1
|
Zhu CZ, Li GZ, Lyu HF, Lu YY, Li Y, Zhang XN. Modulation of autophagy by melatonin and its receptors: implications in brain disorders. Acta Pharmacol Sin 2024:10.1038/s41401-024-01398-2. [PMID: 39448859 DOI: 10.1038/s41401-024-01398-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/17/2024] [Indexed: 10/26/2024] Open
Abstract
Autophagy plays a crucial role in maintaining neuronal homeostasis and function, and its disruption is linked to various brain diseases. Melatonin, an endogenous hormone that primarily acts through MT1 and MT2 receptors, regulates autophagy via multiple pathways. Growing evidence indicates that melatonin's ability to modulate autophagy provides therapeutic and preventive benefits in brain disorders, including neurodegenerative and affective diseases. In this review, we summarize the key mechanisms by which melatonin affects autophagy and explore its therapeutic potential in the treatment of brain disorders.
Collapse
Affiliation(s)
- Chen-Ze Zhu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, 310058, China
| | - Gui-Zhi Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 311399, China
| | - Hai-Feng Lyu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 311399, China
| | - Yang-Yang Lu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, 310058, China
| | - Yue Li
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, 310058, China
| | - Xiang-Nan Zhang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, 310058, China.
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China.
| |
Collapse
|
2
|
Ahmadi S, Taghizadieh M, Mehdizadehfar E, Hasani A, Khalili Fard J, Feizi H, Hamishehkar H, Ansarin M, Yekani M, Memar MY. Gut microbiota in neurological diseases: Melatonin plays an important regulatory role. Biomed Pharmacother 2024; 174:116487. [PMID: 38518598 DOI: 10.1016/j.biopha.2024.116487] [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: 11/23/2023] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024] Open
Abstract
Melatonin is a highly conserved molecule produced in the human pineal gland as a hormone. It is known for its essential biological effects, such as antioxidant activity, circadian rhythm regulator, and immunomodulatory effects. The gut is one of the primary known sources of melatonin. The gut microbiota helps produce melatonin from tryptophan, and melatonin has been shown to have a beneficial effect on gut barrier function and microbial population. Dysbiosis of the intestinal microbiota is associated with bacterial imbalance and decreased beneficial microbial metabolites, including melatonin. In this way, low melatonin levels may be related to several human diseases. Melatonin has shown both preventive and therapeutic effects against various conditions, including neurological diseases such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis. This review was aimed to discuss the role of melatonin in the body, and to describe the possible relationship between gut microbiota and melatonin production, as well as the potential therapeutic effects of melatonin on neurological diseases.
Collapse
Affiliation(s)
- Somayeh Ahmadi
- Students Research Committee, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Taghizadieh
- Department of Pathology, School of Medicine, Center for Women's Health Research Zahra, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Mehdizadehfar
- Department of Neurosciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alka Hasani
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Clinical Research Development Unit, Sina Educational, Research and Treatment Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Khalili Fard
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Feizi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Microbiology, Aalinasab Hospital, Social Security Organization, Tabriz, Iran
| | - Hammed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masood Ansarin
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mina Yekani
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Haddadi R, Eyvari-Brooshghalan S, Makhdoomi S, Fadaiie A, Komaki A, Daneshvar A. Neuroprotective effects of silymarin in 3-nitropropionic acid-induced neurotoxicity in male mice: improving behavioral deficits by attenuating oxidative stress and neuroinflammation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2447-2463. [PMID: 37847410 DOI: 10.1007/s00210-023-02776-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/04/2023] [Indexed: 10/18/2023]
Abstract
3-Nitropropionic acid (3-NP) is strongly believed to be an irreversible inhibitor of mitochondrial complex II, leading to neural damage. This study aimed to investigate the neuroprotective effects of silymarin against 3-NP-induced neurotoxicity in male mice. Six-week-old mice received subacute doses of 3-NP intraperitoneally for 17 days. Mice were given silymarin (70 mg/kg/day, P.O.) for 2 weeks before 3-NP administration or for 4 weeks after 3-NP administration. At the end of the treatment schedule, animals were evaluated for behavioral alterations. Subsequently, neuronal damage in the hippocampus region of the brain tissues, oxidative stress-related parameters (lipid peroxidation, nitric oxide, superoxide dismutase, glutathione, and total antioxidant capacity), and pro-inflammatory cytokine (TNF-α, IL-17, and IL-1β) levels were evaluated. Our results indicated that 3-NP treatment significantly (p < 0.05) tended to reduce motor coordination, memory, and neuronal antioxidant status while increasing pro-inflammatory cytokine levels. However, silymarin in both treatment and pretreatment protocols markedly (p < 0.05) attenuated the behavioral deficits, oxidative stress status, and neuroinflammation. The results of the current study suggest that the neuroprotective effect of silymarin against 3-NP-induced neurotoxicity might be due to the mitigation of oxidative stress status and provide insight into the therapeutic potential of silymarin.
Collapse
Affiliation(s)
- Rasool Haddadi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran.
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Medicinal Plant and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, 6517838678, Iran.
| | - Shahla Eyvari-Brooshghalan
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sajjad Makhdoomi
- Department of Pharmacology and Toxicology, School of Pharmacy, Medicinal Plant and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, 6517838678, Iran
| | - Ahmad Fadaiie
- Department of Pharmacology and Toxicology, School of Pharmacy, Medicinal Plant and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, 6517838678, Iran
| | - Alireza Komaki
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Afsoon Daneshvar
- Department of Pharmacology and Toxicology, School of Pharmacy, Medicinal Plant and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, 6517838678, Iran
| |
Collapse
|
4
|
Kim J, Li W, Wang J, Baranov SV, Heath BE, Jia J, Suofu Y, Baranova OV, Wang X, Larkin TM, Lariviere WR, Carlisle DL, Friedlander RM. Biosynthesis of neuroprotective melatonin is dysregulated in Huntington's disease. J Pineal Res 2023; 75:e12909. [PMID: 37721126 PMCID: PMC10592086 DOI: 10.1111/jpi.12909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 07/27/2023] [Accepted: 08/13/2023] [Indexed: 09/19/2023]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative brain disorder associated with uncontrolled body movements, cognitive decline, and reduced circulating melatonin levels. Melatonin is a potent antioxidant and exogenous melatonin treatment is neuroprotective in experimental HD models. In neurons, melatonin is exclusively synthesized in the mitochondrial matrix. Thus, we investigated the integrity of melatonin biosynthesis pathways in pineal and extrapineal brain areas in human HD brain samples, in the R6/2 mouse model of HD and in full-length mutant huntingtin knock-in cells. Aralkylamine N-acetyltransferase (AANAT) is the rate-limiting step enzyme in the melatonin biosynthetic pathway. We found that AANAT expression is significantly decreased in the pineal gland and the striatum of HD patients compared to normal controls. In the R6/2 mouse forebrain, AANAT protein expression was decreased in synaptosomal, but not nonsynaptosomal, mitochondria and was associated with decreased synaptosomal melatonin levels compared to wild type mice. We also demonstrate sequestration of AANAT in mutant-huntingtin protein aggregates likely resulting in decreased AANAT bioavailability. Paradoxically, AANAT mRNA expression is increased in tissues where AANAT protein expression is decreased, suggesting a potential feedback loop that is, ultimately unsuccessful. In conclusion, we demonstrate that pineal, extrapineal, and synaptosomal melatonin levels are compromised in the brains of HD patients and R6/2 mice due, at least in part, to protein aggregation.
Collapse
Affiliation(s)
- Jinho Kim
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Wei Li
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jingjing Wang
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sergei V Baranov
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Brianna E Heath
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jiaoying Jia
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yalikun Suofu
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Oxana V Baranova
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Xiaomin Wang
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Timothy M Larkin
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - William R Lariviere
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Diane L Carlisle
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Robert M Friedlander
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
5
|
Verma AK, Singh S, Rizvi SI. Therapeutic potential of melatonin and its derivatives in aging and neurodegenerative diseases. Biogerontology 2023; 24:183-206. [PMID: 36550377 DOI: 10.1007/s10522-022-10006-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Aging is associated with increasing impairments in brain homeostasis and represents the main risk factor across most neurodegenerative disorders. Melatonin, a neuroendocrine hormone that regulates mammalian chronobiology and endocrine functions is well known for its antioxidant potential, exhibiting both cytoprotective and chronobiotic abilities. Age-related decline of melatonin disrupting mitochondrial homeostasis and cytosolic DNA-mediated inflammatory reactions in neurons is a major contributory factor in the emergence of neurological abnormalities. There is scattered literature on the possible use of melatonin against neurodegenerative mechanisms in the aging process and its associated diseases. We have searched PUBMED with many combinations of key words for available literature spanning two decades. Based on the vast number of experimental papers, we hereby review recent advancements concerning the potential impact of melatonin on cellular redox balance and mitochondrial dynamics in the context of neurodegeneration. Next, we discuss a broader explanation of the involvement of disrupted redox homeostasis in the pathophysiology of age-related diseases and its connection to circadian mechanisms. Our effort may result in the discovery of novel therapeutic approaches. Finally, we summarize the current knowledge on molecular and circadian regulatory mechanisms of melatonin to overcome neurodegenerative diseases (NDDs) such as Alzheimer's, Parkinson's, Huntington's disease, and amyotrophic lateral sclerosis, however, these findings need to be confirmed by larger, well-designed clinical trials. This review is also expected to uncover the associated molecular alterations in the aging brain and explain how melatonin-mediated circadian restoration of neuronal homeodynamics may increase healthy lifespan in age-related NDDs.
Collapse
Affiliation(s)
- Avnish Kumar Verma
- Department of Biochemistry, University of Allahabad, Allahabad, 211002, India
| | - Sandeep Singh
- Biological Psychiatry Laboratory, Hadassah Medical Center - Hebrew University, Jerusalem, Israel
| | - Syed Ibrahim Rizvi
- Department of Biochemistry, University of Allahabad, Allahabad, 211002, India.
| |
Collapse
|
6
|
D’Egidio F, Castelli V, Cimini A, d’Angelo M. Cell Rearrangement and Oxidant/Antioxidant Imbalance in Huntington's Disease. Antioxidants (Basel) 2023; 12:571. [PMID: 36978821 PMCID: PMC10045781 DOI: 10.3390/antiox12030571] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Huntington's Disease (HD) is a hereditary neurodegenerative disorder caused by the expansion of a CAG triplet repeat in the HTT gene, resulting in the production of an aberrant huntingtin (Htt) protein. The mutant protein accumulation is responsible for neuronal dysfunction and cell death. This is due to the involvement of oxidative damage, excitotoxicity, inflammation, and mitochondrial impairment. Neurons naturally adapt to bioenergetic alteration and oxidative stress in physiological conditions. However, this dynamic system is compromised when a neurodegenerative disorder occurs, resulting in changes in metabolism, alteration in calcium signaling, and impaired substrates transport. Thus, the aim of this review is to provide an overview of the cell's answer to the stress induced by HD, focusing on the role of oxidative stress and its balance with the antioxidant system.
Collapse
Affiliation(s)
| | | | | | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
7
|
Muñoz-Jurado A, Escribano BM, Agüera E, Caballero-Villarraso J, Galván A, Túnez I. SARS-CoV-2 infection in multiple sclerosis patients: interaction with treatments, adjuvant therapies, and vaccines against COVID-19. J Neurol 2022; 269:4581-4603. [PMID: 35788744 PMCID: PMC9253265 DOI: 10.1007/s00415-022-11237-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 12/12/2022]
Abstract
The SARS-CoV-2 pandemic has raised particular concern for people with Multiple Sclerosis, as these people are believed to be at increased risk of infection, especially those being treated with disease-modifying therapies. Therefore, the objective of this review was to describe how COVID-19 affects people who suffer from Multiple Sclerosis, evaluating the risk they have of suffering an infection by this virus, according to the therapy to which they are subjected as well as the immune response of these patients both to infection and vaccines and the neurological consequences that the virus can have in the long term. The results regarding the increased risk of infection due to treatment are contradictory. B-cell depletion therapies may cause patients to have a lower probability of generating a detectable neutralizing antibody titer. However, more studies are needed to help understand how this virus works, paying special attention to long COVID and the neurological symptoms that it causes.
Collapse
Affiliation(s)
- Ana Muñoz-Jurado
- Department of Cell Biology, Physiology and Immunology, Faculty of Veterinary Medicine, University of Cordoba, Campus of Rabanales, 14071 Cordoba, Spain
| | - Begoña M. Escribano
- Department of Cell Biology, Physiology and Immunology, Faculty of Veterinary Medicine, University of Cordoba, Campus of Rabanales, 14071 Cordoba, Spain
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain
| | - Eduardo Agüera
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain
- Neurology Service, Reina Sofia University Hospital, Cordoba, Spain
| | - Javier Caballero-Villarraso
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Av. Menendez Pidal, 14004 Cordoba, Spain
- Clinical Analysis Service, Reina Sofía University Hospital, Cordoba, Spain
| | - Alberto Galván
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Av. Menendez Pidal, 14004 Cordoba, Spain
| | - Isaac Túnez
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Av. Menendez Pidal, 14004 Cordoba, Spain
- Cooperative Research Thematic Excellent Network on Brain Stimulation (REDESTIM), Madrid, Spain
| |
Collapse
|
8
|
Pharmacological Mechanism of Zuojin Pill for Gastroesophageal Reflux Disease: A Network Pharmacology Study. J FOOD QUALITY 2022. [DOI: 10.1155/2022/5933348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Background. Although Zuojin Pill (ZJP) is widely used in China as a traditional prescription to treat gastroesophageal reflux disease (GERD), its exact mechanism of action is still unknown. Therefore, we employed network pharmacology (NP), molecular docking (MD), and molecular dynamics simulation (MDS) to investigate the pharmacological mechanisms of ZJP against GERD. Methods. Active compounds and target genes corresponding to ZJP and target genes related to GERD were identified through analysis of publicly available datasets. Subsequently, the obtained data were subjected to further network pharmacological analysis to explore the potential key active compounds, core target genes, and biological processes (BPs) associated with the effect of ZJP against GERD. Finally, the prediction results of NP were validated by MD, and MDS of the optimal core protein-ligand for each component obtained by MD were performed using Gromacs 2020 software. Results. Twelve active components of ZJP were identified to act on 82 target genes associated with GERD, and ZJP might exert an anti-GERD effect through the regulation of BPs such as reactive oxygen species (ROS) metabolism, response to oxidative stress (OS), and ROS, as well as the activation of signaling pathways such as apoptosis, p53 signaling, chemical carcinogenesis-ROS, and HIF-1 signaling pathways. Furthermore, quercetin, kaempferol, and coptisine, the three key components of ZJP were shown to stably bond with the 14 core target genes, including AKT1, MMP2, TP53, EGFR, JUN, CASP3, CXCL8, HIF1α, IL-1β, MYC, PPARG, MMP9, PTGS2, and FOS. Results from MDS showed that PPARG-quercetin and MMP2-quercetin bound more stably. Conclusions. The findings suggest that ZJP alleviates the symptoms of GERD and improves the prognosis by regulating ROS metabolism, thereby reducing the secretion of proinflammatory cytokines like IL-1β, COX-2, CXCL8, and MMPs, regulating the expression of oncogenes such as JUN and FOS, and maintaining the normal expression of tumor suppressor genes such as TP53 and MYC. However, whether the effect of this modulation of ROS metabolism is positive or negative needs to be further verified by pharmacological experiments.
Collapse
|
9
|
Melatonin and multiple sclerosis: antioxidant, anti-inflammatory and immunomodulator mechanism of action. Inflammopharmacology 2022; 30:1569-1596. [PMID: 35665873 PMCID: PMC9167428 DOI: 10.1007/s10787-022-01011-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/13/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Melatonin is an indole hormone secreted primarily by the pineal gland that showing anti-oxidant, anti-inflammatory and anti-apoptotic capacity. It can play an important role in the pathophysiological mechanisms of various diseases. In this regard, different studies have shown that there is a relationship between Melatonin and Multiple Sclerosis (MS). MS is a chronic immune-mediated disease of the Central Nervous System. AIM The objective of this review was to evaluate the mechanisms of action of melatonin on oxidative stress, inflammation and intestinal dysbiosis caused by MS, as well as its interaction with different hormones and factors that can influence the pathophysiology of the disease. RESULTS Melatonin causes a significant increase in the levels of catalase, superoxide dismutase, glutathione peroxidase, glutathione and can counteract and inhibit the effects of the NLRP3 inflammasome, which would also be beneficial during SARS-CoV-2 infection. In addition, melatonin increases antimicrobial peptides, especially Reg3β, which could be useful in controlling the microbiota. CONCLUSION Melatonin could exert a beneficial effect in people suffering from MS, running as a promising candidate for the treatment of this disease. However, more research in human is needed to help understand the possible interaction between melatonin and certain sex hormones, such as estrogens, to know the potential therapeutic efficacy in both men and women.
Collapse
|
10
|
Srinivasan E, Ram V, Rajasekaran R. A review on Huntington protein Insight into protein aggregation and therapeutic interventions. Curr Drug Metab 2022; 23:260-282. [PMID: 35319359 DOI: 10.2174/1389200223666220321103942] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/13/2021] [Accepted: 01/15/2022] [Indexed: 11/22/2022]
Abstract
Huntington disease (HD) is a distressing, innate neurodegenerative disease that descends from CAG repeat expansion in the huntingtin gene causing behavioral changes, motor dysfunction, and dementia in children and adults. Mutation in huntingtin (HTT) protein has been suggested to cause neuron loss in the cortex and striatum through various mechanisms including abnormal regulation of transcription, proteasomal dysfunction, post-translational modification, and other events, regulating toxicity. Pathogenesis of HD involves cleavage of the huntingtin protein followed by the neuronal accumulation of its aggregated form. Several research groups made possible efforts to reduce huntingtin gene expression, protein accumulation, and protein aggregation using inhibitors and molecular chaperones as developing drugs against HD. Herein, we review the mechanism proposed towards the formation of HTT protein aggregation and the impact of therapeutic strategies for the treatment of HD.
Collapse
Affiliation(s)
- E Srinivasan
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore - 632014, Tamil Nadu, India
- Department of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai - 602105, Tamil Nadu, India
| | - Vavish Ram
- Bioinformatics Lab, Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore - 632014, Tamil Nadu, India
| | - R Rajasekaran
- Bioinformatics Lab, Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore - 632014, Tamil Nadu, India
| |
Collapse
|
11
|
Escribano BM, Muñoz-Jurado A, Caballero-Villarraso J, Valdelvira ME, Giraldo AI, Paz-Rojas E, Gascón F, Santamaría A, Agüera E, Túnez I. Protective effects of melatonin on changes occurring in the experimental autoimmune encephalomyelitis model of multiple sclerosis. Mult Scler Relat Disord 2022; 58:103520. [PMID: 35038645 DOI: 10.1016/j.msard.2022.103520] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/27/2021] [Accepted: 01/09/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Melatonin has been related to the pathophysiology of multiple sclerosis (MS), and its anti-inflammatory and immunomodulatory properties have been proved in numerous neurodegenerative diseases. This study aimed to find out whether a melatonin supplement in MS is able to act as a benefit to its clinical status, i.e. oxidative stress, inflammation and indirect biomarkers of bacterial dysbiosis, lipopolysaccharide (LPS) and LPS-binding protein (LBP), verifying its therapeutic potential and its possible clinical use in patients with MS. METHODS The animal MS model, experimental autoimmune encephalomyelitis (EAE), was employed whereby 25 male Dark Agouti rats (5 animals per group) were divided into: a control group (not manipulated); a control+vehicle group; a control+melatonin group; an EAE group; an EAE+melatonin group. Melatonin was administered daily for 51 days, at a dose of 1 mg/kg body weight/i.p., once a day, five days a week. RESULTS The results from the administration of melatonin demonstrated an improvement in clinical status, a diminution in oxidative stress and inflammation, as well as in bacterial dysbiosis. CONCLUSION Melatonin could play an effective role against MS, either alone or as a therapy combined with traditional agents.
Collapse
Affiliation(s)
- Begoña María Escribano
- Department of Cell Biology, Physiology and Immunology, Faculty of Veterinary Medicine, Campus of Rabanales, University of Cordoba, Cordoba 14071, Spain; Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain.
| | - A Muñoz-Jurado
- Department of Cell Biology, Physiology and Immunology, Faculty of Veterinary Medicine, Campus of Rabanales, University of Cordoba, Cordoba 14071, Spain
| | - J Caballero-Villarraso
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain; Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Av. Menendez Pidal, Cordoba 14004, Spain; Clinical Analysis Service, Reina Sofia University Hospital, Cordoba, Spain
| | - M E Valdelvira
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Av. Menendez Pidal, Cordoba 14004, Spain
| | - A I Giraldo
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain; Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Av. Menendez Pidal, Cordoba 14004, Spain
| | - E Paz-Rojas
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain; Multiplex Biopharma S.L., Rabanales 21, Cordoba, Spain
| | - F Gascón
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain; Clinical Analysis Service, Valle de los Pedroches, Hospital, Cordoba, Spain
| | - A Santamaría
- Laboratory of Exciting Amino Acids, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - E Agüera
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain; Neurology Service, Reina Sofia University Hospital, Cordoba, Spain
| | - Isaac Túnez
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain; Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Av. Menendez Pidal, Cordoba 14004, Spain; Cooperative Research Thematic Excellent Network on Brain Stimulation (REDESTIM), Spain.
| |
Collapse
|
12
|
Purushothaman B, Sumathi T. Research Paper5,6,7 trihydroxy flavone armoured neurodegeneration caused by Quinolinic acid induced huntington's like disease in rat striatum - reinstating the level of brain neurotrophins with special reference to cognitive-socio behaviour, biochemical and histopathological aspects. Neurosci Res 2021; 174:25-35. [PMID: 34416310 DOI: 10.1016/j.neures.2021.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 08/13/2021] [Accepted: 08/15/2021] [Indexed: 10/20/2022]
Abstract
Huntington Disease (HD), a predominant Neurodegenerative Disorder which might be induced by endogenous neurotoxin called Quinolinic Acid (QA), an N-methyl-D aspartate receptor (NMDAR) agonist, the bilaterally intrastriatal administration (200 nm/2 μL of saline) offers rise to the toxic events like neuronal death, neuroinflammation by inflicting excitotoxicity and oxidative stress in the striatum of male Wistar rats by exhibiting the behavioural changes which was accessed by rotarod, open field analysis. In this study, the neuropharmacological effect of Baicalein (BC) against QA induced HD was evaluated. Baicalein (BC), scientifically 5,6,7 trihydroxy flavone present naturally in the edible plants like Scutellaria baicalensis and Oroxylum indicum possess a better neuroprotective effect in the dosage of 10 mg/kg and 30 mg/kg intraperitoneally in the striatum of HD induced rats. This study proved that BC is efficient to revive the level of enzymatic & non-enzymatic antioxidants and mitochondrial complexes by decreasing the number of inflammatory mediators such as MDA, protein carbonyls and Nitric Oxide at the significance of P < 0.01 and restores the amount of BDNF and GDNF thereby preventing the neurophysiological changes which were analysed by haematoxylin & eosin staining. Thus finally, the protective effect of Baicalein displays the up-gradation of psychological and behavioural changes induced by QA.
Collapse
Affiliation(s)
- Bhagyalakshmi Purushothaman
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, 600 113, Tamil Nadu, India
| | - Thangarajan Sumathi
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, 600 113, Tamil Nadu, India.
| |
Collapse
|
13
|
Castagnola E, Robbins EM, Woeppel KM, McGuier M, Golabchi A, Taylor IM, Michael AC, Cui XT. Real-Time Fast Scan Cyclic Voltammetry Detection and Quantification of Exogenously Administered Melatonin in Mice Brain. Front Bioeng Biotechnol 2020; 8:602216. [PMID: 33330433 PMCID: PMC7732424 DOI: 10.3389/fbioe.2020.602216] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/30/2020] [Indexed: 01/19/2023] Open
Abstract
Melatonin (MT) has been recently considered an excellent candidate for the treatment of sleep disorders, neural injuries, and neurological diseases. To better investigate the actions of MT in various brain functions, real-time detection of MT concentrations in specific brain regions is much desired. Previously, we have demonstrated detection of exogenously administered MT in anesthetized mouse brain using square wave voltammetry (SWV). Here, for the first time, we show successful detection of exogenous MT in the brain using fast scan cyclic voltammetry (FSCV) on electrochemically pre-activated carbon fiber microelectrodes (CFEs). In vitro evaluation showed the highest sensitivity (28.1 nA/μM) and lowest detection limit (20.2 ± 4.8 nM) ever reported for MT detection at carbon surface. Additionally, an extensive CFE stability and fouling assessment demonstrated that a prolonged CFE pre-conditioning stabilizes the background, in vitro and in vivo, and provides consistent CFE sensitivity over time even in the presence of a high MT concentration. Finally, the stable in vivo background, with minimized CFE fouling, allows us to achieve a drift-free FSCV detection of exogenous administered MT in mouse brain over a period of 3 min, which is significantly longer than the duration limit (usually < 90 s) for traditional in vivo FSCV acquisition. The MT concentration and dynamics measured by FSCV are in good agreement with SWV, while microdialysis further validated the concentration range. These results demonstrated reliable MT detection using FSCV that has the potential to monitor MT in the brain over long periods of time.
Collapse
Affiliation(s)
- Elisa Castagnola
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Elaine M. Robbins
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kevin M. Woeppel
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States
| | - Moriah McGuier
- Department of Chemistry, Saint Vincent College, Latrobe, PA, United States
| | - Asiyeh Golabchi
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States
| | - I. Mitch Taylor
- Department of Chemistry, Saint Vincent College, Latrobe, PA, United States
| | - Adrian C. Michael
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
14
|
Khan A, Jahan S, Imtiyaz Z, Alshahrani S, Antar Makeen H, Mohammed Alshehri B, Kumar A, Arafah A, Rehman MU. Neuroprotection: Targeting Multiple Pathways by Naturally Occurring Phytochemicals. Biomedicines 2020; 8:E284. [PMID: 32806490 PMCID: PMC7459826 DOI: 10.3390/biomedicines8080284] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/27/2020] [Accepted: 08/05/2020] [Indexed: 12/16/2022] Open
Abstract
With the increase in the expectancy of the life span of humans, neurodegenerative diseases (NDs) have imposed a considerable burden on the family, society, and nation. In defiance of the breakthroughs in the knowledge of the pathogenesis and underlying mechanisms of various NDs, very little success has been achieved in developing effective therapies. This review draws a bead on the availability of the nutraceuticals to date for various NDs (Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis, Huntington's disease, vascular cognitive impairment, Prion disease, Spinocerebellar ataxia, Spinal muscular atrophy, Frontotemporal dementia, and Pick's disease) focusing on their various mechanisms of action in various in vivo and in vitro models of NDs. This review is distinctive in its compilation to critically review preclinical and clinical studies of the maximum phytochemicals in amelioration and prevention of almost all kinds of neurodegenerative diseases and address their possible mechanism of action. PubMed, Embase, and Cochrane Library searches were used for preclinical studies, while ClinicalTrials.gov and PubMed were searched for clinical updates. The results from preclinical studies demonstrate the efficacious effects of the phytochemicals in various NDs while clinical reports showing mixed results with promise for phytochemical use as an adjunct to the conventional treatment in various NDs. These studies together suggest that phytochemicals can significantly act upon different mechanisms of disease such as oxidative stress, inflammation, apoptotic pathways, and gene regulation. However, further clinical studies are needed that should include the appropriate biomarkers of NDs and the effect of phytochemicals on them as well as targeting the appropriate population.
Collapse
Affiliation(s)
- Andleeb Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
| | - Sadaf Jahan
- Medical Laboratories Department, College of Applied Medical Sciences, Majmaah University, Majmaah 15341, Saudi Arabia; (S.J.); (B.M.A.)
| | - Zuha Imtiyaz
- Clinical Drug Development, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan;
| | - Saeed Alshahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
| | - Hafiz Antar Makeen
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
| | - Bader Mohammed Alshehri
- Medical Laboratories Department, College of Applied Medical Sciences, Majmaah University, Majmaah 15341, Saudi Arabia; (S.J.); (B.M.A.)
| | - Ajay Kumar
- Institute of Nano Science and Technology, Habitat Centre, Phase-10, Sector-64, Mohali 160062, India;
| | - Azher Arafah
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.); (M.U.R.)
| | - Muneeb U. Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.); (M.U.R.)
| |
Collapse
|
15
|
Chen D, Zhang T, Lee TH. Cellular Mechanisms of Melatonin: Insight from Neurodegenerative Diseases. Biomolecules 2020; 10:biom10081158. [PMID: 32784556 PMCID: PMC7464852 DOI: 10.3390/biom10081158] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/23/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases are the second most common cause of death and characterized by progressive impairments in movement or mental functioning in the central or peripheral nervous system. The prevention of neurodegenerative disorders has become an emerging public health challenge for our society. Melatonin, a pineal hormone, has various physiological functions in the brain, including regulating circadian rhythms, clearing free radicals, inhibiting biomolecular oxidation, and suppressing neuroinflammation. Cumulative evidence indicates that melatonin has a wide range of neuroprotective roles by regulating pathophysiological mechanisms and signaling pathways. Moreover, melatonin levels are decreased in patients with neurodegenerative diseases. In this review, we summarize current knowledge on the regulation, molecular mechanisms and biological functions of melatonin in neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, vascular dementia and multiple sclerosis. We also discuss the clinical application of melatonin in neurodegenerative disorders. This information will lead to a better understanding of the regulation of melatonin in the brain and provide therapeutic options for the treatment of various neurodegenerative diseases.
Collapse
Affiliation(s)
- Dongmei Chen
- Correspondence: (D.C.); (T.H.L.); Tel.: +86-591-2286-2498 (D.C.); +86-591-2286-2498 (T.H.L.)
| | | | - Tae Ho Lee
- Correspondence: (D.C.); (T.H.L.); Tel.: +86-591-2286-2498 (D.C.); +86-591-2286-2498 (T.H.L.)
| |
Collapse
|
16
|
Gunata M, Parlakpinar H, Acet H. Melatonin: A review of its potential functions and effects on neurological diseases. Rev Neurol (Paris) 2020; 176:148-165. [DOI: 10.1016/j.neurol.2019.07.025] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/03/2019] [Accepted: 07/16/2019] [Indexed: 12/12/2022]
|
17
|
Martinelli C, Pucci C, Battaglini M, Marino A, Ciofani G. Antioxidants and Nanotechnology: Promises and Limits of Potentially Disruptive Approaches in the Treatment of Central Nervous System Diseases. Adv Healthc Mater 2020; 9:e1901589. [PMID: 31854132 DOI: 10.1002/adhm.201901589] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 11/26/2019] [Indexed: 12/11/2022]
Abstract
Many central nervous system (CNS) diseases are still incurable and only symptomatic treatments are available. Oxidative stress is suggested to be a common hallmark, being able to cause and exacerbate the neuronal cell dysfunctions at the basis of these pathologies, such as mitochondrial impairments, accumulation of misfolded proteins, cell membrane damages, and apoptosis induction. Several antioxidant compounds are tested as potential countermeasures for CNS disorders, but their efficacy is often hindered by the loss of antioxidant properties due to enzymatic degradation, low bioavailability, poor water solubility, and insufficient blood-brain barrier crossing efficiency. To overcome the limitations of antioxidant molecules, exploitation of nanostructures, either for their delivery or with inherent antioxidant properties, is proposed. In this review, after a brief discussion concerning the role of the blood-brain barrier in the CNS and the involvement of oxidative stress in some neurodegenerative diseases, the most interesting research concerning the use of nano-antioxidants is introduced and discussed, focusing on the synthesis procedures, functionalization strategies, in vitro and in vivo tests, and on recent clinical trials.
Collapse
Affiliation(s)
- Chiara Martinelli
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy
| | - Carlotta Pucci
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy
| | - Matteo Battaglini
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy
- Scuola Superiore Sant'Anna, The Biorobotics Institute, Viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy
| | - Attilio Marino
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy
| | - Gianni Ciofani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy
| |
Collapse
|
18
|
Paul BD, Snyder SH. Impaired Redox Signaling in Huntington's Disease: Therapeutic Implications. Front Mol Neurosci 2019; 12:68. [PMID: 30941013 PMCID: PMC6433839 DOI: 10.3389/fnmol.2019.00068] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/04/2019] [Indexed: 12/22/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disease triggered by expansion of polyglutamine repeats in the protein huntingtin. Mutant huntingtin (mHtt) aggregates and elicits toxicity by multiple mechanisms which range from dysregulated transcription to disturbances in several metabolic pathways in both the brain and peripheral tissues. Hallmarks of HD include elevated oxidative stress and imbalanced redox signaling. Disruption of antioxidant defense mechanisms, involving antioxidant molecules and enzymes involved in scavenging or reversing oxidative damage, have been linked to the pathophysiology of HD. In addition, mitochondrial function is compromised in HD leading to impaired bioenergetics and elevated production of free radicals in cells. However, the exact mechanisms linking redox imbalance to neurodegeneration are still elusive. This review will focus on the current understanding of aberrant redox homeostasis in HD and potential therapeutic interventions.
Collapse
Affiliation(s)
- Bindu D. Paul
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Solomon H. Snyder
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
19
|
Essa MM, Moghadas M, Ba-Omar T, Walid Qoronfleh M, Guillemin GJ, Manivasagam T, Justin-Thenmozhi A, Ray B, Bhat A, Chidambaram SB, Fernandes AJ, Song BJ, Akbar M. Protective Effects of Antioxidants in Huntington’s Disease: an Extensive Review. Neurotox Res 2019; 35:739-774. [DOI: 10.1007/s12640-018-9989-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 12/09/2018] [Accepted: 12/11/2018] [Indexed: 01/18/2023]
|
20
|
Chodari L, Smailnejad S, Fallahi M, Khalaji N, Ghorbanzadeh V. OXIDATIVE STRESS IS MARKEDLY REDUCED BY COMBINED VOLUNTARY EXERCISE AND TESTOSTERONE IN THE HEART OF DIABETIC RATS. ACTA ENDOCRINOLOGICA-BUCHAREST 2019; 15:173-181. [PMID: 31508173 DOI: 10.4183/aeb.2019.173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Objective Cardiovascular disorders in diabetes condition arise from increased oxidative stress. Both regular mild exercise and testosterone influence on body's antioxidant system in diabetes. In this study, we evaluated treatment of testosterone and voluntary exercise, alone or together on oxidative stress in the heart and blood of diabetic rats. Methods Type 1 diabetes was induced by intraperitoneal injection of 50 mg/kg of streptozotocin in rats. Sixty three rats have been divided into eight groups as follows: Diabetes, diabetes+ testosterone, diabetes+ exercise, diabetes+ testosterone+ exercise, diabetes+ castration, diabetes+ castration+ testosterone, Diabetes+ castration+ exercise, Diabetes+ castration+ exercise+ testosterone. Type 1 diabetes was induced by intraperitoneal injection of 50 mg/kg of streptozotocin in the male Wistar rats and after a week, castration was performed. After 42 days of treatment with testosterone (2 mg/kg/day) or voluntary exercise alone or in combination, SOD, GPX and CAT activities and MDA levels were measured in the blood and heart tissue samples in the groups of study. In the end of study, SOD, GPX and CAT activities and MDA levels were measured in blood and heart tissue samples in the groups of study. Results SOD, GPX and CAT activities significantly (p<0.05) increased in groups that treated either testosterone or exercise and MDA level significantly (p<0.01) decreased in the blood and heart tissue of diabetic and castrated diabetic rats. Simultaneously, treatment with testosterone and exercise had a synergistic effect on antioxidant enzymes level in diabetic and diabetic castrated rats. In the castrated animals with diabetes, SOD, GPX and CAT activities significantly decreased (p<0.05) and MDA levels significantly increased (p<0.05) in blood and heart tissue. Conclusion Voluntary exercise and testosterone alone or together heightened body's antioxidant system and were able to reduce the MDA levels in blood and heart of diabetic and castrated diabetic rats.
Collapse
Affiliation(s)
- L Chodari
- Department of Physiology, Urmia University of Medical Sciences, Urmia, Iran.,Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - S Smailnejad
- Department of Physiology, Urmia University of Medical Sciences, Urmia, Iran
| | - M Fallahi
- Drug Applied Research Center of Tabriz, Urmia, Iran
| | - N Khalaji
- Department of Physiology, Urmia University of Medical Sciences, Urmia, Iran
| | - V Ghorbanzadeh
- Drug Applied Research Center of Tabriz, Urmia, Iran.,Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
21
|
Golomb BA. Diplomats' Mystery Illness and Pulsed Radiofrequency/Microwave Radiation. Neural Comput 2018; 30:2882-2985. [PMID: 30183509 DOI: 10.1162/neco_a_01133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Importance: A mystery illness striking U.S. and Canadian diplomats to Cuba (and now China) "has confounded the FBI, the State Department and US intelligence agencies" (Lederman, Weissenstein, & Lee, 2017). Sonic explanations for the so-called health attacks have long dominated media reports, propelled by peculiar sounds heard and auditory symptoms experienced. Sonic mediation was justly rejected by experts. We assessed whether pulsed radiofrequency/microwave radiation (RF/MW) exposure can accommodate reported facts in diplomats, including unusual ones. Observations: (1) Noises: Many diplomats heard chirping, ringing or grinding noises at night during episodes reportedly triggering health problems. Some reported that noises were localized with laser-like precision or said the sounds seemed to follow them (within the territory in which they were perceived). Pulsed RF/MW engenders just these apparent "sounds" via the Frey effect. Perceived "sounds" differ by head dimensions and pulse characteristics and can be perceived as located behind in or above the head. Ability to hear the "sounds" depends on high-frequency hearing and low ambient noise. (2) Signs/symptoms: Hearing loss and tinnitus are prominent in affected diplomats and in RF/MW-affected individuals. Each of the protean symptoms that diplomats report also affect persons reporting symptoms from RF/MW: sleep problems, headaches, and cognitive problems dominate in both groups. Sensations of pressure or vibration figure in each. Both encompass vision, balance, and speech problems and nosebleeds. Brain injury and brain swelling are reported in both. (3) Mechanisms: Oxidative stress provides a documented mechanism of RF/MW injury compatible with reported signs and symptoms; sequelae of endothelial dysfunction (yielding blood flow compromise), membrane damage, blood-brain barrier disruption, mitochondrial injury, apoptosis, and autoimmune triggering afford downstream mechanisms, of varying persistence, that merit investigation. (4) Of note, microwaving of the U.S. embassy in Moscow is historically documented. Conclusions and relevance: Reported facts appear consistent with pulsed RF/MW as the source of injury in affected diplomats. Nondiplomats citing symptoms from RF/MW, often with an inciting pulsed-RF/MW exposure, report compatible health conditions. Under the RF/MW hypothesis, lessons learned for diplomats and for RF/MW-affected civilians may each aid the other.
Collapse
|
22
|
Kotlar I, Colonnello A, Aguilera-González MF, Avila DS, de Lima ME, García-Contreras R, Ortíz-Plata A, Soares FAA, Aschner M, Santamaría A. Comparison of the Toxic Effects of Quinolinic Acid and 3-Nitropropionic Acid in C. elegans: Involvement of the SKN-1 Pathway. Neurotox Res 2018; 33:259-267. [PMID: 28822104 DOI: 10.1007/s12640-017-9794-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/01/2017] [Accepted: 08/03/2017] [Indexed: 12/23/2022]
Abstract
The tryptophan metabolite, quinolinic acid (QUIN), and the mitochondrial toxin 3-nitropropionic acid (3-NP) are two important tools for toxicological research commonly used in neurotoxic models of excitotoxicity, oxidative stress, energy depletion, and neuronal cell death in mammals. However, their toxic properties have yet to be explored in the nematode Caenorhabditis elegans (C. elegans) for the establishment of novel, simpler, complementary, alternative, and predictive neurotoxic model of mammalian neurotoxicity. In this work, the effects of QUIN (1-100 mM) and 3-NP (1-10 mM) were evaluated on various physiological parameters (survival, locomotion, and longevity) in a wild-type (WT) strand of C. elegans (N2). Their effects were also tested in the VC1772 strain (knock out for the antioxidant SKN-1 pathway) and the VP596 strain (worms with a reporter gene for glutathione S-transferase (GST) transcription) in order to establish the role of the SKN-1 pathway in the mode of action of QUIN and 3-NP. In N2, the higher doses of both toxins decreased survival, though only QUIN altered motor activity. Both toxins also reduced longevity in the VC1772 strain (as compared to N2 strain) and augmented GST transcription in the VP596 strain at the highest doses. The changes induced by both toxins require high doses, and therefore appear moderate when compared with other toxic agents. Nevertheless, the alterations produced by QUIN and 3-NP in C. elegans are relevant to mammalian neurotoxicity as they provide novel mechanistic approaches to the assessment of neurotoxic events comprising oxidative stress and excitotoxicity, in the nematode model.
Collapse
Affiliation(s)
- Ilan Kotlar
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Insurgentes Sur 3877, 14269, Ciudad de México, Mexico
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Aline Colonnello
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Insurgentes Sur 3877, 14269, Ciudad de México, Mexico
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - María Fernanda Aguilera-González
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Insurgentes Sur 3877, 14269, Ciudad de México, Mexico
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | | | - María Eduarda de Lima
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Insurgentes Sur 3877, 14269, Ciudad de México, Mexico
- Universidade Federal do Pampa, Uruguaiana, RS, Brazil
| | - Rodolfo García-Contreras
- Laboratorio de Bacteriología, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Alma Ortíz-Plata
- Laboratorio de Patología Experimental, Instituto Nacional de Neurología y Neurocirugía, 14269, Mexico City, Mexico
| | | | - Michael Aschner
- Albert Einstein College of Medicine, Jack and Pearl Resnick Campus, Bronx, NY, 10461, USA
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Insurgentes Sur 3877, 14269, Ciudad de México, Mexico.
| |
Collapse
|
23
|
Wongprayoon P, Govitrapong P. Melatonin as a mitochondrial protector in neurodegenerative diseases. Cell Mol Life Sci 2017; 74:3999-4014. [PMID: 28791420 PMCID: PMC11107580 DOI: 10.1007/s00018-017-2614-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/03/2017] [Indexed: 12/19/2022]
Abstract
Mitochondria are crucial organelles as their role in cellular energy production of eukaryotes. Because the brain cells demand high energy for maintaining their normal activities, disturbances in mitochondrial physiology may lead to neuropathological events underlying neurodegenerative conditions such as Alzheimer's disease, Parkinson's disease and Huntington's disease. Melatonin is an endogenous compound with a variety of physiological roles. In addition, it possesses potent antioxidant properties which effectively play protective roles in several pathological conditions. Several lines of evidence also reveal roles of melatonin in mitochondrial protection, which could prevent development and progression of neurodegeneration. Since the mitochondrial dysfunction is a primary event in neurodegeneration, the neuroprotection afforded by melatonin is thereby more effective in early stages of the diseases. This article reviews mechanisms which melatonin exerts its protective roles on mitochondria as a potential therapeutic strategy against neurodegenerative disorders.
Collapse
Affiliation(s)
- Pawaris Wongprayoon
- Department of Biopharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand
| | - Piyarat Govitrapong
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, 73170, Thailand.
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, 10210, Thailand.
| |
Collapse
|
24
|
Wu HJ, Wu C, Niu HJ, Wang K, Mo LJ, Shao AW, Dixon BJ, Zhang JM, Yang SX, Wang YR. Neuroprotective Mechanisms of Melatonin in Hemorrhagic Stroke. Cell Mol Neurobiol 2017; 37:1173-1185. [PMID: 28132129 DOI: 10.1007/s10571-017-0461-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/05/2017] [Indexed: 12/30/2022]
Abstract
Hemorrhagic stroke which consists of subarachnoid hemorrhage and intracerebral hemorrhage is a dominant cause of death and disability worldwide. Although great efforts have been made, the physiological mechanisms of these diseases are not fully understood and effective pharmacological interventions are still lacking. Melatonin (N-acetyl-5-methoxytryptamine), a neurohormone produced by the pineal gland, is a broad-spectrum antioxidant and potent free radical scavenger. More importantly, there is extensive evidence demonstrating that melatonin confers neuroprotective effects in experimental models of hemorrhagic stroke. Multiple molecular mechanisms such as antioxidant, anti-apoptosis, and anti-inflammation, contribute to melatonin-mediated neuroprotection against brain injury after hemorrhagic stroke. This review article aims to summarize current knowledge regarding the beneficial effects of melatonin in experimental models of hemorrhagic stroke and explores the underlying mechanisms. We propose that melatonin is a promising neuroprotective candidate that is worthy of further evaluation for its potential therapeutic applications in hemorrhagic stroke.
Collapse
Affiliation(s)
- Hai-Jian Wu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Cheng Wu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Huan-Jiang Niu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Kun Wang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Lian-Jie Mo
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - An-Wen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Brandon J Dixon
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jian-Min Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shu-Xu Yang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
| | - Yi-Rong Wang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
25
|
Wahdan SA, Tadros MG, Khalifa AE. Antioxidant and antiapoptotic actions of selegiline protect against 3-NP-induced neurotoxicity in rats. Naunyn Schmiedebergs Arch Pharmacol 2017. [DOI: 10.1007/s00210-017-1392-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
26
|
The Role of Reactive Oxygen Species in the Pathogenesis of Alzheimer's Disease, Parkinson's Disease, and Huntington's Disease: A Mini Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8590578. [PMID: 28116038 PMCID: PMC5223034 DOI: 10.1155/2016/8590578] [Citation(s) in RCA: 302] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/06/2016] [Accepted: 11/13/2016] [Indexed: 11/18/2022]
Abstract
Neurodegenerative diseases affect not only the life quality of aging populations, but also their life spans. All forms of neurodegenerative diseases have a massive impact on the elderly. The major threat of these brain diseases includes progressive loss of memory, Alzheimer's disease (AD), impairments in the movement, Parkinson's disease (PD), and the inability to walk, talk, and think, Huntington's disease (HD). Oxidative stress and mitochondrial dysfunction are highlighted as a central feature of brain degenerative diseases. Oxidative stress, a condition that occurs due to imbalance in oxidant and antioxidant status, has been known to play a vital role in the pathophysiology of neurodegenerative diseases including AD, PD, and HD. A large number of studies have utilized oxidative stress biomarkers to investigate the severity of these neurodegenerative diseases and medications are available, but these only treat the symptoms. In traditional medicine, a large number of medicinal plants have been used to treat the symptoms of these neurodegenerative diseases. Extensive studies scientifically validated the beneficial effect of natural products against neurodegenerative diseases using suitable animal models. This short review focuses the role of oxidative stress in the pathogenesis of AD, PD, and HD and the protective efficacy of natural products against these diseases.
Collapse
|
27
|
A novel therapeutic application of solid lipid nanoparticles encapsulated thymoquinone (TQ-SLNs) on 3-nitroproponic acid induced Huntington's disease-like symptoms in wistar rats. Chem Biol Interact 2016; 256:25-36. [PMID: 27206696 DOI: 10.1016/j.cbi.2016.05.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/20/2016] [Accepted: 05/15/2016] [Indexed: 01/02/2023]
Abstract
Huntington's disease (HD), a devastating neurodegenerative disease causing a remarkable pathogenesis involves mitochondrial dysfunction and bioenergetics failure. 3-Nitropropionic acid (3-NP) is a unique toxin model of HD that are mainly confined to mitochondrial complex-II inhibition and free radical generation. Recently, several nanoparticle formulations were developed to treat against various neurodegenerative diseases including HD. One among them is solid lipid nanoparticles (SLNs), a colloidal carrier designed to enhance the brain drug delivery and to prolong the bio-availability of drugs in the system. Hence, the present study was framed to evaluate solid lipid nanoparticles encapsulated thymoquinone (TQ-SLNs) in comparison with thymoquinone suspension (TQ-S) against 3-NP induced behavioral despair, oxidative injury and striatal pathology. This study reports that theTQ-SLNs (10 and 20 mg/kg) and TQ-S (80 mg/kg) treated animals showed a significant (P < 0.01) improvement in the muscle strength, rigidity, movement and memory performances on 7th and 14th day behavioral analysis than TQ-S (40 mg/kg) treated group. Similarly, TQ-SLNs highly attenuated the levels of oxidative stress markers such as LPO, NO and protein carbonylsin 3-NP induced animals. Further, TQ-SLNs significantly restored the antioxidant defense system, controls the mitochondrial SDH inhibition and alleviates anti-cholinergic effect upon 3-NP induction. In addition, TQ-SLNs efficiently protected the striatal structural microelements against 3-NP toxicity, which was confirmed by light microscopic studies. Thus, the present investigation, collectively suggests that the low dose of TQ-SLNs supplementation is highly sufficient to attain the effect of TQ-S (80 mg/kg) to attenuate behavioral, biochemical and histological modifications in 3-NP exposed HD model.
Collapse
|
28
|
Shinomol GK, Ranganayaki S, Joshi AK, Gayathri N, Gowda H, Muralidhara, Srinivas Bharath MM. Characterization of age-dependent changes in the striatum: Response to the mitochondrial toxin 3-nitropropionic acid. Mech Ageing Dev 2016; 161:66-82. [PMID: 27143313 DOI: 10.1016/j.mad.2016.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 04/10/2016] [Accepted: 04/23/2016] [Indexed: 11/26/2022]
Abstract
Neurodegenerative phenomena are associated with mitochondrial dysfunction and this could be exacerbated by aging. Age-dependence of mitochondrial response to toxins could help understand these mechanisms and evolve novel therapeutics. 3-Nitropropionic acid (3-NPA) is a mitochondrial toxin that induces neurotoxicity in the striatum via inhibition of complex II. We investigated the age-related events that contribute to 3-NPA toxicity. 3-NPA induced neuronal death, oxidative stress and altered mitochondrial structure in neuronal cells. 3-NPA injection in vivo caused motor impairment, mitochondrial dysfunction and oxidative damage with different trend in young and adult mice. To understand the age-dependent mechanisms, we carried out proteomic analysis of the striatal protein extract from young mice (control: YC vs. 3-NPA treated: YT) and adult mice (control: AC vs. 3-NPA treated: AT). Among the 3752 identified proteins, 33 differentially expressed proteins (mitochondrial, synaptic and microsomal proteins) were unique either to YT or AT. Interestingly, comparison of the proteomic profile in AC and YC indicated that 161 proteins (linked with cytoskeletal structure, neuronal development, axogenesis, protein transport, cell adhesion and synaptic function) were down-regulated in AC compared to YC. We surmise that aging contributes to the cellular and molecular architecture in the mouse striatum with implications for neurodegeneration.
Collapse
Affiliation(s)
- G K Shinomol
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India; Neurotoxicology laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India
| | - S Ranganayaki
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India; Neurotoxicology laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India
| | - Apurva K Joshi
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India; Neurotoxicology laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India
| | - N Gayathri
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India
| | - Harsha Gowda
- Institute of Bioinformatics (IOB), Discoverer, Industrial Technology Park Limited (ITPL), Whitefield, Bangalore 560066, Karnataka, India
| | - Muralidhara
- Department of Biochemistry and Nutrition, Central Food Technological Research Institute, Mysore 570020, Karnataka, India
| | - M M Srinivas Bharath
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India; Neurotoxicology laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India.
| |
Collapse
|
29
|
Ganie SA, Dar TA, Bhat AH, Dar KB, Anees S, Zargar MA, Masood A. Melatonin: A Potential Anti-Oxidant Therapeutic Agent for Mitochondrial Dysfunctions and Related Disorders. Rejuvenation Res 2015; 19:21-40. [PMID: 26087000 DOI: 10.1089/rej.2015.1704] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Mitochondria play a central role in cellular physiology. Besides their classic function of energy metabolism, mitochondria are involved in multiple cell functions, including energy distribution through the cell, energy/heat modulation, regulation of reactive oxygen species (ROS), calcium homeostasis, and control of apoptosis. Simultaneously, mitochondria are the main producer and target of ROS with the result that multiple mitochondrial diseases are related to ROS-induced mitochondrial injuries. Increased free radical generation, enhanced mitochondrial inducible nitric oxide synthase (iNOS) activity, enhanced nitric oxide (NO) production, decreased respiratory complex activity, impaired electron transport system, and opening of mitochondrial permeability transition pores have all been suggested as factors responsible for impaired mitochondrial function. Because of these, neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and aging, are caused by ROS-induced mitochondrial dysfunctions. Melatonin, the major hormone of the pineal gland, also acts as an anti-oxidant and as a regulator of mitochondrial bioenergetic function. Melatonin is selectively taken up by mitochondrial membranes, a function not shared by other anti-oxidants, and thus has emerged as a major potential therapeutic tool for treating neurodegenerative disorders. Multiple in vitro and in vivo experiments have shown the protective role of melatonin for preventing oxidative stress-induced mitochondrial dysfunction seen in experimental models of PD, AD, and HD. With these functions in mind, this article reviews the protective role of melatonin with mechanistic insights against mitochondrial diseases and suggests new avenues for safe and effective treatment modalities against these devastating neurodegenerative diseases. Future insights are also discussed.
Collapse
Affiliation(s)
- Showkat Ahmad Ganie
- 1 Department of Clinical Biochemistry, University of Kashmir Srinagar , India
| | - Tanveer Ali Dar
- 1 Department of Clinical Biochemistry, University of Kashmir Srinagar , India
| | - Aashiq Hussain Bhat
- 1 Department of Clinical Biochemistry, University of Kashmir Srinagar , India
| | - Khalid B Dar
- 1 Department of Clinical Biochemistry, University of Kashmir Srinagar , India
| | - Suhail Anees
- 1 Department of Clinical Biochemistry, University of Kashmir Srinagar , India
| | | | - Akbar Masood
- 2 Department of Biochemistry, University of Kashmir Srinagar , India
| |
Collapse
|
30
|
Hanna DMF, Tadros MG, Khalifa AE. ADIOL protects against 3-NP-induced neurotoxicity in rats: Possible impact of its anti-oxidant, anti-inflammatory and anti-apoptotic actions. Prog Neuropsychopharmacol Biol Psychiatry 2015; 60:36-51. [PMID: 25689821 DOI: 10.1016/j.pnpbp.2015.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 02/08/2015] [Accepted: 02/09/2015] [Indexed: 01/20/2023]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder with a wide spectrum of cognitive, behavioral and motor abnormalities. The mitochondrial toxin 3-nitropropionic acid (3-NP) effectively induces specific behavioral changes and selective striatal lesions similar to that observed in HD. Some neurosteroids, synthesized in neurons and glial cells, previously showed neuroprotective abilities. 5-Androstene-3β-17β-diol (ADIOL) is a major metabolite of dehydroepiandrosterone (DHEA) with previously reported anti-inflammatory, anti-apoptotic and neuroprotective activities. The neuroprotective potential of ADIOL in HD was not previously investigated. Therefore, the present study investigated the neuroprotective effects of ADIOL against 3-NP-induced behavioral changes, oxidative stress, inflammation and apoptosis. Intraperitoneal administration of 3-NP (20mg/kg) for 4 consecutive days in rats caused significant loss in body weight, reduced prepulse inhibition (PPI) of acoustic startle response, locomotor hypoactivity with altered cortical/striatal histological structure, increased cortical/striatal oxidative stress, inflammation and apoptosis. Administration of ADIOL (25mg/kg, s.c.) for two days before 3-NP significantly attenuated the reduction in body weights and PPI, increased locomotor activity and restored cortical/striatal histological structure nearly to normal. Moreover, it displayed anti-oxidant, anti-inflammatory and anti-apoptotic activities as evidenced by the elevation of cortical and striatal reduced glutathione levels, reductions of cortical and striatal malondialdehyde, striatal tumor necrosis factor alpha and interleukin-6 levels. Only a small number of iNOS and caspase-3 positive cells were detected in sections from rats pretreated with ADIOL. This study suggests a potential neuroprotective role of ADIOL against 3-NP-induced Huntington's disease-like manifestations. Such neuroprotection can be attributed to its anti-oxidant, anti-inflammatory and anti-apoptotic activities.
Collapse
Affiliation(s)
- Diana M F Hanna
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mariane G Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Amani E Khalifa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
31
|
Neuroprotective therapeutics from botanicals and phytochemicals against Huntington's disease and related neurodegenerative disorders. J Herb Med 2015. [DOI: 10.1016/j.hermed.2015.01.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
32
|
Joshi N, Biswas J, Nath C, Singh S. Promising Role of Melatonin as Neuroprotectant in Neurodegenerative Pathology. Mol Neurobiol 2014; 52:330-40. [DOI: 10.1007/s12035-014-8865-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 08/14/2014] [Indexed: 12/14/2022]
|
33
|
Chakraborty J, Nthenge-Ngumbau D, Rajamma U, Mohanakumar K. Melatonin protects against behavioural dysfunctions and dendritic spine damage in 3-nitropropionic acid-induced rat model of Huntington's disease. Behav Brain Res 2014; 264:91-104. [DOI: 10.1016/j.bbr.2014.01.048] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 01/26/2014] [Indexed: 12/15/2022]
|
34
|
Singh M, Jadhav HR. Melatonin: functions and ligands. Drug Discov Today 2014; 19:1410-8. [PMID: 24792719 DOI: 10.1016/j.drudis.2014.04.014] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 03/16/2014] [Accepted: 04/22/2014] [Indexed: 12/15/2022]
Abstract
Melatonin is a chronobiotic substance that acts as synchronizer by stabilizing bodily rhythms. Its synthesis occurs in various locations throughout the body, including the pineal gland, skin, lymphocytes and gastrointestinal tract (GIT). Its synthesis and secretion is controlled by light and dark conditions, whereby light decreases and darkness increases its production. Thus, melatonin is also known as the 'hormone of darkness'. Melatonin and analogs that bind to the melatonin receptors are important because of their role in the management of depression, insomnia, epilepsy, Alzheimer's disease (AD), diabetes, obesity, alopecia, migraine, cancer, and immune and cardiac disorders. In this review, we discuss the mechanism of action of melatonin in these disorders, which could aid in the design of novel melatonin receptor ligands.
Collapse
Affiliation(s)
- Mahaveer Singh
- Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Pilani Campus, Vidya Vihar, Pilani 333031, Rajasthan, India.
| | - Hemant R Jadhav
- Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Pilani Campus, Vidya Vihar, Pilani 333031, Rajasthan, India
| |
Collapse
|
35
|
Zhang JQ, Shen M, Zhu CC, Yu FX, Liu ZQ, Ally N, Sun SC, Li K, Liu HL. 3-Nitropropionic acid induces ovarian oxidative stress and impairs follicle in mouse. PLoS One 2014; 9:e86589. [PMID: 24505260 PMCID: PMC3914797 DOI: 10.1371/journal.pone.0086589] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 12/12/2013] [Indexed: 12/19/2022] Open
Abstract
Oxidative stress induces many serious reproductive diseases in female mammals and thus poses a serious threat to reproductive health. However, the relationship between reactive oxygen species (ROS)-induced oxidative stress and follicular development, oocyte and embryo quality is not clear. The aim of this study was to investigate the effect of ovarian oxidative stress on the health of follicle and oocyte development. Female ICR mice were dosed with 3-nitropropionic acid (3-NPA) at three different concentrations (6.25, 12.5 and 25 mg/kg) and saline (control) via continuous intraperitoneal injection for 7 days. The treatment with 12.5 mg/kg reduced the weight of mouse ovaries, and significantly increased ROS levels and the activities of antioxidant enzymes--total superoxide dismutase (T-SOD), glutathione peroxidase (GPx) and catalase (CAT)--in granulosa cells and ovarian tissues, but not in other tissues (brain, liver, kidney and spleen). The same treatment significantly increased the percentage of atretic large follicles, and reduced the number of large follicles, the number of ovulated oocytes, and the capacity for early embryonic development compared with controls. It also significantly decreased the ratio of Bcl-2 to Bax, while causing an increase in the mRNA expression of (SOD2, CAT and GP X) and ROS levels in granulosa cells. Collectively, these data indicate that 3-NPA induces granulosa cell apoptosis, large follicle atresia, and an increase of ROS levels in the ovary. Therefore, we have established an in vivo model of ovarian oxidative stress for studying the mechanism of resulting damage induced by free radicals and for the screening of novel antioxidants.
Collapse
Affiliation(s)
- Jia-Qing Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Ming Shen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Cheng-Cheng Zhu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Feng-Xiang Yu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Ze-Qun Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Nazim Ally
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Shao-Chen Sun
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Kui Li
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hong-Lin Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
36
|
Chakraborty J, Rajamma U, Mohanakumar KP. A mitochondrial basis for Huntington's disease: therapeutic prospects. Mol Cell Biochem 2013; 389:277-91. [PMID: 24374792 DOI: 10.1007/s11010-013-1951-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Accepted: 12/19/2013] [Indexed: 01/12/2023]
Abstract
Huntington's disease (HD) is an autosomal dominant disease, with overt movement dysfunctions. Despite focused research on the basis of neurodegeneration in HD for last few decades, the mechanism for the site-specific lesion of neurons in the brain is not clear. All the explanations that partially clarify the phenomenon of neurodegeneration leads to one organelle, mitochondrion, which is severely affected in HD at the level of electron transport chain, Ca(2+) buffering efficiency and morphology. But, with the existing knowledge, it is not clear whether the cell death processes in HD initiate from mitochondria, though the Huntingtin (Htt) aggregates show close proximity to this organelle, or do some extracellular stimuli like TNFα or FasL trigger the process. Mainly because of the disparity in the different available experimental models, the results are quite confusing or at least inconsistent to a great extent. The fact remains that the mutant Htt protein was seen to be associated with mitochondria directly, and as the striatum is highly enriched with dopamine and glutamate, it may make the striatal mitochondria more vulnerable because of the presence of dopa-quinones, and due to an imbalance in Ca(2+). The current therapeutic strategies are based on symptomatic relief, and, therefore, mainly target neurotransmitter(s) and their receptors to modulate behavioral outputs, but none of them targets mitochondria or try to address the basic molecular events that cause neurons to die in discrete regions of the brain, which could probably be resulting from grave mitochondrial dysfunctions. Therefore, targeting mitochondria for their protection, while addressing symptomatic recovery, holds a great potential to tone down the progression of the disease, and to provide better relief to the patients and caretakers.
Collapse
Affiliation(s)
- J Chakraborty
- Laboratory of Clinical and Experimental Neuroscience, Division of Cell Biology & Physiology, CSIR-Indian Institute of Chemical Biology, Rooms 117&119, 4, Raja S. C. Mullick Road, Kolkata, 700 032, India
| | | | | |
Collapse
|
37
|
Sandhir R, Yadav A, Mehrotra A, Sunkaria A, Singh A, Sharma S. Curcumin nanoparticles attenuate neurochemical and neurobehavioral deficits in experimental model of Huntington's disease. Neuromolecular Med 2013; 16:106-18. [PMID: 24008671 DOI: 10.1007/s12017-013-8261-y] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 08/16/2013] [Indexed: 12/13/2022]
Abstract
Till date, an exact causative pathway responsible for neurodegeneration in Huntington's disease (HD) remains elusive; however, mitochondrial dysfunction appears to play an important role in HD pathogenesis. Therefore, strategies to attenuate mitochondrial impairments could provide a potential therapeutic intervention. In the present study, we used curcumin encapsulated solid lipid nanoparticles (C-SLNs) to ameliorate 3-nitropropionic acid (3-NP)-induced HD in rats. Results of MTT (3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide) assay and succinate dehydrogenase (SDH) staining of striatum revealed a marked decrease in Complex II activity. However, C-SLN-treated animals showed significant increase in the activity of mitochondrial complexes and cytochrome levels. C-SLNs also restored the glutathione levels and superoxide dismutase activity. Moreover, significant reduction in mitochondrial swelling, lipid peroxidation, protein carbonyls and reactive oxygen species was observed in rats treated with C-SLNs. Quantitative PCR and Western blot results revealed the activation of nuclear factor-erythroid 2 antioxidant pathway after C-SLNs administration in 3-NP-treated animals. In addition, C-SLN-treated rats showed significant improvement in neuromotor coordination when compared with 3-NP-treated rats. Thus, the results of this study suggest that C-SLNs administration might be a promising therapeutic intervention to ameliorate mitochondrial dysfunctions in HD.
Collapse
Affiliation(s)
- Rajat Sandhir
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India,
| | | | | | | | | | | |
Collapse
|
38
|
Tasset I, Pontes AJ, Hinojosa AJ, de la Torre R, Túnez I. Olive oil reduces oxidative damage in a 3-nitropropionic acid-induced Huntington's disease-like rat model. Nutr Neurosci 2013; 14:106-11. [DOI: 10.1179/1476830511y.0000000005] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
39
|
Kasote DM, Hegde MV, Katyare SS. Mitochondrial dysfunction in psychiatric and neurological diseases: cause(s), consequence(s), and implications of antioxidant therapy. Biofactors 2013; 39:392-406. [PMID: 23460132 DOI: 10.1002/biof.1093] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 12/25/2012] [Indexed: 12/12/2022]
Abstract
Mitochondrial dysfunction is at the base of development and progression of several psychiatric and neurologic diseases with different etiologies. MtDNA/nDNA mutational damage, failure of endogenous antioxidant defenses, hormonal malfunction, altered membrane permeability, metabolic dysregulation, disruption of calcium buffering capacity and ageing have been found to be the root causes of mitochondrial dysfunction in psychatric and neurodegenerative diseases. However, the overall consequences of mitochondrial dysfunction are only limited to increase in oxidative/nitrosative stress and cellular energy crises. Thus far, extensive efforts have been made to improve mitochondrial function through specific cause-dependent antioxidant therapy. However, owing to complex genetic and interlinked causes of mitochondrial dysfunction, it has not been possible to achieve any common, unique supportive antioxidant therapeutic strategy for the treatment of psychiatric and neurologic diseases. Hence, we propose an antioxidant therapeutic strategy for management of consequences of mitochondrial dysfunction in psychiatric and neurologic diseases. It is expected that this will not only reduces oxidative stress, but also promote anaerobic energy production.
Collapse
Affiliation(s)
- Deepak M Kasote
- MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune, MS, India.
| | | | | |
Collapse
|
40
|
Kovacic P, Somanathan R. Redox processes in neurodegenerative disease involving reactive oxygen species. Curr Neuropharmacol 2013; 10:289-302. [PMID: 23730253 PMCID: PMC3520039 DOI: 10.2174/157015912804143487] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 06/12/2012] [Accepted: 06/20/2012] [Indexed: 11/22/2022] Open
Abstract
Much attention has been devoted to neurodegenerative diseases involving redox processes. This review comprises an update involving redox processes reported in the considerable literature in recent years. The mechanism involves reactive oxygen species and oxidative stress, usually in the brain. There are many examples including Parkinson’s, Huntington’s, Alzheimer’s, prions, Down’s syndrome, ataxia, multiple sclerosis, Creutzfeldt-Jacob disease, amyotrophic lateral sclerosis, schizophrenia, and Tardive Dyskinesia. Evidence indicates a protective role for antioxidants, which may have clinical implications. A multifaceted approach to mode of action appears reasonable.
Collapse
Affiliation(s)
- Peter Kovacic
- Department of Chemistry and Biochemistry, San Diego State University, San Diego CA 92182 USA
| | | |
Collapse
|
41
|
Santhosh MS, Sundaram MS, Sunitha K, Kemparaju K, Girish KS. Viper venom-induced oxidative stress and activation of inflammatory cytokines: a therapeutic approach for overlooked issues of snakebite management. Inflamm Res 2013; 62:721-31. [DOI: 10.1007/s00011-013-0627-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 01/02/2013] [Accepted: 04/23/2013] [Indexed: 11/24/2022] Open
|
42
|
Cardinali DP, Pagano ES, Scacchi Bernasconi PA, Reynoso R, Scacchi P. Melatonin and mitochondrial dysfunction in the central nervous system. Horm Behav 2013; 63:322-30. [PMID: 22391273 DOI: 10.1016/j.yhbeh.2012.02.020] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 02/16/2012] [Accepted: 02/20/2012] [Indexed: 01/19/2023]
Abstract
Cell death and survival are critical events for neurodegeneration, mitochondria being increasingly seen as important determinants of both. Mitochondrial dysfunction is considered a major causative factor in Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD). Increased free radical generation, enhanced mitochondrial inducible nitric oxide (NO) synthase activity and NO production, and disrupted electron transport system and mitochondrial permeability transition, have all been involved in impaired mitochondrial function. Melatonin, the major secretory product of the pineal gland, is an antioxidant and an effective protector of mitochondrial bioenergetic function. Both in vitro and in vivo, melatonin was effective to prevent oxidative stress/nitrosative stress-induced mitochondrial dysfunction seen in experimental models of AD, PD and HD. These effects are seen at doses 2-3 orders of magnitude higher than those required to affect sleep and circadian rhythms, both conspicuous targets of melatonin action. Melatonin is selectively taken up by mitochondria, a function not shared by other antioxidants. A limited number of clinical studies indicate that melatonin can improve sleep and circadian rhythm disruption in PD and AD patients. More recently, attention has been focused on the development of potent melatonin analogs with prolonged effects which were employed in clinical trials in sleep-disturbed or depressed patients in doses considerably higher than those employed for melatonin. In view that the relative potencies of the analogs are higher than that of the natural compound, clinical trials employing melatonin in the range of 50-100mg/day are needed to assess its therapeutic validity in neurodegenerative disorders.
Collapse
Affiliation(s)
- Daniel P Cardinali
- Pontificia Universidad Católica Argentina, Facultad de Ciencias Médicas, 1107 Buenos Aires, Argentina.
| | | | | | | | | |
Collapse
|
43
|
Correlations between behavioural and oxidative parameters in a rat quinolinic acid model of Huntington's disease: protective effect of melatonin. Eur J Pharmacol 2013; 701:65-72. [PMID: 23340221 DOI: 10.1016/j.ejphar.2013.01.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 12/20/2012] [Accepted: 01/09/2013] [Indexed: 11/23/2022]
Abstract
The present study was designed to examine the correlations between behavioural and oxidative parameters in a quinolinic acid model of Huntington's disease in rats. The protective effect of melatonin against the excitotoxicity induced by quinolinic acid was investigated. Rats were pre-treated with melatonin (5 or 20mg/kg) before injection of quinolinic acid (240nmol/site; 1μl) into their right corpora striata. The locomotor and exploratory activities as well as the circling behaviour were recorded. The elevated body swing test was also performed. After behavioural experiments, biochemical determinations were carried out. Melatonin partially protected against the increase of circling behaviour caused by quinolinic acid injection. No alteration was found in the number of crossings and rearings of animals treated with melatonin and/or quinolinic acid. Melatonin decreased the percentage of contralateral biased swings induced by quinolinic acid. Melatonin protected against the increase in reactive species and protein carbonyl levels as well as the inhibition of superoxide dismutase activity resulting from quinolinic acid injection. Melatonin was partially effective against the inhibition of striatal catalase activity and a decrease of non-protein thiol levels induced by quinolinic acid. Melatonin was not effective against the inhibition of Na(+), K(+) ATPase activity caused by quinolinic acid injection. There were significant correlations between circling behaviour and oxidative parameters. The antioxidant property of melatonin is involved, at least in part, in its neuroprotective effect. The results reinforce the idea that melatonin could be useful in overwhelming neurotoxicity caused by quinolinic acid, a rat model of Huntington's disease.
Collapse
|
44
|
Nagpal K, Singh SK, Mishra DN. Formulation, Optimization, in Vivo Pharmacokinetic, Behavioral and Biochemical Estimations of Minocycline Loaded Chitosan Nanoparticles for Enhanced Brain Uptake. Chem Pharm Bull (Tokyo) 2013; 61:258-72. [DOI: 10.1248/cpb.c12-00732] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Kalpana Nagpal
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science & Technology
| | - Shailendra Kumar Singh
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science & Technology
| | - Dina Nath Mishra
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science & Technology
| |
Collapse
|
45
|
Jeong JK, Moon MH, Lee YJ, Seol JW, Park SY. Melatonin-induced autophagy protects against human prion protein-mediated neurotoxicity. J Pineal Res 2012; 53:138-46. [PMID: 22335252 DOI: 10.1111/j.1600-079x.2012.00980.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Melatonin has neuroprotective effects in the models of neurodegenerative disease including Alzheimer's and Parkinson's disease. Several studies have shown that melatonin prevents neurodegeneration by regulation of mitochondrial function. However, the protective action of melatonin has not been reported in prion disease. We investigated the influence of melatonin on prion-mediated neurotoxicity. Melatonin rescued neuronal cells from PrP(106-126)-induced neurotoxicity by prevention of mitochondrial dysfunction. Moreover, the protective effect of melatonin against mitochondrial dysfunction was related with autophagy activation. Melatonin-treated cells were dose-dependently increased in LC3-II, an autophagy marker. Melatonin-induced autophagy prevented a PrP(106-126)-induced reduction in mitochondrial potential and translocation of Bax to the mitochondria and cytochrome c release. On the other hand, downregulation of autophagy protein 5 with Atg5 siRNA or the autophagy blocker 3-methyladenine prevented the melatonin-mediated neuroprotective effects. This is the first report demonstrating that treatment with melatonin appears to protect against prion-mediated neurotoxicity and that the neuroprotection is induced by melatonin-mediated autophagy signals. The results of this study suggest that regulation of melatonin is a therapeutic strategy for prion peptide-induced apoptosis.
Collapse
Affiliation(s)
- Jae-Kyo Jeong
- Korea Zoonoses Research Institute, Bio-Safety Research Institute, Center for Healthcare Technology Development, College of Veterinary Medicine, Chonbuk National University, Jeonju, Korea
| | | | | | | | | |
Collapse
|
46
|
Melatonin Antioxidative Defense: Therapeutical Implications for Aging and Neurodegenerative Processes. Neurotox Res 2012; 23:267-300. [DOI: 10.1007/s12640-012-9337-4] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 06/12/2012] [Accepted: 06/13/2012] [Indexed: 12/12/2022]
|
47
|
Aziz NA, Pijl H, Frölich M, Schröder-van der Elst JP, van der Bent C, Roelfsema F, Roos RAC. Delayed onset of the diurnal melatonin rise in patients with Huntington's disease. J Neurol 2012; 256:1961-5. [PMID: 19562249 PMCID: PMC2780627 DOI: 10.1007/s00415-009-5196-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Accepted: 05/24/2009] [Indexed: 11/27/2022]
Abstract
Sleep disturbances are very prevalent in Huntington’s disease (HD) patients and can substantially impair their quality of life. Accumulating evidence suggests considerable dysfunction of the hypothalamic suprachiasmatic nucleus (SCN), the biological clock, in both HD patients and transgenic mouse models of the disease. As melatonin has a major role in the regulation of sleep and other cyclical bodily activities and its synthesis is directly regulated by the SCN, we postulated that disturbed SCN function is likely to give rise to abnormal melatonin secretion in HD. Therefore, we compared 24 h melatonin secretion profiles between early stage HD patients and age-, sex- and body mass index-matched controls. Although mean diurnal melatonin levels were not different between the two groups (p = 0.691), the timing of the evening rise in melatonin levels was significantly delayed by more than 01:30 h in HD patients (p = 0.048). Moreover, diurnal melatonin levels strongly correlated with both motor (r = −0.70, p = 0.036) and functional impairment (r = +0.78, p = 0.013). These findings suggest a delayed sleep phase syndrome-like circadian rhythm disorder in early stage HD patients and suggest that melatonin levels may progressively decline with advancing disease.
Collapse
Affiliation(s)
- N Ahmad Aziz
- Department of Neurology, K-05-Q 110, Leiden University Medical Center, P.O. Box 9600, Albinusdreef 2, 2300 RC Leiden, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
48
|
Bhateja DK, Dhull DK, Gill A, Sidhu A, Sharma S, Reddy BK, Padi SS. Peroxisome proliferator-activated receptor-α activation attenuates 3-nitropropionic acid induced behavioral and biochemical alterations in rats: Possible neuroprotective mechanisms. Eur J Pharmacol 2012; 674:33-43. [DOI: 10.1016/j.ejphar.2011.10.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 10/07/2011] [Accepted: 10/15/2011] [Indexed: 01/01/2023]
|
49
|
Abstract
Melatonin mediates neuroprotection in several experimental models of neurodegeneration. It is not yet known, however, whether melatonin provides neuroprotection in genetic models of Huntington's disease (HD). We report that melatonin delays disease onset and mortality in a transgenic mouse model of HD. Moreover, mutant huntingtin (htt)-mediated toxicity in cells, mice, and humans is associated with loss of the type 1 melatonin receptor (MT1). We observe high levels of MT1 receptor in mitochondria from the brains of wild-type mice but much less in brains from HD mice. Moreover, we demonstrate that melatonin inhibits mutant htt-induced caspase activation and preserves MT1 receptor expression. This observation is critical, because melatonin-mediated protection is dependent on the presence and activation of the MT1 receptor. In summary, we delineate a pathologic process whereby mutant htt-induced loss of the mitochondrial MT1 receptor enhances neuronal vulnerability and potentially accelerates the neurodegenerative process.
Collapse
|
50
|
Tasset I, Agüera E, Olmo-Camacho R, Escribano B, Sánchez-López F, Delgado MJ, Cruz AH, Gascón F, Luque E, Peña J, Jimena IM, Túnez I. Melatonin improves 3-nitropropionic acid induced behavioral alterations and neurotrophic factors levels. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:1944-9. [PMID: 21939726 DOI: 10.1016/j.pnpbp.2011.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 08/31/2011] [Accepted: 09/06/2011] [Indexed: 11/17/2022]
Abstract
OBJECTIVE This study sought to determine whether melatonin causes changes in neurotrophic factors and it protects against the mycotoxin 3-nitropropionic acid (3-NP) in brain tissue. METHODS Rats were given 3-NP over four consecutive days (20 mg/kg BW), while melatonin was administered over 21 days (1 mg/kg/BW), starting after the last injection of 3-NP. RESULTS Rats treated with 3-NP displayed significant changes in neurotrophic factor (BDNF and GDNF) levels, together with alterations in behavior; they also displayed extensive oxidative stress and a massive neuronal damage. CONCLUSIONS Melatonin improved behavioral alterations, reduced oxidative damage, lowered neurotrophic factor levels and neuronal loss in 3-NP-treated rats. These results suggest that melatonin exerts a neuroprotective action.
Collapse
Affiliation(s)
- Inmaculada Tasset
- Dpto. Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)/Universidad de Córdoba, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|