1
|
Ohishi K, Rahman AA, Ohkura T, Burns AJ, Goldstein AM, Hotta R. Effects of aged garlic extract on aging?related changes in gastrointestinal function and enteric nervous system cells. Exp Ther Med 2025; 29:103. [PMID: 40171138 PMCID: PMC11959352 DOI: 10.3892/etm.2025.12853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/05/2025] [Indexed: 04/03/2025] Open
Abstract
Dysmotility of the gastrointestinal (GI) tract is commonly seen in elderly individuals, where it causes significant morbidity and can lead to more severe conditions, including sarcopenia and frailty. Although the precise mechanisms underlying aging-related GI dysmotility are not fully understood, neuronal loss or degeneration in the enteric nervous system (ENS) may be involved. Aged garlic extract (AGE) has been shown to have several beneficial effects in the GI tract; however, it is not known whether AGE can improve GI motility in older animals. The aim of the present study was to examine the effects of AGE on the ENS and gut motility in older mice and elucidate potential mechanisms of action. An AGE-formulated diet was given to 18-month-old female mice for 2 weeks. Organ bath studies and cell culture demonstrated that AGE: i) Altered gut contractile activity; ii) enhanced viability of ENS cells; and iii) exhibited neuroprotective effects on the ENS via reduction in oxidative stress. These findings suggest that AGE could be used to develop novel dietary therapeutics for aging-related GI dysmotility by targeting the associated loss and damage of the ENS.
Collapse
Affiliation(s)
- Kensuke Ohishi
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co., Ltd., Akitakata, Hiroshima 739-1195, Japan
| | - Ahmed A. Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Takahiro Ohkura
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Alan J. Burns
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Allan M. Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
2
|
Kheirouri S, Alizadeh H. Dietary diversity and cognitive performance in older adults: a systematic review. BMC Neurol 2025; 25:144. [PMID: 40188075 PMCID: PMC11972542 DOI: 10.1186/s12883-025-04096-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/19/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Promoting dietary diversity (DD), which refers to the variety or the number of different food groups that people eat over the time given, is important for brain health maintenance and may be beneficial for inhibiting neurodegenerative diseases. This research aimed to review the literature and summarize research evidence for achieving an inclusive estimate concerning the relationship between DD and cognitive function in adults. METHODS We systematically queried the databases of PubMed, Web of Science, and Google Scholar, without imposing any date restrictions, up to June 2024 to identify original literature that sheds light on the intricate relationship between DD and cognitive function. Employing rigorous criteria, we meticulously screened studies, eliminating duplicates or those unrelated to our focus. Subsequently, we critically evaluated the findings from the selected studies, descriptively summarizing them. Additionally, we engaged in an in-depth exploration of potential mechanistic pathways linking DD to cognitive performance. RESULTS Of the 388 citations obtained, 23 articles were included in the final review. All the studies reported a positive association between DD score and cognitive functioning and indicated that higher DD was accompanied by good memory (n = 3) and lower risk of cognitive decline (n = 19), dementia (n = 3), and Alzheimer's disease (n = 1). CONCLUSION The results indicate that sustaining a diverse diet among older people may help maintain cognitive functioning. Dietary diversity represents a promising clinical avenue for mitigating cognitive decline associated with diverse brain disorders, potentially preventing or attenuating deterioration.
Collapse
Affiliation(s)
- Sorayya Kheirouri
- Department of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Attar Nishabouri St., POBOX: 14711, Tabriz, 5166614711, Iran.
| | - Hamed Alizadeh
- Student Research Committee, Khoy University of Medical Sciences, Khoy, Iran
| |
Collapse
|
3
|
Changkakoti L, Rajabalaya R, David SR, Balaraman AK, Sivasubramanian H, Mukherjee AK, Bala A. Exploration of the Role of Vitamins in Preventing Neurodegenerative Diseases: Comprehensive Review on Preclinical and Clinical Findings. Curr Neuropharmacol 2025; 23:547-563. [PMID: 39572918 DOI: 10.2174/011570159x327677240902105443] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 04/11/2025] Open
Abstract
Neurodegenerative diseases (NDDs) are a multifaceted and heterogeneous group of complex diseases. Unfortunately, a cure for these conditions has yet to be found, but there are ways to reduce the risk of developing them. Studies have shown that specific vitamins regulate the brain molecules and signaling pathways, which may help prevent degeneration. This review focuses on examining the role of vitamins in preventing five significant types of neurodegenerative diseases, including Parkinson's disease (PD), Alzheimer's disease (AD), Huntington's disease (HD), Multiple Sclerosis (MS), and Amyotrophic Lateral Sclerosis (ALS). This review also highlights promising and controversial findings about the potential impact of vitamins on this group of diseases. Several developed countries standardize daily dietary vitamin intake to meet nutrient requirements, improve health, and prevent chronic diseases like NDDs. However, more research is necessary to gain a more comprehensive understanding of their therapeutic benefits, including studies exploring different drug-dose paradigms, diverse humanized animal models, and clinical trials conducted in various locations.
Collapse
Affiliation(s)
- Liza Changkakoti
- Pharmacology and Drug Discovery Research Laboratory, Division of Life Sciences; Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Guwahati, PIN-781035, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), AcSIR (an Indian Institute of National Importance), Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh PIN-201002, India
| | - Rajan Rajabalaya
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, BE 1410 Bandar Seri Begawan, Brunei Darussalam
| | - Sheba R David
- School of Pharmacy, University of Wyoming, Laramie, Wyoming, 82071, USA
| | - Ashok Kumar Balaraman
- Research Management Unit, University of Cyberjaya, Persiaran Bestari, Cyber 11, 63000, Cyberjaya, Selangor, Malaysia
| | - Hemalatha Sivasubramanian
- Department of Pharmacognosy, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Porur, Chennai, India
| | - Ashis K Mukherjee
- Microbial Biotechnology and Protein Research Laboratory, Division of Life Sciences; Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Guwahati, PIN-781035, Assam, India
| | - Asis Bala
- Pharmacology and Drug Discovery Research Laboratory, Division of Life Sciences; Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Guwahati, PIN-781035, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), AcSIR (an Indian Institute of National Importance), Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh PIN-201002, India
| |
Collapse
|
4
|
Kaya K, Şahin Y, Demirel HH, Çiftçi O. Investigation of oxidative, inflammatory and apoptotic effects of favipiravir use alone and combined with vitamin C on brain tissue of elderly rats. Drug Chem Toxicol 2024; 47:640-648. [PMID: 37424396 DOI: 10.1080/01480545.2023.2233054] [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: 01/06/2023] [Revised: 06/09/2023] [Accepted: 06/23/2023] [Indexed: 07/11/2023]
Abstract
Favipiravir is a nucleoside analogue antiviral drug and inhibits the replication of many RNA viruses, especially influenza viruses. Favipiravir has also been used to treat patients with mild to moderate COVID-19 disease. However, various side effects, including neurological side effects, have been reported related to the use of favipiravir. Therefore, in this study, we aimed to investigate the possible effects of favipiravir alone or in combination with vitamin C on aged rats' brain tissue and the possible mechanisms of these effects. A total of 30 rats used in the study were randomly divided into 5 equal groups and the first group was kept as the control group. High-dose (100 mg/kg) or low-dose (20 mg/kg) favipiravir was administered alone or in combination with vitamin C (150 mg/kg) to other groups. Administration of both high and low doses of favipiravir significantly increased TBARS levels in brain tissue of aged rats. Similarly, both high and low doses of favipiravir led to significant increases in Bcl-2 and caspase-3 relative mRNA expression. However, only low dose favipiravir caused a significant increase in iNOS and IL-1β relative mRNA expression levels. Similar results were also observed in histopathological examinations. However, co-administration of vitamin C with favipiravir attenuated some of the adverse effects of favipiravir. In conclusion, in this study, it was shown that the use of favipiravir caused some adverse effects through oxidative, inflammatory and apoptotic processes in the brain tissue of aged rats, and the potential of vitamin C to alleviate these effects.
Collapse
Affiliation(s)
- Kürşat Kaya
- Department of Medical Biochemistry, Faculty of Medicine, Pamukkale University, Denizli, Türkiye
| | - Yasemin Şahin
- Department of Medical Pharmacology, Faculty of Medicine, Pamukkale University, Denizli, Türkiye
| | - Hasan Hüseyin Demirel
- Bayat Laborant & Veterinary Health Division, Afyon Kocatepe University, Afyon, Türkiye
| | - Osman Çiftçi
- Department of Medical Pharmacology, Faculty of Medicine, Pamukkale University, Denizli, Türkiye
| |
Collapse
|
5
|
Adebayo OL, Agu VA, Idowu GA, Ezejiaku BC, Atunnise AK. The Role of Vitamin C on ATPases Activities in Monosodium Glutamate-Induced Oxidative Stress in Rat Striatum and Cerebellum. Neurotox Res 2024; 42:40. [PMID: 39212807 DOI: 10.1007/s12640-024-00719-x] [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: 01/03/2024] [Revised: 06/04/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
Monosodium glutamate (MSG) is a silent excitotoxin used as a flavour enhancer but exerts serious health hazards to consumers. MSG plays a role in neuronal function as the dominant excitatory neurotransmitter. It is transferred into the blood and ultimately increases brain glutamate levels, causing functional disruptions notably via oxidative stress. The study evaluated the toxic effect of high consumption of MSG and the modulatory role of vitamin C on ATPase activities in the striatum and cerebellum of male Wistar rats for five weeks. Rats were grouped into four (A-D): group A was fed with rat's show only; Group B was fed with diet containing 15% MSG; Group C was treated with vitamin C (200 mg/kg b.wgt orally in 0.9% saline solution) only for 3 weeks; and group D rats were fed with MSG and vitamin C. The findings show that MSG does not affect body and cerebellum weights but increases striatal weight. MSG increases the malondialdehyde (MDA) level and significantly decreases catalase (CAT) and superoxide dismutase (SOD) activities and glutathione (GSH) levels. MSG significantly impaired striatal and cerebellar ATPases activities (Na+/K+-, Ca2+-, Mg2+- and total ATPases). Vitamin C treatment abolishes MSG-induced oxidative stress and improves ATPase activities. The findings show that vitamin C has beneficial effects in improving the functions of membrane-bound ATPases against MSG toxicity in rat's striatum and cerebellum.
Collapse
Affiliation(s)
- Olusegun L Adebayo
- Department of Biochemistry, Faculty of Basic Medical Sciences, Redeemer's University, PMB 230, Ede, Osun State, Nigeria.
| | - Vivian A Agu
- Department of Biochemistry, Faculty of Basic Medical Sciences, Redeemer's University, PMB 230, Ede, Osun State, Nigeria
| | - Grace A Idowu
- Department of Biochemistry, Faculty of Basic Medical Sciences, Redeemer's University, PMB 230, Ede, Osun State, Nigeria
| | - Blessing C Ezejiaku
- Department of Biochemistry, Faculty of Basic Medical Sciences, Redeemer's University, PMB 230, Ede, Osun State, Nigeria
| | - Adeleke K Atunnise
- Department of Biochemistry, Faculty of Basic Medical Sciences, Redeemer's University, PMB 230, Ede, Osun State, Nigeria
| |
Collapse
|
6
|
Ali J, Choe K, Park JS, Park HY, Kang H, Park TJ, Kim MO. The Interplay of Protein Aggregation, Genetics, and Oxidative Stress in Alzheimer's Disease: Role for Natural Antioxidants and Immunotherapeutics. Antioxidants (Basel) 2024; 13:862. [PMID: 39061930 PMCID: PMC11274292 DOI: 10.3390/antiox13070862] [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: 06/03/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that comprises amyloid-beta protein (Aβ) as a main component of neuritic plaques. Its deposition is considered a trigger for AD pathogenesis, progression, and the clinical symptoms of cognitive impairment. Some distinct pathological features of AD include phosphorylation of tau protein, oxidative stress, and mitochondrial dysfunction. These pathological consequences tend to produce reactive oxygen species (ROS), resulting in the dysregulation of various signaling pathways of neuroinflammation and neurodegeneration. The relationship between the Aβ cascade and oxidative stress in AD pathogenesis is like a "chicken and egg" story, with the etiology of the disease regarding these two factors remaining a question of "which comes first." However, in this review, we have tried our best to clarify the interconnection between these two mechanisms and to show the precise cause-and-effect relationship. Based on the above hallmarks of AD, several therapeutic strategies using natural antioxidants, monoclonal antibodies, and vaccines are employed as anti-Aβ therapy to decrease ROS, Aβ burden, chronic neuroinflammation, and synaptic failure. These natural antioxidants and immunotherapeutics have demonstrated significant neuroprotective effects and symptomatic relief in various in vitro and in vivo models, as well as in clinical trials for AD. However, none of them have received final approval to enter the drug market for mitigating AD. In this review, we extensively elaborate on the pitfalls, assurances, and important crosstalk between oxidative stress and Aβ concerning current anti-Aβ therapy. Additionally, we discuss future strategies for the development of more Aβ-targeted approaches and the optimization of AD treatment and mitigation.
Collapse
Affiliation(s)
- Jawad Ali
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.A.); (K.C.); (J.S.P.)
| | - Kyonghwan Choe
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.A.); (K.C.); (J.S.P.)
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Jun Sung Park
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.A.); (K.C.); (J.S.P.)
| | - Hyun Young Park
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands;
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), 6202 AZ Maastricht, The Netherlands
| | - Heeyoung Kang
- Department of Neurology, Gyeongsang National University Hospital & College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea;
| | - Tae Ju Park
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences (MVLS), University of Glasgow, Glasgow G12 0ZD, UK
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.A.); (K.C.); (J.S.P.)
- Alz-Dementia Korea Co., Jinju 52828, Republic of Korea
| |
Collapse
|
7
|
Obadimu AA, Adebayo OL, Tugbobo-Amisu AO, Fagbohunka BS, Adenuga GA. Effect of Selenium and Zinc Supplementation on Reproductive Organs Following Postnatal Protein Malnutrition. Biol Trace Elem Res 2024; 202:1126-1139. [PMID: 37393387 DOI: 10.1007/s12011-023-03751-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/24/2023] [Indexed: 07/03/2023]
Abstract
Protein diets are required for the normal development of the reproductive system and their inadequacy or deficiency might have hazardous functional complications during maturational and developmental stages. The study was carried out to evaluate the effect of selenium (Se) and zinc (Zn) supplementation on the male and female reproductive organs of rats with postnatal protein malnutrition. Male and female weanling rats were randomly assigned to six groups respectively. The adequate protein diet rats were fed with 16% casein diet while the protein malnourished diet (PMD) rats were fed with 5% casein diet. After the 8th week of feeding, Se (sodium selenite; Na2SeO3) and Zn (zinc sulfate; ZnSO4·7H2O) were supplemented for 3 weeks. The growth curve of body weights, lipid profile, testosterone and progesterone level, Na+-K+-ATPase activity, oxidative stress, and antioxidant status were evaluated. The results showed that PMD reduced the body weights of male and female rats. It also reduced the activities of catalase and glutathione peroxidase in the testes, but reductions in superoxide dismutase and glutathione-S-transferase activities, glutathione, vitamins C and E, testosterone, and progesterone levels were observed in both the testes and ovaries. Furthermore, PMD increased the nitric oxide level in both organs and altered the plasma lipid profiles in both sexes. Se and Zn supplementation, however, restored almost all the alterations observed in all the parameters analyzed. In conclusion, Se and Zn supplementation protects the male and female reproductive organs of rats against postnatal protein malnutrition.
Collapse
Affiliation(s)
- Adedayo Adedeji Obadimu
- Department of Biochemistry, Faculty of Basic Medical Sciences, Obafemi Awolowo College of Health Sciences, Olabisi Onabanjo University, Sagamu, Ogun State, Nigeria
| | - Olusegun Lateef Adebayo
- Department of Biochemistry, Faculty of Basic Medical Sciences, Redeemer's University, Ede, Osun State, P.M.B. 230, Nigeria.
| | - Adesewa Omolara Tugbobo-Amisu
- Department of Food Technology,, Federal Institute of Industrial Research Oshodi (FIIRO), Lagos, Lagos State, Nigeria
| | - Bamidele Sanya Fagbohunka
- Department of Biochemistry, Faculty of Basic Medical Sciences, Obafemi Awolowo College of Health Sciences, Olabisi Onabanjo University, Sagamu, Ogun State, Nigeria
| | - Gbenga Adebola Adenuga
- Department of Biochemistry, Faculty of Basic Medical Sciences, Obafemi Awolowo College of Health Sciences, Olabisi Onabanjo University, Sagamu, Ogun State, Nigeria
| |
Collapse
|
8
|
Holton K. The potential role of dietary intervention for the treatment of neuroinflammation. TRANSLATIONAL NEUROIMMUNOLOGY, VOLUME 7 2023:239-266. [DOI: 10.1016/b978-0-323-85841-0.00022-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
9
|
Bulama I, Nasiru S, Bello A, Abbas AY, Nasiru JI, Saidu Y, Chiroma MS, Mohd Moklas MA, Mat Taib CN, Waziri A, Suleman BL. Antioxidant-based neuroprotective effect of dimethylsulfoxide against induced traumatic brain injury in a rats model. Front Pharmacol 2022; 13:998179. [PMID: 36353489 PMCID: PMC9638698 DOI: 10.3389/fphar.2022.998179] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/17/2022] [Indexed: 08/03/2023] Open
Abstract
Traumatic brain injury (TBI) has been the result of neurological deficit and oxidative stress. This study evaluated the antioxidative neuroprotective property and learning and memory-enhancing effects of dimethyl sulfoxide (DMSO) in a rat model after the induction of TBI. 21 albino rats with 7 rats per group were used in this study. Group I was induced with TBI and treated with DMSO at 67.5 mg/kg orally once daily which started 30 min after the induction of TBI and lasted 21 days. Group II was induced with TBI but not treated while Group III was neither induced with TBI nor treated. Assessment of behavioral function (Learning and memory, anxiety and motor function), the level of an antioxidant enzymes and their gene expression (superoxide dismutase, catalase, glutathione peroxidase), the biomarkers of oxidative stress (malondialdehyde) and S100B levels as well as brain tissues histological studies were conducted. Administration of DMSO to rats with induced TBI has improved learning and memory, locomotor function and decreased anxiety in Group I compared to Group II. Moreover, the level of S100B was significantly (p < 0.05) lower in Group I compared to Group II. Treatment with DMSO also decreased lipid peroxidation significantly (p < 0.05) compared to Group II. There exists a significant (p < 0.05) increase in CAT, SOD, and GPX activities in Group I compared to Group II. Therefore, DMSO has demonstrated a potential antioxidative neuroprotective effect through its ability to increase the level of antioxidant enzymes which they quench and inhibit the formation of ROS, thereby improving cognitive functions.
Collapse
Affiliation(s)
- Ibrahim Bulama
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Maiduguri, Maiduguri, Nigeria
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Suleiman Nasiru
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, Usman Danfodiyo University, Sokoto, Nigeria
| | - Abubakar Bello
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Abdullahi Yahaya Abbas
- Department of Biochemistry, Faculty of Chemical and Life Sciences, Usman Danfodiyo University, Sokoto, Nigeria
| | - Jinjiri Ismail Nasiru
- Department of Surgery, Faculty of Clinical Sciences, Usman Danfodiyo University Teaching Hospital, Sokoto, Nigeria
| | - Yusuf Saidu
- Department of Biochemistry, Faculty of Chemical and Life Sciences, Usman Danfodiyo University, Sokoto, Nigeria
| | - Musa Samaila Chiroma
- Department of Human Anatomy, Faculty of Basic Clinical Sciences, University of Maiduguri, Maiduguri, Nigeria
| | - Mohamad Aris Mohd Moklas
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Che Norma Mat Taib
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Ali Waziri
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Maiduguri, Maiduguri, Nigeria
| | - Bilbis Lawal Suleman
- Department of Biochemistry, Faculty of Chemical and Life Sciences, Usman Danfodiyo University, Sokoto, Nigeria
| |
Collapse
|
10
|
Hamid M, Mansoor S, Amber S, Zahid S. A quantitative meta-analysis of vitamin C in the pathophysiology of Alzheimer’s disease. Front Aging Neurosci 2022; 14:970263. [PMID: 36158537 PMCID: PMC9490219 DOI: 10.3389/fnagi.2022.970263] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/29/2022] [Indexed: 11/28/2022] Open
Abstract
Purpose Alzheimer’s disease (AD) is a multifaceted neurodegenerative disorder with many complex pathways feeding into its pathogenesis and progression. Vitamin C, an essential dietary antioxidant, is vital for proper neurological development and maintenance. This meta-analysis and systematic review attempted to define the relationship between vitamin C plasma levels and AD while highlighting the importance and involvement of vitamin C in the pathogenesis of AD. Materials and methods PRISMA guidelines were used to obtain studies quantifying the plasma levels of vitamin C in AD and control subjects. The literature was searched in the online databases PubMed, Google Scholar, and Web of Science. A total of 12 studies were included (n = 1,100) and analyzed using Comprehensive Meta-Analysis 3.0. Results The results show that there is a significant decrease in the plasma vitamin C levels of AD patients as compared to healthy controls (pooled SMD with random-effect model: −1.164, with 95%CI: −1.720 to −0.608, Z = −4.102, p = 0.00) with significant heterogeneity (I2 = 93.218). The sensitivity analysis showed directionally similar results. Egger’s regression test (p = 0.11) and visual inspection of the funnel plot showed no publication bias. Conclusion Based on these studies, it can be deduced that the deficiency of vitamin C is involved in disease progression and supplementation is a plausible preventive and treatment strategy. However, clinical studies are warranted to elucidate its exact mechanistic role in AD pathophysiology and prevention.
Collapse
|
11
|
Marino A, Battaglini M, Moles N, Ciofani G. Natural Antioxidant Compounds as Potential Pharmaceutical Tools against Neurodegenerative Diseases. ACS OMEGA 2022; 7:25974-25990. [PMID: 35936442 PMCID: PMC9352343 DOI: 10.1021/acsomega.2c03291] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/29/2022] [Indexed: 06/01/2023]
Abstract
Natural antioxidants are a very large diversified family of molecules classified by activity (enzymatic or nonenzymatic), chemical-physical properties (e.g., hydrophilic or lipophilic), and chemical structure (e.g., vitamins, polyphenols, etc.). Research on natural antioxidants in various fields, such as pharmaceutics, nutraceutics, and cosmetics, is among the biggest challenges for industry and science. From a biomedical point of view, the scavenging activity of reactive oxygen species (ROS) makes them a potential tool for the treatment of neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, dementia, and amyotrophic lateral sclerosis (ALS). In addition to the purified phytochemical compounds, a variety of natural extracts characterized by a complex mixture of antioxidants and anti-inflammatory molecules have been successfully exploited to rescue preclinical models of these diseases. Extracts derived from Ginkgo biloba, grape, oregano, curcumin, tea, and ginseng show multitherapeutic effects by synergically acting on different biochemical pathways. Furthermore, the reduced toxicity associated with many of these compounds limits the occurrence of side effects. The support of nanotechnology for improving brain delivery, controlling release, and preventing rapid degradation and excretion of these compounds is of fundamental importance. This review reports on the most promising results obtained on in vitro systems, in vivo models, and in clinical trials, by exploiting natural-derived antioxidant compounds and extracts, in their free form or encapsulated in nanocarriers.
Collapse
Affiliation(s)
- Attilio Marino
- Istituto
Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
| | - Matteo Battaglini
- Istituto
Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
| | - Nadia Moles
- Istituto
Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
- Politecnico
di Torino, Department of Mechanical
and Aerospace Engineering, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Gianni Ciofani
- Istituto
Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
| |
Collapse
|
12
|
AL-Nasser MN, Mellor IR, Carter WG. Is L-Glutamate Toxic to Neurons and Thereby Contributes to Neuronal Loss and Neurodegeneration? A Systematic Review. Brain Sci 2022; 12:577. [PMID: 35624964 PMCID: PMC9139234 DOI: 10.3390/brainsci12050577] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 01/27/2023] Open
Abstract
L-glutamate (L-Glu) is a nonessential amino acid, but an extensively utilised excitatory neurotransmitter with critical roles in normal brain function. Aberrant accumulation of L-Glu has been linked to neurotoxicity and neurodegeneration. To investigate this further, we systematically reviewed the literature to evaluate the effects of L-Glu on neuronal viability linked to the pathogenesis and/or progression of neurodegenerative diseases (NDDs). A search in PubMed, Medline, Embase, and Web of Science Core Collection was conducted to retrieve studies that investigated an association between L-Glu and pathology for five NDDs: Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). Together, 4060 studies were identified, of which 71 met eligibility criteria. Despite several inadequacies, including small sample size, employment of supraphysiological concentrations, and a range of administration routes, it was concluded that exposure to L-Glu in vitro or in vivo has multiple pathogenic mechanisms that influence neuronal viability. These mechanisms include oxidative stress, reduced antioxidant defence, neuroinflammation, altered neurotransmitter levels, protein accumulations, excitotoxicity, mitochondrial dysfunction, intracellular calcium level changes, and effects on neuronal histology, cognitive function, and animal behaviour. This implies that clinical and epidemiological studies are required to assess the potential neuronal harm arising from excessive intake of exogenous L-Glu.
Collapse
Affiliation(s)
- Maryam N. AL-Nasser
- Department of Biological Sciences, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
- School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 3DT, UK
| | - Ian R. Mellor
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Wayne G. Carter
- School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 3DT, UK
| |
Collapse
|
13
|
Kumar RR, Singh L, Thakur A, Singh S, Kumar B. Role of Vitamins in Neurodegenerative Diseases: A Review. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2021; 21:766-773. [PMID: 34802410 DOI: 10.2174/1871527320666211119122150] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/12/2021] [Accepted: 08/30/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Vitamins are the micronutrients required for boosting the immune system and managing any future infection. Vitamins are involved in neurogenesis, a defense mechanism working in neurons, metabolic reactions, neuronal survival, and neuronal transmission. Their deficiency leads to abnormal functions in the brain like oxidative stress, mitochondrial dysfunction, accumulation of proteins (synuclein, Aβ plaques), neurodegeneration, and excitotoxicity. METHODS In this review, we have compiled various reports collected from PubMed, Scholar Google, Research gate, and Science direct. The findings were evaluated, compiled, and represented in this manuscript. CONCLUSION The deficiency of vitamins in the body causes various neurological disorders like Alzheimer's disease, Parkinson's disease, Huntington's disease, and depression. We have discussed the role of vitamins in neurological disorders and the normal human body. Depression is linked to a deficiency of vitamin-C and vitamin B. In the case of Alzheimer's disease, there is a lack of vitamin-B1, B12, and vitamin-A, which results in Aβ-plaques. Similarly, in Parkinson's disease, vitamin-D deficiency leads to a decrease in the level of dopamine, and imbalance in vitamin D leads to accumulation of synuclein. In MS, Vitamin-C and Vitamin-D deficiency causes demyelination of neurons. In Huntington's disease, vitamin- C deficiency decreases the antioxidant level, enhances oxidative stress, and disrupts the glucose cycle. Vitamin B5 deficiency in Huntington's disease disrupts the synthesis of acetylcholine and hormones in the brain.
Collapse
Affiliation(s)
- Ravi Ranjan Kumar
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Ferozpur G.T. Road MOGA-142001, Punjab. India
| | - Lovekesh Singh
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Ferozpur G.T. Road MOGA-142001, Punjab. India
| | - Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031. Taiwan
| | - Shamsher Singh
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Ferozpur G.T. Road MOGA-142001, Punjab. India
| | - Bhupinder Kumar
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Ferozpur G.T. Road MOGA-142001, Punjab. India
| |
Collapse
|
14
|
Altaher W, Alhelo H, Chosky D, Kulesza RJ. Neonatal exposure to monosodium glutamate results in impaired auditory brainstem structure and function. Hear Res 2021; 405:108243. [PMID: 33865019 DOI: 10.1016/j.heares.2021.108243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 03/14/2021] [Accepted: 03/31/2021] [Indexed: 10/21/2022]
Abstract
Excitotoxic injury during the neonatal period has been shown to result in neurodegenerative changes in several different brain regions. Exposure to monosodium glutamate (MSG) during the first two postnatal weeks results in glutamate neurotoxicity in the cochlea and has been shown to result in damage to cochlear hair cells and fewer neurons in the spiral ganglion. Further, we have shown that such exposure results in fewer neurons in the cochlear nucleus and superior olivary complex and abnormal expression of the calcium binding proteins calbindin and calretinin. Based on these findings, we hypothesized that neonatal MSG exposure would result in loss of neurons at more rostral levels in the auditory brainstem, and this exposure would result in abnormal brainstem auditory evoked potentials. We identified a significantly lower density of neurons in the spiral ganglion, heterogenous loss of neurons in the globular bushy cell-trapezoid body circuit, and fewer neurons in the nuclei of the lateral lemniscus and central nucleus of the inferior colliculus. The most severe loss of neurons was found in the inferior colliculus. Click-evoked auditory brainstem responses revealed significantly higher thresholds and longer latency responses, but these did not deteriorate with age. These results, together with our previous findings, indicate that neonatal exposure to MSG results in fewer neurons throughout the entire auditory brainstem and results in abnormal auditory brainstem responses.
Collapse
Affiliation(s)
- Weam Altaher
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, 1858 West Grandview Blvd, Erie, PA 16504, United States
| | - Hasan Alhelo
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, 1858 West Grandview Blvd, Erie, PA 16504, United States
| | - Devon Chosky
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, 1858 West Grandview Blvd, Erie, PA 16504, United States
| | - Randy J Kulesza
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, 1858 West Grandview Blvd, Erie, PA 16504, United States.
| |
Collapse
|
15
|
Ho TC, Teresi GI, Segarra JR, Ojha A, Walker JC, Gu M, Spielman DM, Sacchet MD, Jiang F, Rosenberg-Hasson Y, Maecker H, Gotlib IH. Higher Levels of Pro-inflammatory Cytokines Are Associated With Higher Levels of Glutamate in the Anterior Cingulate Cortex in Depressed Adolescents. Front Psychiatry 2021; 12:642976. [PMID: 33935833 PMCID: PMC8081972 DOI: 10.3389/fpsyt.2021.642976] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/26/2021] [Indexed: 12/14/2022] Open
Abstract
Animal models of stress and related conditions, including depression, have shown that elevated peripheral levels of inflammatory cytokines have downstream consequences on glutamate (Glu) in the brain. Although studies in human adults with depression have reported evidence of higher inflammation but lower Glu in the anterior cingulate cortex (ACC), the extent to which peripheral inflammation contributes to glutamatergic abnormalities in adolescents with depression is not well-understood. It is also unclear whether antioxidants, such as ascorbate (Asc), may buffer against the effects of inflammation on Glu metabolism. Fifty-five depressed adolescents were recruited in the present cross-sectional study and provided blood samples, from which we assayed pro-inflammatory cytokines, and underwent a short-TE proton magnetic spectroscopy scan at 3T, from which we estimated Glu and Asc in the dorsal ACC. In the 31 adolescents with usable cytokine and Glu data, we found that IL-6 was significantly positively associated with dorsal ACC Glu (β = 0.466 ± 0.199, p = 0.029). Of the 16 participants who had usable Asc data, we found that at higher levels of dorsal ACC Asc, there was a negative association between IL-6 and Glu (interaction effect: β = -0.906 ± 0.433, p = 0.034). Importantly, these results remained significant when controlling for age, gender, percentage of gray matter in the dorsal ACC voxel, BMI, and medication (antidepressant and anti-inflammatory) usage. While preliminary, our results underscore the importance of examining both immune and neural contributors to depression and highlight the potential role of anti-inflammatory compounds in mitigating the adverse effects of inflammation (e.g., glutamatergic neuroexcitotoxicity). Future studies that experimentally manipulate levels of inflammation, and of ascorbate, and that characterize these effects on cortical glutamate concentrations and subsequent behavior in animals and in humans are needed.
Collapse
Affiliation(s)
- Tiffany C Ho
- Department of Psychiatry and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Giana I Teresi
- Department of Psychology, Stanford University, Stanford, CA, United States
| | - Jillian R Segarra
- Department of Psychology, Stanford University, Stanford, CA, United States
| | - Amar Ojha
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - Johanna C Walker
- Department of Psychology, Stanford University, Stanford, CA, United States
| | - Meng Gu
- Department of Radiology, Stanford University, Stanford, CA, United States
| | - Daniel M Spielman
- Department of Radiology, Stanford University, Stanford, CA, United States
| | - Matthew D Sacchet
- Center for Depression, Anxiety, and Stress Research, McLean Hospital and Harvard Medical School, Belmont, MA, United States
| | - Fei Jiang
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, United States
| | - Yael Rosenberg-Hasson
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Holden Maecker
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Ian H Gotlib
- Department of Psychology, Stanford University, Stanford, CA, United States
| |
Collapse
|
16
|
Plasma Vitamin C Concentrations Were Negatively Associated with Tingling, Prickling or Pins and Needles Sensation in Patients with Postherpetic Neuralgia. Nutrients 2020; 12:nu12082384. [PMID: 32784896 PMCID: PMC7468915 DOI: 10.3390/nu12082384] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Vitamin C deficiency increases the risk of postherpetic neuralgia (PHN). In this cross-sectional study, the relationships among plasma vitamin C concentrations, pain and Leeds assessment of neuropathic symptoms and signs (LANSS) items were investigated during their first pain clinic visit of 120 PHN patients. The factors associated with vitamin C deficiency were determined. Independent predictors of vitamin C deficiency were presented as adjusted odds ratios (AOR) and 95% confidence intervals (CI). The patients had a high prevalence (52.5%) of vitamin C deficiency. Their plasma vitamin C concentrations were negatively associated with spontaneous pain and tingling, prickling or pins and needles sensation according to the LANSS questionnaire. Based on the receiver operator characteristic curve, the cutoffs for plasma vitamin C to predict moderate-to-severe and severe symptoms of sharp sensation were <7.05 and <5.68 mg/L, respectively. By comparison, the patients well-nourished with vitamin C had lower incidences of sharp sensations, sharp pain, and reddish skin. Multivariate analyses revealed that vitamin C deficiency was associated with the low intake of fruit/vegetables (AOR 2.66, 95% CI 1.09–6.48, p = 0.032), peptic ulcer disease (AOR 3.25, 95% CI 1.28–8.28, p = 0.014), and smoking (AOR 3.60, 95% CI 1.33–9.77, p = 0.010). Future studies are needed to substantiate these findings.
Collapse
|
17
|
White matter injury and neurodevelopmental disabilities: A cross-disease (dis)connection. Prog Neurobiol 2020; 193:101845. [PMID: 32505757 DOI: 10.1016/j.pneurobio.2020.101845] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 05/19/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022]
Abstract
White matter (WM) injury, once known primarily in preterm newborns, is emerging in its non-focal (diffused), non-necrotic form as a critical component of subtle brain injuries in many early-life diseases like prematurity, intrauterine growth restriction, congenital heart defects, and hypoxic-ischemic encephalopathy. While advances in medical techniques have reduced the number of severe outcomes, the incidence of tardive impairments in complex cognitive functions or psychopathology remains high, with lifelong detrimental effects. The importance of WM in coordinating neuronal assemblies firing and neural groups synchronizing within multiple frequency bands through myelination, even mild alterations in WM structure, may interfere with the cognitive performance that increasing social and learning demands would exploit tardively during children growth. This phenomenon may contribute to explaining longitudinally the high incidence of late-appearing impairments that affect children with a history of perinatal insults. Furthermore, WM abnormalities have been highlighted in several neuropsychiatric disorders, such as autism and schizophrenia. In this review, we gather and organize evidence on how diffused WM injuries contribute to neurodevelopmental disorders through different perinatal diseases and insults. An insight into a possible common, cross-disease, mechanism, neuroimaging and monitoring, biomarkers, and neuroprotective strategies will also be presented.
Collapse
|
18
|
Ahmad F, Haque S, Ravinayagam V, Ahmad A, Kamli MR, Barreto GE, Ghulam Md Ashraf. Developmental lead (Pb)-induced deficits in redox and bioenergetic status of cerebellar synapses are ameliorated by ascorbate supplementation. Toxicology 2020; 440:152492. [PMID: 32407874 DOI: 10.1016/j.tox.2020.152492] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/08/2020] [Accepted: 05/07/2020] [Indexed: 10/24/2022]
Abstract
Neurotoxicity induced by exposure to heavy metal lead (Pb) is a concern of utmost importance particularly for countries with industrial-based economies. The developing brain is especially sensitive to exposure to even minute quantities of Pb which can alter neurodevelopmental trajectory with irreversible effects on motor, emotive-social and cognitive attributes even into later adulthood. Chemical synapses form the major pathway of inter-neuronal communications and are prime candidates for higher order brain (motor, memory and behavior) functions and determine the resistance/susceptibility for neurological disorders, including neuropsychopathologies. The synaptic pathways and mechanisms underlying Pb-mediated alterations in neuronal signaling and plasticity are not completely understood. Employing a biochemically isolated synaptosomal fraction which is enriched in synaptic terminals and synaptic mitochondria, this study aimed to analyze the alterations in bioenergetic and redox/antioxidant status of cerebellar synapses induced by developmental exposure to Pb (0.2 %). Moreover, we test the efficacy of vitamin C (ascorbate; 500 mg/kg body weight), a neuroprotective and neuromodulatory antioxidant, in mitigation of Pb-induced neuronal deficits. Our results implicate redox and bioenergetic disruptions as an underlying feature of the synaptic dysfunction observed in developmental Pb neurotoxicity, potentially contributing to consequent deficits in motor, behavioral and psychological attributes of the organisms. In addition, we establish ascorbate as a key ingredient for therapeutic approach against Pb induced neurotoxicity, particularly for early-life exposures.
Collapse
Affiliation(s)
- Faraz Ahmad
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand.
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia
| | - Vijaya Ravinayagam
- Deanship of Scientific Research, Department of Nano-medicine Research, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
| | - Aqeel Ahmad
- Department of Medical Biochemistry, College of Medicine, Shaqra University, Shaqra, 11961, Saudi Arabia
| | - Majid Rasool Kamli
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, 21589, Saudi Arabia; Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Ireland
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
19
|
Zhao L, Zhang J, Hu C, Wang T, Lu J, Wu C, Chen L, Jin M, Ji G, Cao Q, Jiang Y. Apigenin Prevents Acetaminophen-Induced Liver Injury by Activating the SIRT1 Pathway. Front Pharmacol 2020; 11:514. [PMID: 32425778 PMCID: PMC7212374 DOI: 10.3389/fphar.2020.00514] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 04/01/2020] [Indexed: 01/29/2023] Open
Abstract
Acetaminophen (APAP) overdose is the main cause of acute liver failure. Apigenin (API) is a natural dietary flavonol with high antioxidant capacity. Herein, we investigated protection by API against APAP-induced liver injury in mice, and explored the potential mechanism. Cell viability assays and mice were used to evaluate the effects of API against APAP-induced liver injury. Western blotting, immunofluorescence staining, RT-PCR, and Transmission Electron Microscope were carried out to determine the signalling pathways affected by API. Analysis of mouse serum levels of alanine/aspartate aminotransferase (ALT/AST), malondialdehyde (MDA), liver myeloperoxidase (MPO) activity, glutathione (GSH), and reactive oxygen species (ROS) revealed that API (80 mg/kg) owned protective effect on APAP-induced liver injury. Meanwhile, API ameliorated the decreased cell viability in L-02 cells incubated by APAP with a dose dependent. Furthermore, API promoted SIRT1 expression and deacetylated p53. Western blotting showed that API promoted APAP-induced autophagy, activated the NRF2 pathway, and inhibited the transcriptional activation of nuclear p65 in the presence of APAP. Furthermore, SIRT1 inhibitor EX-527 reduced protection by API against APAP-induced hepatotoxicity. Molecular docking results indicate potential interaction between API and SIRT1. API prevents APAP-induced liver injury by regulating the SIRT1-p53 axis, thereby promoting APAP-induced autophagy and ameliorating APAP-induced inflammatory responses and oxidative stress injury.
Collapse
Affiliation(s)
- Licong Zhao
- Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Second Clinical College, China Medical University, Shenyang, China
| | - Jiaqi Zhang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cheng Hu
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Wang
- Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Juan Lu
- Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenqu Wu
- Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Long Chen
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingming Jin
- Shanghai University of Medicine & Health Sciences of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qin Cao
- Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanye Jiang
- Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
20
|
Rajabian A, Sadeghnia HR, Hosseini A, Mousavi SH, Boroushaki MT. 3-Acetyl-11-keto-β-boswellic acid attenuated oxidative glutamate toxicity in neuron-like cell lines by apoptosis inhibition. J Cell Biochem 2020; 121:1778-1789. [PMID: 31642100 DOI: 10.1002/jcb.29413] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 10/04/2019] [Indexed: 12/18/2022]
Abstract
3-Acetyl-11-keto-β-boswellic acid (AKBA), a pentacyclic triterpenic acid present in gum resin of Boswellia serrata, has been found to possess antioxidant and neuroprotective properties. In this study, we aimed to examine protective properties of AKBA against glutamate-induced neuronal injury. To investigate the effects of AKBA (2.5-10 µM) on glutamate injury in neuron-like cells PC12 and N2a, two treatment regimens (incubation for 2 or 0 hours before glutamate exposure) were used. Then, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method was used to determine viability of the cells. Cellular redox status was evaluated using fluorimetry and comet assays. Annexin V/propidium iodide double staining and Western blot analysis of relative apoptotic proteins were conducted. Based on the results, 24 hours incubation with glutamate (8 mM) increased the cell mortality of PC12 and N2a (P < .001). However, AKBA (2.5-10 µM) enhanced the cell viability in both treatment regimens (P < .001). Also co- and pretreatment with AKBA significantly attenuated lipid peroxidation, reactive oxygen species production, and DNA injury (P < .05 and P < .001). AKBA also restored the activity of cellular superoxide dismutase under glutamate toxicity; this effect was seen to be more significant during the pretreatment regimen (P < .001). Moreover, Western blot analysis indicated that AKBA inhibited glutamate-induced programmed cell death through depressing the elevation of the expression ratio of Bax/Bcl-2 and cleaved-caspase-3 proteins, concentration-dependently. Overall, the present findings suggest the neuroprotective activities of AKBA against glutamate-induced cell injury probably by inhibiting oxidative damage and reducing apoptotic cell death.
Collapse
Affiliation(s)
- Arezoo Rajabian
- Pharmacological Research Center of Medicinal Plants, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Sadeghnia
- Pharmacological Research Center of Medicinal Plants, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Azar Hosseini
- Pharmacological Research Center of Medicinal Plants, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Hadi Mousavi
- Pharmacological Research Center of Medicinal Plants, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Taher Boroushaki
- Pharmacological Research Center of Medicinal Plants, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
21
|
Ballaz SJ, Rebec GV. Neurobiology of vitamin C: Expanding the focus from antioxidant to endogenous neuromodulator. Pharmacol Res 2019; 146:104321. [PMID: 31229562 DOI: 10.1016/j.phrs.2019.104321] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/16/2019] [Accepted: 06/18/2019] [Indexed: 01/06/2023]
Abstract
Ascorbic acid (AA) is a water-soluble vitamin (C) found in all bodily organs. Most mammals synthesize it, humans are required to eat it, but all mammals need it for healthy functioning. AA reaches its highest concentration in the brain where both neurons and glia rely on tightly regulated uptake from blood via the glucose transport system and sodium-coupled active transport to accumulate and maintain AA at millimolar levels. As a prototype antioxidant, AA is not only neuroprotective, but also functions as a cofactor in redox-coupled reactions essential for the synthesis of neurotransmitters (e.g., dopamine and norepinephrine) and paracrine lipid mediators (e.g., epoxiecoisatrienoic acids) as well as the epigenetic regulation of DNA. Although redox capacity led to the promotion of AA in high doses as potential treatment for various neuropathological and psychiatric conditions, ample evidence has not supported this therapeutic strategy. Here, we focus on some long-neglected aspects of AA neurobiology, including its modulatory role in synaptic transmission as demonstrated by the long-established link between release of endogenous AA in brain extracellular fluid and the clearance of glutamate, an excitatory amino acid. Evidence that this link can be disrupted in animal models of Huntington´s disease is revealing opportunities for new research pathways and therapeutic applications (e.g., epilepsy and pain management). In fact, we suggest that improved understanding of the regulation of endogenous AA and its interaction with key brain neurotransmitter systems, rather than administration of AA in excess, should be the target of future brain-based therapies.
Collapse
Affiliation(s)
- Santiago J Ballaz
- School of Biological Sciences and Engineering, Yachay Tech University, Urcuqui, Ecuador.
| | - George V Rebec
- Program in Neuroscience, Department Psychological & Brain Sciences, Indiana University, Bloomington, USA.
| |
Collapse
|
22
|
Walia V, Garg C, Garg M. Nitrergic signaling modulation by ascorbic acid treatment is responsible for anxiolysis in mouse model of anxiety. Behav Brain Res 2019; 364:85-98. [PMID: 30738102 DOI: 10.1016/j.bbr.2019.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/16/2019] [Accepted: 02/04/2019] [Indexed: 10/27/2022]
Abstract
The present study was designed to investigate the effect of ascorbic acid (AA) treatment on the anxiety related behavioral and neurochemical alterations. AA (50, 100 and 200 mg/kg, i.p.) was administered to the mice and anxiety related behavior and levels of glutamate and nitrite in the brain of mice were determined. The results obtained revealed that the administration of AA (100 mg/kg, i.p.) significantly reduced the anxiety related behavior and the levels of nitrite in the brain of mice. Nitrergic interactions were further determined by the pretreatment of mice with nitric oxide (NO) modulator and AA treatment followed by behavioral and neurochemical measurements. The results obtained suggested that NO inhibition potentiated the anxiolytic like activity of AA in mice. It was also observed that the glutamate and nitrite level in the brain of mice were significantly reduced by the NO inhibitor pretreatment. Thus, the present study demonstrated the possible nitrergic pathways modulation in the anxiolytic like activity of AA in mice.
Collapse
Affiliation(s)
- Vaibhav Walia
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak-124001, Haryana, India.
| | - Chanchal Garg
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak-124001, Haryana, India.
| | - Munish Garg
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak-124001, Haryana, India.
| |
Collapse
|
23
|
Xiong S, Song Y, Liu J, Du Y, Ding Y, Wei H, Bryan K, Ma F, Mao L. Neuroprotective effects of MK-801 on auditory cortex in salicylate-induced tinnitus: Involvement of neural activity, glutamate and ascorbate. Hear Res 2019; 375:44-52. [PMID: 30795964 DOI: 10.1016/j.heares.2019.01.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 01/17/2019] [Accepted: 01/30/2019] [Indexed: 12/14/2022]
Abstract
Tinnitus may cause anxiety, depression, insomnia, which impair the quality of life of millions worldwide. However, the mechanism of tinnitus remains to be understood, it has been previously hypothesized that the activation of N-methyl-D-aspartate (NMDA) receptor is involved in the tinnitus processes and blockade of the NMDA receptor is regarded as a therapeutic strategy for tinnitus treatment even if the rescue treatment is still proved invalid in some cases. To demonstrate the therapeutic effect of the NMDA receptor blocker on tinnitus, we examined here the spontaneous firing rate (SFR) and the neurochemical dynamics in the auditory cortex (AC) of rats after sodium salicylate (SS) injection, which is a widely used model for tinnitus research. We also recorded their responses to MK-801 treatment. Electrophysiological studies showed that MK-801 significantly suppresses SFR in AC of rats with SS-induced tinnitus. In addition, by using a technique that combining in vivo microdialysis with an online electrochemical system (OECS) and a high-performance liquid chromatography (HPLC), we found that the levels of both glutamate and ascorbate in AC dramatically increased after SS injection and that MK-801 administration attenuated those response. Further studies found that MK-801 given at a time point of 30 min pre- or post-injection of SS were more effective than that given at a time point of 60 min post-SS injection, indicating that the time point of MK-801 intervention has a critical impact on the therapeutic effect. These findings suggest that MK-801 plays a neuroprotective role against hyperactivity during tinnitus induced by SS and that the therapeutic effect depends on the time point of MK-801 intervention, which would advance the studies on understanding of the therapeutic potential of NMDA receptor antagonist in tinnitus therapy.
Collapse
Affiliation(s)
- Shan Xiong
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Yu Song
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Junxiu Liu
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Yali Du
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Yujing Ding
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Huan Wei
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, The Chinese Academy of Sciences, Beijing, 100190, China
| | - Kevin Bryan
- Junipero Serra High School, San Mateo, CA, USA
| | - Furong Ma
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, 100191, China.
| | - Lanqun Mao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, The Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
24
|
Fedoce ADG, Ferreira F, Bota RG, Bonet-Costa V, Sun PY, Davies KJA. The role of oxidative stress in anxiety disorder: cause or consequence? Free Radic Res 2018; 52:737-750. [PMID: 29742940 PMCID: PMC6218334 DOI: 10.1080/10715762.2018.1475733] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Anxiety disorders are the most common mental illness in the USA affecting 18% of the population. The cause(s) of anxiety disorders is/are not completely clear, and research in the neurobiology of anxiety at the molecular level is still rather limited. Although mounting clinical and preclinical evidence now indicates that oxidative stress may be a major component of anxiety pathology, whether oxidative stress is the cause or consequence remains elusive. Studies conducted over the past few years suggest that anxiety disorders may be characterised by lowered antioxidant defences and increased oxidative damage to proteins, lipids, and nucleic acids. In particular, oxidative modifications to proteins have actually been proposed as a potential factor in the onset and progression of several psychiatric disorders, including anxiety and depressive disorders. Oxidised proteins are normally degraded by the proteasome proteolytic complex in the cell cytoplasm, nucleus, and endoplasmic reticulum. The Lon protease performs a similar protective function inside mitochondria. Impairment of the proteasome and/or the Lon protease results in the accumulation of toxic oxidised proteins in the brain, which can cause severe neuronal trauma. Recent evidence points to possible proteolytic dysfunction and accumulation of damaged, oxidised proteins as factors that may determine the appearance and severity of psychotic symptoms in mood disorders. Thus, critical interactions between oxidative stress, proteasome, and the Lon protease may provide keys to the molecular mechanisms involved in emotional regulation, and may also be of great help in designing and screening novel anxiolytics and antidepressants.
Collapse
Affiliation(s)
- Alessandra das Graças Fedoce
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, The University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Frederico Ferreira
- Oswaldo Cruz Foundation, Oswaldo Cruz Institute, Laboratory on Thymus Research, Rio de Janeiro, Brazil
| | - Robert G. Bota
- Department of Psychiatry, University of California, Irvine, Orange, CA 92868
| | - Vicent Bonet-Costa
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, The University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Patrick Y. Sun
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, The University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Kelvin J. A. Davies
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, The University of Southern California, Los Angeles, CA 90089-0191, USA
- Division of Molecular & Computational Biology, Department of Biological Sciences, Dornsife College of Letters, Arts, & Sciences, The University of Southern California, Los Angeles, CA 90089-0191, USA
| |
Collapse
|
25
|
Saffarpour S, Nasirinezhad F. Functional interaction between N-methyl-D-aspartate receptor and ascorbic acid during neuropathic pain induced by chronic constriction injury of the sciatic nerve. J Basic Clin Physiol Pharmacol 2018; 28:601-608. [PMID: 28902622 DOI: 10.1515/jbcpp-2017-0015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 06/29/2017] [Indexed: 01/10/2023]
Abstract
BACKGROUND Neuropathic pain is a chronic pain condition, which is resistant to therapy. Ascorbate was released because of the activation of glutaminergic neurons. Due to the important role of N-methyl-D-aspartate (NMDA) receptors in the pathophysiology of neuropathic pain, this study investigated the analgesic efficacy of ascorbic acid (AA) in neuropathic pain condition and the role of NMDA receptors in this effect. METHODS For this purpose, adult male rats were randomly allocated to experimental groups (n=8 in each group). Neuropathic pain was induced by chronic constriction injury (CCI) of the sciatic nerve. During the second week after CCI, animals received a single injection of 1, 3, 5, or 10 mg/kg of AA intraperitoneally and pain threshold was determined 15 and 60 min later. The antinociceptive effect of chronic administration was also evaluated by intraperitoneal injection (IP) of 3 mg/kg AA for 3 weeks. To determine the role of NMDA receptors, separate groups of animals 30 min after single injection of AA (1 mg/kg) animals received i.p. injection of ketamine (5 mg/kg), MK-801 (0.01 mg/kg), or glutamate (1000 nmol) and were tested 20 min afterwards. Data analyzed by ANOVA and Newman-Keuls tests and p<0.05 were considered as significant. RESULTS IP of 3, 5 and 10 mg/kg increased the pain threshold during the second week after CCI (p<0.05, F=3 in tactile allodynia and p<0.01, F=3.2 in thermal and mechanical hyperalgeisa). Chronic administration of AA also produced antinociceptive effect. Ascorbic acid (1 mg/kg, i.p.) inhibited MK-801 and ketamine-induced antinociception response significantly (p<0.001, F=2). It also prevented the analgesic effect of glutamate administration (p<0.001, F=2). CONCLUSIONS The results indicated that AA produced a dose-dependent antinociceptive effect that seems to mediate through its interaction with NMDA receptors.
Collapse
|
26
|
Bansal R, Singh R. Exploring the potential of natural and synthetic neuroprotective steroids against neurodegenerative disorders: A literature review. Med Res Rev 2017; 38:1126-1158. [PMID: 28697282 DOI: 10.1002/med.21458] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/01/2017] [Accepted: 06/20/2017] [Indexed: 12/18/2022]
Abstract
Neurodegeneration is a complex process, which leads to progressive brain damage due to loss of neurons. Despite exhaustive research, the cause of neuronal loss in various degenerative disorders is not entirely understood. Neuroprotective steroids constitute an important line of attack, which could play a major role against the common mechanisms associated with various neurodegenerative disorders like Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Natural endogenous steroids induce the neuroprotection by protecting the nerve cells from neuronal injury through multiple mechanisms, therefore the structural modifications of the endogenous steroids could be helpful in the generation of new therapeutically useful neuroprotective agents. The review article will keep the readers apprised of the detailed description of natural as well as synthetic neuroprotective steroids from the medicinal chemistry point of view, which would be helpful in drug discovery efforts aimed toward neurodegenerative diseases.
Collapse
Affiliation(s)
- Ranju Bansal
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Ranjit Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
27
|
Mortensen A, Aguilar F, Crebelli R, Di Domenico A, Dusemund B, Frutos MJ, Galtier P, Gott D, Gundert-Remy U, Leblanc JC, Lindtner O, Moldeus P, Mosesso P, Parent-Massin D, Oskarsson A, Stankovic I, Waalkens-Berendsen I, Woutersen RA, Wright M, Younes M, Boon P, Chrysafidis D, Gürtler R, Tobback P, Altieri A, Rincon AM, Lambré C. Re-evaluation of glutamic acid (E 620), sodium glutamate (E 621), potassium glutamate (E 622), calcium glutamate (E 623), ammonium glutamate (E 624) and magnesium glutamate (E 625) as food additives. EFSA J 2017; 15:e04910. [PMID: 32625571 PMCID: PMC7009848 DOI: 10.2903/j.efsa.2017.4910] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The EFSA Panel on Food Additives and Nutrient Sources added to Food (ANS) provides a scientific opinion re-evaluating the safety of glutamic acid-glutamates (E 620-625) when used as food additives. Glutamate is absorbed in the intestine and it is presystemically metabolised in the gut wall. No adverse effects were observed in the available short-term, subchronic, chronic, reproductive and developmental studies. The only effect observed was increased kidney weight and increased spleen weight; however, the increase in organ weight was not accompanied by adverse histopathological findings and, therefore, the increase in organ weight was not considered as an adverse effect. The Panel considered that glutamic acid-glutamates (E 620-625) did not raise concern with regards to genotoxicity. From a neurodevelopmental toxicity study, a no observed adverse effect level (NOAEL) of 3,200 mg monosodium glutamate/kg body weight (bw) per day could be identified. The Panel assessed the suitability of human data to be used for the derivation of a health-based guidance value. Although effects on humans were identified human data were not suitable due to the lack of dose-response data from which a dose without effect could be identified. Based on the NOAEL of 3,200 mg monosodium glutamate/kg bw per day from the neurodevelopmental toxicity study and applying the default uncertainty factor of 100, the Panel derived a group acceptable daily intake (ADI) of 30 mg/kg bw per day, expressed as glutamic acid, for glutamic acid and glutamates (E 620-625). The Panel noted that the exposure to glutamic acid and glutamates (E 620-625) exceeded not only the proposed ADI, but also doses associated with adverse effects in humans for some population groups.
Collapse
|
28
|
Kocot J, Luchowska-Kocot D, Kiełczykowska M, Musik I, Kurzepa J. Does Vitamin C Influence Neurodegenerative Diseases and Psychiatric Disorders? Nutrients 2017; 9:E659. [PMID: 28654017 PMCID: PMC5537779 DOI: 10.3390/nu9070659] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/15/2017] [Accepted: 06/21/2017] [Indexed: 02/07/2023] Open
Abstract
Vitamin C (Vit C) is considered to be a vital antioxidant molecule in the brain. Intracellular Vit C helps maintain integrity and function of several processes in the central nervous system (CNS), including neuronal maturation and differentiation, myelin formation, synthesis of catecholamine, modulation of neurotransmission and antioxidant protection. The importance of Vit C for CNS function has been proven by the fact that targeted deletion of the sodium-vitamin C co-transporter in mice results in widespread cerebral hemorrhage and death on post-natal day one. Since neurological diseases are characterized by increased free radical generation and the highest concentrations of Vit C in the body are found in the brain and neuroendocrine tissues, it is suggested that Vit C may change the course of neurological diseases and display potential therapeutic roles. The aim of this review is to update the current state of knowledge of the role of vitamin C on neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis and amyotrophic sclerosis, as well as psychiatric disorders including depression, anxiety and schizophrenia. The particular attention is attributed to understanding of the mechanisms underlying possible therapeutic properties of ascorbic acid in the presented disorders.
Collapse
Affiliation(s)
- Joanna Kocot
- Chair and Department of Medical Chemistry, Medical University of Lublin, 4A Chodźki Street, 20-093 Lublin, Poland.
| | - Dorota Luchowska-Kocot
- Chair and Department of Medical Chemistry, Medical University of Lublin, 4A Chodźki Street, 20-093 Lublin, Poland.
| | - Małgorzata Kiełczykowska
- Chair and Department of Medical Chemistry, Medical University of Lublin, 4A Chodźki Street, 20-093 Lublin, Poland.
| | - Irena Musik
- Chair and Department of Medical Chemistry, Medical University of Lublin, 4A Chodźki Street, 20-093 Lublin, Poland.
| | - Jacek Kurzepa
- Chair and Department of Medical Chemistry, Medical University of Lublin, 4A Chodźki Street, 20-093 Lublin, Poland.
| |
Collapse
|
29
|
17β-Estradiol via SIRT1/Acetyl-p53/NF-kB Signaling Pathway Rescued Postnatal Rat Brain Against Acute Ethanol Intoxication. Mol Neurobiol 2017; 55:3067-3078. [PMID: 28466267 DOI: 10.1007/s12035-017-0520-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 04/06/2017] [Indexed: 10/19/2022]
Abstract
Growing evidences reveal that 17β-estradiol has a wide variety of neuroprotective potential. Recently, it has been shown that 17β-estradiol can limit ethanol-induced neurotoxicity in neonatal rats. Whether it can stimulate SIRT1 signaling against ethanol intoxicity in developing brain remain elusive. Here, we report for the first time that 17β-estradiol activated SIRT1 to deacetylate p53 proteins against acute ethanol-induced oxidative stress, neuroinflammation, and neurodegeneration. A single subcutaneous injection of ethanol-induced oxidative stress triggered phospho c-jun N terminal kinase (p-JNK) and phospho mammalian target of rapamycin (p-mTOR) accompanied by neuroinflammation and widespread neurodegeneration. In contrast, 17β-estradiol cotreatment positively regulated SIRT1, inhibited p53 acetylation, reactive oxygen species (ROS) production, p-JNK, and p-mTOR activation and reduced neuroinflammation and neuronal cell death in the postnatal rat brain. Interestingly, SIRT1 inhibition with its inhibitor, i.e., EX527 further enhanced ethanol intoxication and also abolished the beneficial effects of 17β-estradiol against ethanol in the young rat's brain. Indeed, 17β-estradiol treatment increased the cell viability (HT22 cells), inhibited ROS production via the SIRT1/Acetyl-p53 pathway, and reduced the nuclear translocation of phospho-nuclear factor kappa B (p-NF-kB) in the BV2 microglia cells. Taken together, these results show that 17β-estradiol can be used as a potential neuroprotective agent against acute ethanol intoxication.
Collapse
|
30
|
Abstract
Disturbance of cerebral redox homeostasis is the primary cause of human neurodegenerative disorders, such as Parkinson's disease or Alzheimer's disease. Well known experimental research demonstrates that oxidative stress is a main cause of cell death. A high concentration of reactive oxygen and nitrogen species leads to damage of a lot of proteins, lipids and also DNA. Synthetic compounds used for the treatment in the neurodegenerative diseases failed to meet the hopes they had raised and often exhibit a number of side effects. Therefore, in recent years interest in natural compounds derived from plants appears to be on the rise. This review describes a few natural compounds (1MeTIQ, resveratrol, curcumin, vitamin C and Gingko biloba) which revealed neuroprotective potential both in experimental studies and clinical trials. 1MeTIQ has a privileged position because, as opposed to the remaining compounds, it is an endogenous amine synthesized in human and animal brain. Based on evidence from research, it seems that a common protective mechanism for all the above-mentioned natural compounds relies on their ability to inhibit or even scavenge the excess of free radicals generated in oxidative and neurotoxin-induced processes in nerve cells of the brain. However, it was demonstrated that further different molecular processes connected with neurotoxicity (e.g. the inhibition of mitochondrial complex I, activation of caspase-3, apoptosis) follow later and are initiated by the reactive oxygen species. What is more, these natural compounds are able to inhibit further stages of apoptosis triggered by neurotoxins in the brain.
Collapse
Affiliation(s)
- Agnieszka Wąsik
- Institute of Pharmacology, Polish Academy of Sciences, Department of Neurochemistry, Kraków, Poland.
| | | |
Collapse
|
31
|
Foran L, Blackburn K, Kulesza RJ. Auditory hindbrain atrophy and anomalous calcium binding protein expression after neonatal exposure to monosodium glutamate. Neuroscience 2017; 344:406-417. [DOI: 10.1016/j.neuroscience.2017.01.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 12/31/2016] [Accepted: 01/03/2017] [Indexed: 01/29/2023]
|
32
|
Shah SA, Yoon GH, Chung SS, Abid MN, Kim TH, Lee HY, Kim MO. Novel osmotin inhibits SREBP2 via the AdipoR1/AMPK/SIRT1 pathway to improve Alzheimer's disease neuropathological deficits. Mol Psychiatry 2017; 22:407-416. [PMID: 27001618 PMCID: PMC5322276 DOI: 10.1038/mp.2016.23] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 02/02/2016] [Accepted: 02/04/2016] [Indexed: 12/22/2022]
Abstract
Extensive evidence has indicated that a high rate of cholesterol biogenesis and abnormal neuronal energy metabolism play key roles in Alzheimer's disease (AD) pathogenesis. Here, for we believe the first time, we used osmotin, a plant protein homolog of mammalian adiponectin, to determine its therapeutic efficacy in different AD models. Our results reveal that osmotin treatment modulated adiponectin receptor 1 (AdipoR1), significantly induced AMP-activated protein kinase (AMPK)/Sirtuin 1 (SIRT1) activation and reduced SREBP2 (sterol regulatory element-binding protein 2) expression in both in vitro and in vivo AD models and in Adipo-/- mice. Via the AdipoR1/AMPK/SIRT1/SREBP2 signaling pathway, osmotin significantly diminished amyloidogenic Aβ production, abundance and aggregation, accompanied by improved pre- and post-synaptic dysfunction, cognitive impairment, memory deficits and, most importantly, reversed the suppression of long-term potentiation in AD mice. Interestingly, AdipoR1, AMPK and SIRT1 silencing not only abolished osmotin capability but also further enhanced AD pathology by increasing SREBP2, amyloid precursor protein (APP) and β-secretase (BACE1) expression and the levels of toxic Aβ production. However, the opposite was true for SREBP2 when silenced using small interfering RNA in APPswe/ind-transfected SH-SY5Y cells. Similarly, osmotin treatment also enhanced the non-amyloidogenic pathway by activating the α-secretase gene that is, ADAM10, in an AMPK/SIRT1-dependent manner. These results suggest that osmotin or osmotin-based therapeutic agents might be potential candidates for AD treatment.
Collapse
Affiliation(s)
- S A Shah
- Division of Life Science and Applied Life Science (BK21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - G H Yoon
- Division of Life Science and Applied Life Science (BK21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - S S Chung
- Department of Physiology, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - M N Abid
- Division of Life Science and Applied Life Science (BK21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - T H Kim
- Division of Life Science and Applied Life Science (BK21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - H Y Lee
- Division of Life Science and Applied Life Science (BK21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - M O Kim
- Division of Life Science and Applied Life Science (BK21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea,Neuroscience Pioneer Research Center, Department of Biology, College of Natural Sciences, Gyeongsang National University, Jinju 660-701, Republic of Korea. E-mail:
| |
Collapse
|
33
|
Shah SA, Amin FU, Khan M, Abid MN, Rehman SU, Kim TH, Kim MW, Kim MO. Anthocyanins abrogate glutamate-induced AMPK activation, oxidative stress, neuroinflammation, and neurodegeneration in postnatal rat brain. J Neuroinflammation 2016; 13:286. [PMID: 27821173 PMCID: PMC5100309 DOI: 10.1186/s12974-016-0752-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/26/2016] [Indexed: 12/20/2022] Open
Abstract
Background Glutamate-induced excitotoxicity, oxidative damage, and neuroinflammation are believed to play an important role in the development of a number of CNS disorders. We recently reported that a high dose of glutamate could induce AMPK-mediated neurodegeneration in the postnatal day 7 (PND7) rat brain. Yet, the mechanism of glutamate-induced oxidative stress and neuroinflammation in the postnatal brain is not well understood. Here, we report for the first time the mechanism of glutamate-induced oxidative damage, neuroinflammation, and neuroprotection by polyphenolic anthocyanins in PND7. Methods PND7 rat brains, SH-SY5Y, and BV2 cells treated either alone with glutamate or in combination with anthocyanins and compound C were examined with Western blot and immunofluorescence techniques. Additionally, reactive oxygen species (ROS) assay and other ELISA kit assays were employed to know the therapeutic efficacy of anthocyanins against glutamate. Results A single injection of glutamate to developing rats significantly increased brain glutamate levels, activated and phosphorylated AMPK induction, and inhibited nuclear factor-E2-related factor 2 (Nrf2) after 2, 3, and 4 h in a time-dependent manner. In contrast, anthocyanin co-treatment significantly reduced glutamate-induced AMPK induction, ROS production, neuroinflammation, and neurodegeneration in the developing rat brain. Most importantly, anthocyanins increased glutathione (GSH and GSSG) levels and stimulated the endogenous antioxidant system, including Nrf2 and heme oxygenase-1 (HO-1), against glutamate-induced oxidative stress. Interestingly, blocking AMPK with compound C in young rats abolished glutamate-induced neurotoxicity. Similarly, all these experiments were replicated in SH-SY5Y cells by silencing AMPK with siRNA, which suggests that AMPK is the key mediator in glutamate-induced neurotoxicity. Conclusions Here, we report for the first time that anthocyanins can potentially decrease glutamate-induced neurotoxicity in young rats. Our work demonstrates that glutamate is toxic to the developing rat brain and that anthocyanins can minimize the severity of glutamate-induced neurotoxicity in an AMPK-dependent manner. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0752-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shahid Ali Shah
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Faiz Ul Amin
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Mehtab Khan
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Muhammad Noman Abid
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Shafiq Ur Rehman
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Tae Hyun Kim
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Min Woo Kim
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Myeong Ok Kim
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea.
| |
Collapse
|
34
|
Moosavirad SA, Rabbani M, Sharifzadeh M, Hosseini-Sharifabad A. Protective effect of vitamin C, vitamin B12 and omega-3 on lead-induced memory impairment in rat. Res Pharm Sci 2016; 11:390-396. [PMID: 27920821 PMCID: PMC5122828 DOI: 10.4103/1735-5362.192490] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Lead belongs to the heavy metal group and is considered as an environmental contaminant. Acute or chronic contact to lead can change the physiological function of human organs. One of the most important disorders following the lead exposure is neurotoxicity. Lead neurotoxicity consists of the neurobehavioral disturbances like cognitive impairment. The aim of the current study is to evaluate the possible protective effect of vitamin C (Vit C), vitamin B12 (Vit B12), omega 3 (ω-3), or their combination on the lead-induced memory disorder. Adult wistar rats were orally administered Vit C (120 mg/kg/day) or Vit B12 (1 mg/kg/day) or ω-3 (1000 mg/kg/day) or their combination for 3 weeks in groups of 7 animals each. Then lead acetate (15 mg/kg/day) was injected intraperitoneally for one week to all pretreated animals. The control group received normal saline as a vehicle while the positive control for cognitive impairment received just lead acetate. At the end of treatments animal memories were evaluated in Object Recognition Task. The results showed, although 15 mg/kg lead acetate significantly declines the memory-evaluating parameters, pretreatment with Vit C, Vit B12, ω-3, or their combination considerably inverted the lead induced reduction in discrimination (d2) index (P < 0.001) and recognition (R) index (P < 0.001, P < 0.05, P < 0.05, and P < 0.001, respectively). Our findings indicate while lead acetate impairs spatial memory in rat, administration of Vit C, Vit B12, ω-3, or their combination prior to the lead exposure inhibits the lead induced cognitive loss. There was no remarkable difference in this effect between the used supplements.
Collapse
Affiliation(s)
- Saeedeh Alsadat Moosavirad
- Department of Pharmacology and Toxicology, Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mohammad Rabbani
- Department of Pharmacology and Toxicology, Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mohammad Sharifzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, I.R. Iran
| | - Ali Hosseini-Sharifabad
- Department of Pharmacology and Toxicology, Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
35
|
Dual Role of Vitamin C on the Neuroinflammation Mediated Neurodegeneration and Memory Impairments in Colchicine Induced Rat Model of Alzheimer Disease. J Mol Neurosci 2016; 60:421-435. [PMID: 27665568 DOI: 10.1007/s12031-016-0817-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 08/10/2016] [Indexed: 12/22/2022]
Abstract
The neurodegeneration in colchicine induced AD rats (cAD) is mediated by cox-2 linked neuroinflammation. The importance of ROS in the inflammatory process in cAD has not been identified, which may be deciphered by blocking oxidative stress in this model by a well-known anti-oxidant vitamin C. Therefore, the present study was designed to investigate the role of vitamin C on colchicine induced oxidative stress linked neuroinflammation mediated neurodegeneration and memory impairments along with peripheral immune responses in cAD. The impairments of working and reference memory were associated with neuroinflammation and neurodegeneration in the hippocampus of cAD. Administration of vitamin C (200 and 400 mg/kg BW) in cAD resulted in recovery of memory impairments, with prevention of neurodegeneration and neuroinflammation in the hippocampus. The neuroinflammation in the hippocampus also influenced the peripheral immune responses and inflammation in the serum of cAD and all of these parameters were also recovered at 200 and 400 mg dose of vitamin C. However, cAD treated with 600 mg dose did not recover but resulted in increase of memory impairments, neurodegeneration and neuroinflammation in hippocampus along with alteration of peripheral immune responses in comparison to cAD of the present study. Therefore, the present study showed that ROS played an important role in the colchicine induced neuroinflammation linked neurodegeneration and memory impairments along with alteration of peripheral immune responses. It also appears from the results that vitamin C at lower doses showed anti-oxidant effect and at higher dose resulted in pro-oxidant effects in cAD.
Collapse
|
36
|
Shah SA, Khan M, Jo MH, Jo MG, Amin FU, Kim MO. Melatonin Stimulates the SIRT1/Nrf2 Signaling Pathway Counteracting Lipopolysaccharide (LPS)-Induced Oxidative Stress to Rescue Postnatal Rat Brain. CNS Neurosci Ther 2016; 23:33-44. [PMID: 27421686 DOI: 10.1111/cns.12588] [Citation(s) in RCA: 195] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 12/12/2022] Open
Abstract
AIMS Lipopolysaccharide (LPS) induces oxidative stress and neuroinflammation both in vivo and in vitro. Here, we provided the first detailed description of the mechanism of melatonin neuroprotection against LPS-induced oxidative stress, acute neuroinflammation, and neurodegeneration in the hippocampal dentate gyrus (DG) region of the postnatal day 7 (PND7) rat brain. METHODS The neuroprotective effects of melatonin against LPS-induced neurotoxicity were analyzed using multiple research techniques, including Western blotting, immunofluorescence, and enzyme-linked immunosorbent assays (ELISAs) in PND7 rat brain homogenates and BV2 cell lysates in vitro. We also used EX527 to inhibit silent information regulator transcript-1 (SIRT1). RESULTS A single intraperitoneal (i.p) injection of LPS to PND7 rats significantly induced glial cell activation, acute neuroinflammation, reactive oxygen species (ROS) production and apoptotic neurodegeneration in hippocampal DG region after 4 h. However, the coadministration of melatonin significantly inhibited both LPS-induced acute neuroinflammation and apoptotic neurodegeneration and improved synaptic dysfunction in the hippocampal DG region of PND7 rats. Most importantly, melatonin stimulated the SIRT1/Nrf2 (nuclear factor-erythroid 2-related factor 2) signaling pathway to reduce LPS-induced ROS generation. The beneficial effects of melatonin were further confirmed in LPS-stimulated BV2 microglia cell lines in vitro using EX527 as an inhibitor of SIRT1. LPS-induced oxidative stress, Nrf2 inhibition, and neuroinflammation are SIRT1-dependent in BV2 microglia cell lines. CONCLUSION These results demonstrated that melatonin treatment rescued the hippocampal DG region of PND7 rat brains against LPS-induced oxidative stress damage, acute neuroinflammation, and apoptotic neurodegeneration via SIRT1/Nrf2 signaling pathway activation.
Collapse
Affiliation(s)
- Shahid Ali Shah
- Department of Biology and Applied Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Mehtab Khan
- Department of Biology and Applied Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Myeung-Hoon Jo
- Department of Biology and Applied Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Min Gi Jo
- Department of Biology and Applied Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Faiz Ul Amin
- Department of Biology and Applied Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Myeong Ok Kim
- Department of Biology and Applied Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
37
|
Amin FU, Shah SA, Kim MO. Glycine inhibits ethanol-induced oxidative stress, neuroinflammation and apoptotic neurodegeneration in postnatal rat brain. Neurochem Int 2016; 96:1-12. [DOI: 10.1016/j.neuint.2016.04.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 03/31/2016] [Accepted: 04/01/2016] [Indexed: 12/22/2022]
|
38
|
The gene BRAF is underexpressed in bipolar subject olfactory neuroepithelial progenitor cells undergoing apoptosis. Psychiatry Res 2016; 236:130-135. [PMID: 26753950 DOI: 10.1016/j.psychres.2015.12.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 09/30/2015] [Accepted: 12/15/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND Bipolar disorder is a devastating psychiatric condition that frequently results in various degrees of brain tissue loss, cognitive decline, and premature death. The documentation of brain tissue loss implicates apoptosis as the likely underlying degenerative process, but direct experimental demonstration is lacking. METHODS Olfactory neuroepithelial biopsies from individuals with and without bipolar I disorder yielded olfactory neuroepithelial progenitor cells (ONPs), which spontaneously differentiate into neurons and glia. Glutamate, 0.1M, for 3 and 6h was used to induce apoptosis. Genes involved in the apoptotic pathway were interrogated with micro-array analysis before and after glutamate treatment for 6h. Confirmation was accomplished with real-time PCR. Total and phospho-B-Raf protein levels were measured using Western blot analysis. RESULTS ONPs from bipolar individuals demonstrated significantly greater apoptosis than cells from non-bipolar subjects. Microarray results revealed 12 differentially expressed genes. Five genes were further examined. BRAF mRNA and protein levels were significantly reduced in bipolar ONPs. CONCLUSIONS ONPs with the genetic heritage of bipolar I disorder were more sensitive to glutamate induced apoptosis. Under expression of the BRAF gene and protein, which plays a role in regulating the pro-survival MEK/ERK signaling pathway, may contribute to this apoptotic sensitivity.
Collapse
|
39
|
Shah SA, Yoon GH, Ahmad A, Ullah F, Ul Amin F, Kim MO. Nanoscale-alumina induces oxidative stress and accelerates amyloid beta (Aβ) production in ICR female mice. NANOSCALE 2015; 7:15225-37. [PMID: 26315713 DOI: 10.1039/c5nr03598h] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The adverse effects of nanoscale-alumina (Al2O3-NPs) have been previously demonstrated in both in vitro and in vivo studies, whereas little is known about their mechanism of neurotoxicity. It is the goal of this research to determine the toxic effects of nano-alumina on human neuroblastoma SH-SY5Y and mouse hippocampal HT22 cells in vitro and on ICR female mice in vivo. Nano-alumina displayed toxic effects on SH-SY5Y cell lines in three different concentrations also increased aluminium abundance and induced oxidative stress in HT22 cells. Nano-alumina peripherally administered to ICR female mice for three weeks increased brain aluminium and ROS production, disturbing brain energy homeostasis, and led to the impairment of hippocampus-dependent memory. Most importantly, these nano-particles induced Alzheimer disease (AD) neuropathology by enhancing the amyloidogenic pathway of Amyloid Beta (Aβ) production, aggregation and implied the progression of neurodegeneration in the cortex and hippocampus of these mice. In conclusion, these data demonstrate that nano-alumina is toxic to both cells and female mice and that prolonged exposure may heighten the chances of developing a neurodegenerative disease, such as AD.
Collapse
Affiliation(s)
- Shahid Ali Shah
- Department of Biology and Applied Life Science (BK 21), College of Natural Sciences (RINS), Gyeongsang National University, Jinju, 660-701, Republic of Korea.
| | | | | | | | | | | |
Collapse
|