental models of the disease has been developed and applied in diverse animal species. The two most common toxin models used employ 6-hydroxydopamine (6-OHDA) and the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine/1-methyl-4-phenilpyridinium ion (MPTP/MPP+), either given systemically or locally applied into the nigrostriatal pathway, to resemble PD features in animals. Both neurotoxins selectively and rapidly destroy catecolaminergic neurons, although with different mechanisms. Since in vivo microdialysis coupled to high-performance liquid chromatography is an established technique for studying physiological, pharmacological, and pathological changes of a wide range of low molecular weight substances in the brain extracellular fluid, here we review the most prominent animal and human data obtained by the use of this technique in PD research.
Collapse
Affiliation(s)
- Giuseppe Di Giovanni
- Dipartimento di Medicina Sperimentale, Sezione di Fisiologia Umana, G. Pagano, Universitá degli Studi di Palermo, 90134, Palermo, Italy
| | | | | |
Collapse
|
43
|
Ortiz GG, Benítez-King GA, Rosales-Corral SA, Pacheco-Moisés FP, Velázquez-Brizuela IE. Cellular and biochemical actions of melatonin which protect against free radicals: role in neurodegenerative disorders. Curr Neuropharmacol 2008; 6:203-14. [PMID: 19506721 PMCID: PMC2687933 DOI: 10.2174/157015908785777201] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2007] [Revised: 01/01/2008] [Accepted: 02/19/2008] [Indexed: 01/21/2023] Open
Abstract
Molecular oxygen is toxic for anaerobic organisms but it is also obvious that oxygen is poisonous to aerobic organisms as well, since oxygen plays an essential role for inducing molecular damage. Molecular oxygen is a triplet radical in its ground-stage (.O-O.) and has two unpaired electrons that can undergoes consecutive reductions of one electron and generates other more reactive forms of oxygen known as free radicals and reactive oxygen species. These reactants (including superoxide radicals, hydroxyl radicals) possess variable degrees of toxicity. Nitric oxide (NO*) contains one unpaired electron and is, therefore, a radical. NO* is generated in biological tissues by specific nitric oxide synthases and acts as an important biological signal. Excessive nitric oxide production, under pathological conditions, leads to detrimental effects of this molecule on tissues, which can be attributed to its diffusion-limited reaction with superoxide to form the powerful and toxic oxidant, peroxynitrite.Reactive oxygen and nitrogen species are molecular "renegades"; these highly unstable products tend to react rapidly with adjacent molecules, donating, abstracting, or even sharing their outer orbital electron(s). This reaction not only changes the target molecule, but often passes the unpaired electron along to the target, generating a second free radical, which can then go on to react with a new target amplifying their effects.This review describes the mechanisms of oxidative damage and its relationship with the most highly studied neurodegenerative diseases and the roles of melatonin as free radical scavenger and neurocytoskeletal protector.
Collapse
Affiliation(s)
- Genaro G Ortiz
- Laboratorio de Desarrollo-Envejecimiento, Enfermedades Neurodegenerativas, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, IMSS, Sierra Mojada 800 C.P. 44340 Guadalajara, Jalisco, México.
| | | | | | | | | |
Collapse
|
44
|
Chapter Two Evaluation of Some Cell Death Features by Real Time Real Space Microscopy. Methods Enzymol 2008; 442:27-50. [DOI: 10.1016/s0076-6879(08)01402-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
45
|
Langerveld AJ, Mihalko D, DeLong C, Walburn J, Ide CF. Gene expression changes in postmortem tissue from the rostral pons of multiple system atrophy patients. Mov Disord 2007; 22:766-77. [PMID: 17290454 DOI: 10.1002/mds.21259] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Multiple system atrophy (MSA) is a neurodegenerative disease characterized by various degrees of Parkinsonism, cerebellar ataxia, and autonomic dysfunction. In this report, Affymetrix DNA microarrays were used to measure changes in gene expression in the rostral pons, an area that undergoes extensive damage in MSA, but not other synucleinopathies. Significant changes in expression of 254 genes (180 downregulated and 74 upregulated) occurred in pons tissue from MSA patients when compared with control patients. The downregulated genes were primarily associated with biological functions known to be impaired in Parkinson's disease (PD) and other neurological diseases; for example, downregulation occurred in genes associated with mitochondrial function, ubiquitin-proteasome function, protein modification, glycolysis/metabolism, and ion transport. On the other hand, upregulated genes were associated with transcription/RNA modification, inflammation, immune system function, and oligodendrocyte maintenance and function. Immunocytochemistry, in conjunction with quantitative image analysis, was carried out to characterize alpha-synuclein protein expression as glial cytoplasmic inclusions in the pontocerebellar tract in rostral pons tissue and to determine the relationship between the amount of aggregated alpha-synuclein protein and changes in specific gene expression. Of the regulated genes, 86 were associated with the amount of observed aggregated alpha-synuclein protein in the rostral pons tissue. These data indicate that cells in the pons of MSA patients show changes in gene expression previously associated with the substantia nigra of PD patients and/or other neurological diseases, with additional changes, for example related to oligodendrocyte function unique to MSA.
Collapse
|
46
|
Liu W, Gnanasambandam R, Benjamin J, Kaur G, Getman PB, Siegel AJ, Shortridge RD, Singh S. Mutations in cytochrome c oxidase subunit VIa cause neurodegeneration and motor dysfunction in Drosophila. Genetics 2007; 176:937-46. [PMID: 17435251 PMCID: PMC1894620 DOI: 10.1534/genetics.107.071688] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mitochondrial dysfunction is involved in many neurodegenerative disorders in humans. Here we report mutations in a gene (designated levy) that codes for subunit VIa of cytochrome c oxidase (COX). The mutations were identified by the phenotype of temperature-induced paralysis and showed the additional phenotypes of decreased COX activity, age-dependent bang-induced paralysis, progressive neurodegeneration, and reduced life span. Germ-line transformation using the levy(+) gene rescued the mutant flies from all phenotypes including neurodegeneration. The data from levy mutants reveal a COX-mediated pathway in Drosophila, disruption of which leads to mitochondrial encephalomyopathic effects including neurodegeneration, motor dysfunction, and premature death. The data present the first case of a mutation in a nuclear-encoded structural subunit of COX that causes mitochondrial encephalomyopathy rather than lethality, whereas several previous attempts to identify such mutations have not been successful. The levy mutants provide a genetic model to understand the mechanisms underlying COX-mediated mitochondrial encephalomyopathies and to explore possible therapeutic interventions.
Collapse
Affiliation(s)
- Wensheng Liu
- Department of Pharmacology and Toxicology, State University of New York, Buffalo, New York 14214 and Department of Biological Sciences, State University of New York, Buffalo, New York 14260
| | - Radhakrishnan Gnanasambandam
- Department of Pharmacology and Toxicology, State University of New York, Buffalo, New York 14214 and Department of Biological Sciences, State University of New York, Buffalo, New York 14260
| | - Jeffery Benjamin
- Department of Pharmacology and Toxicology, State University of New York, Buffalo, New York 14214 and Department of Biological Sciences, State University of New York, Buffalo, New York 14260
| | - Gunisha Kaur
- Department of Pharmacology and Toxicology, State University of New York, Buffalo, New York 14214 and Department of Biological Sciences, State University of New York, Buffalo, New York 14260
| | - Patricia B. Getman
- Department of Pharmacology and Toxicology, State University of New York, Buffalo, New York 14214 and Department of Biological Sciences, State University of New York, Buffalo, New York 14260
| | - Alan J. Siegel
- Department of Pharmacology and Toxicology, State University of New York, Buffalo, New York 14214 and Department of Biological Sciences, State University of New York, Buffalo, New York 14260
| | - Randall D. Shortridge
- Department of Pharmacology and Toxicology, State University of New York, Buffalo, New York 14214 and Department of Biological Sciences, State University of New York, Buffalo, New York 14260
| | - Satpal Singh
- Department of Pharmacology and Toxicology, State University of New York, Buffalo, New York 14214 and Department of Biological Sciences, State University of New York, Buffalo, New York 14260
- Corresponding author: Department of Pharmacology and Toxicology, 102 Farber Hall, State University of New York, Buffalo, NY 14214-3000. E-mail:
| |
Collapse
|
47
|
Okawara M, Katsuki H, Kurimoto E, Shibata H, Kume T, Akaike A. Resveratrol protects dopaminergic neurons in midbrain slice culture from multiple insults. Biochem Pharmacol 2006; 73:550-60. [PMID: 17147953 DOI: 10.1016/j.bcp.2006.11.003] [Citation(s) in RCA: 154] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2006] [Revised: 10/31/2006] [Accepted: 11/06/2006] [Indexed: 12/16/2022]
Abstract
Increasing lines of evidence show that resveratrol, a polyphenol compound contained in several dietary products, exhibits cytoprotective actions. Notably, resveratrol activates sirtuin family of NAD-dependent histone deacetylases implicated in regulation of various cellular processes including gene transcription, DNA repair and apoptosis. Here we examined neuroprotective effect of resveratrol on dopaminergic neurons in organotypic midbrain slice culture. Resveratrol and quercetin, another sirtuin-activating polyphenol, prevented the decrease of dopaminergic neurons and the increase of propidium iodide uptake into slices induced by a dopaminergic neurotoxin 1-methyl-4-phenyl pyridinium (MPP(+)). Resveratrol also provided concentration-dependent neuroprotective effects against sodium azide, a mitochondrial complex IV inhibitor, and thrombin (EC number 3.4.21.5), a microglia-activating agent. Sirtuin inhibitors such as nicotinamide and sirtinol did not attenuate the protective effect of resveratrol against MPP(+) cytotoxicity. Instead, we found that resveratrol prevented accumulation of reactive oxygen species, depletion of cellular glutathione, and cellular oxidative damage induced by MPP(+), suggesting involvement of antioxidative properties in the neuroprotective action of resveratrol. On the other hand, resveratrol as well as a sirtuin activator NAD inhibited dopaminergic neurotoxicity of a DNA alkylating agent, N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). Moreover, MNNG-induced increase in acetylation of p53, a representative target of sirtuin deacetylase activity, was suppressed by resveratrol. These results indicate that resveratrol can exert neuroprotective actions in dopaminergic neurons. Either antioxidative activity or sirtuin-activating potential may play an important role in the neuroprotectice actions of resveratrol against different kinds of insults.
Collapse
Affiliation(s)
- Mitsugi Okawara
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Obata T. Effect of desferrioxamine, a strong iron (III) chelator, on 1-methyl-4-phenylpyridinium ion (MPP+)-induced hydroxyl radical generation in the rat striatum. Eur J Pharmacol 2006; 539:34-8. [PMID: 16650845 DOI: 10.1016/j.ejphar.2006.03.071] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Revised: 03/17/2006] [Accepted: 03/28/2006] [Indexed: 11/26/2022]
Abstract
The present study was examined that the desferrioxamine, a strong iron (III) chelator, enhanced 1 methyl-4-phenylpyridinium ion (MPP+)-induced hydroxyl radical (*OH) generation in the extracellular fluid of caudate nucleus anesthetized rats. Rats were anesthetized, and sodium salicylate in Ringer's solution (0.5 nmol/microl/min) was infused through a microdialysis probe to detect the generation of *OH as reflected by the non-enzymatic formation of 2,3-dihydroxybenzoic acid (DHBA) in the striatum. Induction of desferrioxamine (50 microM) drastically increased the formation of *OH trapped as 2,3-DHBA by the action of MPP+, as compared with MPP+-only-treated animals. Although desferrioxamine did not change the levels of MPP+-induced dopamine, a marked elevation of *OH formation trapped as 2,3-DHBA was observed. When corresponding experiments were performed with reserpinized animals, the level of dopamine and 2,3-DHBA drastically decreased. However, the level of dopamine did not change, but desferrioxamine significantly increased the level of 2,3-DHBA in reserpinized animals. Iron (III) decreased MPP+-induced 2,3-DHBA formations in the presence of dopamine (10 microM). Moreover, when iron (II) was administered to desferrioxamine-treated animals, a marked elevation of 2,3-DHBA was observed, compared with MPP+-only-treated animals, that showed a positive linear correlation between iron (II) and *OH formation trapped as 2,3-DHBA (R2=0.981) in the dialysate. The present study indicates that the suppression of MPP+-induced *OH formation by iron (III) may play a key role in protective effect of iron (III) on the rat brain.
Collapse
Affiliation(s)
- Toshio Obata
- Department of Analytical Chemistry, Ohu University School of Pharmaceutical Sciences, Koriyama, Fukushima 963-8611, Japan.
| |
Collapse
|
49
|
Obata T. Activation of protein kinase C enhances 1-methyl-4-phenylpyridinium ion (MPP+)-induced hydroxylradical generation in rat striatum. Neurosci Lett 2006; 398:50-2. [PMID: 16406319 DOI: 10.1016/j.neulet.2005.12.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2005] [Revised: 11/12/2005] [Accepted: 12/14/2005] [Indexed: 11/30/2022]
Abstract
The present study examined the effect of chelerlythrine, a protein kinase C (PKC) inhibitor, on 1-methyl-4-phenylpyridine (MPP+)-induced hydroxyl radicals (*OH) in rat striatum. Rats were anesthetized, and sodium salicylate (0.5 mM or 0.5 nmol/microl/min) was infused through a microdialysis probe to detect the *OH generation as reflected by the non-enzymatic formation of 2,3-dihydroxybenzoic acid (2,3-DHBA) in the striatum. Dopamine (DA)-selective neurotoxin, MPP+, infusion into the striatum of rats induces *OH formation, trapped as 2,3-DHBA. The application of chelerythrine, a potent and selective protein kinase C (PKC) inhibitor, suppressed MPP+ -induced *OH formation. The results in the present study suggests the protective effect of chelerythrine on *OH generation induced by MPP+.
Collapse
Affiliation(s)
- Toshio Obata
- Department of Analytical Chemistry, Ohu University School of Pharmaceutical Sciences, Koriyama, Fukushima 963-8611, Japan.
| |
Collapse
|
50
|
Obata T. Tamoxifen protect against hydroxyl radical generation induced by phenelzine in rat striatum. Toxicology 2006; 222:46-52. [PMID: 16500014 DOI: 10.1016/j.tox.2006.01.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2005] [Revised: 12/27/2005] [Accepted: 01/20/2006] [Indexed: 10/25/2022]
Abstract
The present study was examined whether tamoxifen, a synthetic nonsteroidal antiestrogen, could suppress antidepressant drug phenelzine can increase an active dopaminergic neurotoxin, 1-methyl-4-phenylpyridinium ion (MPP(+))-induced hydroxyl radical (OH) generation in the extracellular fluid of rat striatum, using in vivo microdialysis system. Rats were anesthetized, and sodium salicylate (0.5 nmol/microl/min) was infused through a microdialysis probe to detect the generation of OH as reflected by the non-enzymatic formation of 2,3-dihydroxybenzoic acid (DHBA) in the striatum. Infusion of phenelzine (100 microM or 0.1 nmol/microl/min) into the striatum drastically increased dopamine (DA) efflux and the OH formation, trapped as 2,3-DHBA by the possible increased production of MPP(+). However, tamoxifen (100 microM) significantly suppressed phenelzine enhanced DA efflux and OH formation by MPP(+). These results in the present study is the first demonstration showing the protective effect of tamoxifen on OH generation induced by phenelzine enhanced MPP(+) by suppressing DA efflux.
Collapse
Affiliation(s)
- Toshio Obata
- Department of Analytical Chemistry, Ohu University, School of Pharmaceutical Sciences, Koriyama, Fukushima 963-8611, Japan.
| |
Collapse
|