1
|
Paslı D, Gürbay A. Assessment of Protective Effects of DTPA, NAC, and Taurine on Possible Cytotoxicity Induced by Individual and Combined Zinc Oxide and Copper Oxide Nanoparticles in SH-SY5Y Cells. Biol Trace Elem Res 2024:10.1007/s12011-024-04161-0. [PMID: 38683268 DOI: 10.1007/s12011-024-04161-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 03/25/2024] [Indexed: 05/01/2024]
Abstract
The present study investigated the cytotoxic effects of ZnO, CuO, and mixed combinations of them on SH-SY5Y cells. For this purpose, the cells were exposed to various concentrations of these NPs alone for 24-96 h and as a mixture for 24 h. Variations in cell viability were noted. MTT results showed that ZnO and/or CuO NPs decreased cell survival by about 59% at 200 (ZnO, at 24 h) and 800 µg/ml (ZnO and/or CuO, at 72 and 96 h). When the NR assay was used, slight decreases were noted with ZnO NPs at 72 and 96 h. With CuO NPs alone and NPs in a mixture, only the highest concentrations caused 40 and 70% decreases in cell survival, respectively. Especially with NR assays, DTPA, NAC, or taurine provided marked protection. ROS levels were increased with the highest concentration of CuO NPs and with all concentrations of the mixture. The highest concentration of ZnO NPs and the lowest concentration of CuO NPs caused slight decreases in mitochondrial membrane potential levels. Additionally, increases were noted in caspase 3/7 levels with ZnO and CuO NPs alone or with a mixture of them. Intracellular calcium levels were decreased in this system. These findings demonstrated that ZnO and CuO NPs, either separately or in combination, had a modest cytotoxic effect on SH-SY5Y cells. Protection obtained with DTPA, NAC, or taurine against the cytotoxicity of these NPs and the ROS-inducing effect of CuO NPs and the NPs' mixture suggests that oxidative stress might be involved in the cytotoxicity mechanisms of these NPs.
Collapse
Affiliation(s)
- Duygu Paslı
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, 06100, Ankara, Turkey
| | - Aylin Gürbay
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, 06100, Ankara, Turkey.
| |
Collapse
|
2
|
Corona-Trejo A, Gonsebatt ME, Trejo-Solis C, Campos-Peña V, Quintas-Granados LI, Villegas-Vázquez EY, Daniel Reyes-Hernández O, Hernández-Abad VJ, Figueroa-González G, Silva-Adaya D. Transsulfuration pathway: a targeting neuromodulator in Parkinson's disease. Rev Neurosci 2023; 34:915-932. [PMID: 37409540 DOI: 10.1515/revneuro-2023-0039] [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: 03/31/2023] [Accepted: 06/04/2023] [Indexed: 07/07/2023]
Abstract
The transsulfuration pathway (TSP) is a metabolic pathway involving sulfur transfer from homocysteine to cysteine. Transsulfuration pathway leads to many sulfur metabolites, principally glutathione, H2S, taurine, and cysteine. Key enzymes of the TSP, such as cystathionine β-synthase and cystathionine γ-lyase, are essential regulators at multiple levels in this pathway. TSP metabolites are implicated in many physiological processes in the central nervous system and other tissues. TSP is important in controlling sulfur balance and optimal cellular functions such as glutathione synthesis. Alterations in the TSP and related pathways (transmethylation and remethylation) are altered in several neurodegenerative diseases, including Parkinson's disease, suggesting their participation in the pathophysiology and progression of these diseases. In Parkinson's disease many cellular processes are comprised mainly those that regulate redox homeostasis, inflammation, reticulum endoplasmic stress, mitochondrial function, oxidative stress, and sulfur content metabolites of TSP are involved in these damage processes. Current research on the transsulfuration pathway in Parkinson's disease has primarily focused on the synthesis and function of certain metabolites, particularly glutathione. However, our understanding of the regulation of other metabolites of the transsulfuration pathway, as well as their relationships with other metabolites, and their synthesis regulation in Parkinson´s disease remain limited. Thus, this paper highlights the importance of studying the molecular dynamics in different metabolites and enzymes that affect the transsulfuration in Parkinson's disease.
Collapse
Affiliation(s)
- Andrea Corona-Trejo
- Carrera de Biología, Laboratorio de Farmacogenética, Unidad Multidisciplinaria de Investigación Experimental Zaragoza, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico
| | - María E Gonsebatt
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Cristina Trejo-Solis
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, Mexico, 14269, Mexico
| | - Victoria Campos-Peña
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, Mexico, 14269, Mexico
| | | | - Edgar Yebrán Villegas-Vázquez
- Laboratorio de Farmacogenética, Unidad Multidisciplinaria de Investigación Experimental Zaragoza, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, 09230 Mexico City, Mexico
| | - Octavio Daniel Reyes-Hernández
- Laboratorio de Biología Molecular del Cáncer, Unidad Multidisciplinaria de Investigación Experimental Zaragoza, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico
| | - Vicente Jesús Hernández-Abad
- Laboratorio de Investigación Farmacéutica, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Batalla de 5 de mayo s/n, Col, Ejército de Oriente, 09230 Mexico City, Mexico
| | - Gabriela Figueroa-González
- Laboratorio de Farmacogenética, Unidad Multidisciplinaria de Investigación Experimental Zaragoza, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, 09230 Mexico City, Mexico
| | - Daniela Silva-Adaya
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, Mexico, 14269, Mexico
| |
Collapse
|
3
|
Hoffman SS, Liang D, Hood RB, Tan Y, Terrell ML, Marder ME, Barton H, Pearson MA, Walker DI, Barr DB, Jones DP, Marcus M. Assessing Metabolic Differences Associated with Exposure to Polybrominated Biphenyl and Polychlorinated Biphenyls in the Michigan PBB Registry. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:107005. [PMID: 37815925 PMCID: PMC10564108 DOI: 10.1289/ehp12657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND Polybrominated biphenyls (PBB) and polychlorinated biphenyls (PCB) are persistent organic pollutants with potential endocrine-disrupting effects linked to adverse health outcomes. OBJECTIVES In this study, we utilize high-resolution metabolomics (HRM) to identify internal exposure and biological responses underlying PCB and multigenerational PBB exposure for participants enrolled in the Michigan PBB Registry. METHODS HRM profiling was conducted on plasma samples collected from 2013 to 2014 from a subset of participants enrolled in the Michigan PBB Registry, including 369 directly exposed individuals (F0) who were alive when PBB mixtures were accidentally introduced into the food chain and 129 participants exposed to PBB in utero or through breastfeeding, if applicable (F1). Metabolome-wide association studies were performed for PBB-153 separately for each generation and Σ PCB (PCB-118, PCB-138, PCB-153, and PCB-180) in the two generations combined, as both had direct PCB exposure. Metabolite and metabolic pathway alterations were evaluated following a well-established untargeted HRM workflow. RESULTS Mean levels were 1.75 ng / mL [standard deviation (SD): 13.9] for PBB-153 and 1.04 ng / mL (SD: 0.788) for Σ PCB . Sixty-two and 26 metabolic features were significantly associated with PBB-153 in F0 and F1 [false discovery rate (FDR) p < 0.2 ], respectively. There were 2,861 features associated with Σ PCB (FDR p < 0.2 ). Metabolic pathway enrichment analysis using a bioinformatics tool revealed perturbations associated with Σ PCB in numerous oxidative stress and inflammation pathways (e.g., carnitine shuttle, glycosphingolipid, and vitamin B9 metabolism). Metabolic perturbations associated with PBB-153 in F0 were related to oxidative stress (e.g., pentose phosphate and vitamin C metabolism) and in F1 were related to energy production (e.g., pyrimidine, amino sugars, and lysine metabolism). Using authentic chemical standards, we confirmed the chemical identity of 29 metabolites associated with Σ PCB levels (level 1 evidence). CONCLUSIONS Our results demonstrate that serum PBB-153 is associated with alterations in inflammation and oxidative stress-related pathways, which differed when stratified by generation. We also found that Σ PCB was associated with the downregulation of important neurotransmitters, serotonin, and 4-aminobutanoate. These findings provide novel insights for future investigations of molecular mechanisms underlying PBB and PCB exposure on health. https://doi.org/10.1289/EHP12657.
Collapse
Affiliation(s)
- Susan S. Hoffman
- Department of Epidemiology, Emory University, Atlanta, Georgia, USA
| | - Donghai Liang
- Department of Epidemiology, Emory University, Atlanta, Georgia, USA
- Gangarosa Department of Environmental Health, Emory University, Atlanta, Georgia, USA
| | - Robert B. Hood
- Department of Epidemiology, Emory University, Atlanta, Georgia, USA
| | - Youran Tan
- Gangarosa Department of Environmental Health, Emory University, Atlanta, Georgia, USA
| | | | - M. Elizabeth Marder
- Department of Environmental Toxicology, University of California, Davis, Davis, California, USA
| | - Hillary Barton
- Department of Epidemiology, Emory University, Atlanta, Georgia, USA
| | - Melanie A. Pearson
- Gangarosa Department of Environmental Health, Emory University, Atlanta, Georgia, USA
| | - Douglas I. Walker
- Gangarosa Department of Environmental Health, Emory University, Atlanta, Georgia, USA
| | - Dana Boyd Barr
- Gangarosa Department of Environmental Health, Emory University, Atlanta, Georgia, USA
| | - Dean P. Jones
- School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Michele Marcus
- Department of Epidemiology, Emory University, Atlanta, Georgia, USA
- Gangarosa Department of Environmental Health, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
4
|
Shevelev OB, Cherkasova OP, Razumov IA, Zavjalov EL. In vivo MRS study of long-term effects of traumatic intracranial injection of a culture medium in mice. Vavilovskii Zhurnal Genet Selektsii 2023; 27:633-640. [PMID: 38223456 PMCID: PMC10784322 DOI: 10.18699/vjgb-23-74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 02/21/2023] [Accepted: 06/01/2023] [Indexed: 01/16/2024] Open
Abstract
Orthotopic transplantation of glioblastoma cells in the brain of laboratory mice is a common animal model for studying brain tumors. It was shown that 1H magnetic resonance spectroscopy (MRS) enables monitoring of the tumor's occurrence and its development during therapy based on the ratio of several metabolites. However, in studying new approaches to the therapy of glioblastoma in the model of orthotopic xenotransplantation of glioma cells into the brain of mice, it is necessary to understand which metabolites are produced by a growing tumor and which are the result of tumor cells injection along the modeling of the pathology. Currently, there are no data on the dynamic metabolic processes in the brain that occur after the introduction of glioblastoma cells into the brain of mice. In addition, there is a lack of data on the delayed effects of invasive brain damage. Therefore, this study investigates the long-term dynamics of the neurometabolic profile, assessed using 1H MRS, after intracranial injection of a culture medium used in orthotopic modeling of glioma in mice. Levels of N-acetylaspartate, N-acetylaspartylglutamic acid, myoinositol, taurine, glutathione, the sum of glycerophosphocholine and phosphocholine, glutamic acid (Glu), glutamine (Gln), and gamma aminobutyric acid (GABA) indicate patterns of neurometabolites in the early stage after intracranial injection similar to brain trauma ones. Most of the metabolites, with the exception of Gln, Glu and GABA, returned to their original values on day 28 after injection. A progressive increase in the Glu/Gln and Glu/GABA ratio up to 28 days after surgery potentially indicates an impaired turnover of these metabolites or increased neurotransmission. Thus, the data indicate that the recovery processes are largely completed on day 28 after the traumatic event in the brain tissue, leaving open the question of the neurotransmitter system impairment. Consequently, when using animal models of human glioma, researchers should clearly distinguish between which changes in neurometabolites are a response to the injection of cancer cells into the brain, and which processes may indicate the early development of a brain tumor. It is important to keep this in mind when modeling human glioblastoma in mice and monitoring new treatments. In addition, these results may be important in the development of approaches for non-invasive diagnostics of traumatic brain injury as well as recovery and rehabilitation processes of patients after certain brain surgeries.
Collapse
Affiliation(s)
- O B Shevelev
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Institute "International Tomografic Center" of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - O P Cherkasova
- Institute of Laser Physics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State Technical University, Novosibirsk, Russia
| | - I A Razumov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | - E L Zavjalov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
5
|
Kamalmaz N, Ben Bacha A, Alonazi M, Albasher G, Khayyat AIA, El-Ansary A. Unveiling sex-based differences in developing propionic acid-induced features in mice as a rodent model of ASD. PeerJ 2023; 11:e15488. [PMID: 37334116 PMCID: PMC10274690 DOI: 10.7717/peerj.15488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/10/2023] [Indexed: 06/20/2023] Open
Abstract
Background Males are more likely to develop autism as a neurodevelopmental disorder than females are, although the mechanisms underlying male vulnerability are not fully understood. Therefore, studying the role of autism etiologies considering sex differences in the propionic acid (PPA) rodent model of autism would build greater understanding of how females are protected from autism spectrum disorder, which may be used as a treatment strategy for males with autism. Objectives This study aimed to investigate the sex differences in oxidative stress, glutamate excitotoxicity, neuroinflammation, and gut microbiota impairment as etiological mechanisms for many neurological diseases, with specific reference to autism. Method Forty albino mice were divided into four groups of 10 animals each with two control and two treated groups of both sexes received only phosphate-buffered saline or a neurotoxic dose of PPA (250 mg/kg body weight) for 3 days, respectively. Biochemical markers of energy metabolism, oxidative stress, neuroinflammation, and excitotoxicity were measured in mouse brain homogenates, whereas the presence of pathogenic bacteria was assessed in mouse stool samples. Furthermore, the repetitive behavior, cognitive ability, and physical-neural coordination of the animals were examined. Results Collectively, selected variables related to oxidative stress, glutamate excitotoxicity, neuroinflammation, and gut bacteria were impaired concomitantly with altered behavior in PPA-induced rodent model, with males being more susceptible than females. Conclusion This study explains the role of sex in the higher vulnerability of males to develop autistic biochemical and behavioral features compared with females. Female sex hormones and the higher detoxification capacity and higher glycolytic flux in females serve as neuroprotective contributors in a rodent model of autism.
Collapse
Affiliation(s)
- Nasreen Kamalmaz
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Abir Ben Bacha
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Mona Alonazi
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Gadah Albasher
- Zoology Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Arwa Ishaq A. Khayyat
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Research Laboratory, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
6
|
Mohammed AS, Al-Hassani AN, Alrawi RA, Tawfeeq RD. The protective effect of taurine, piracetam and vinpocetine on etoposide-induced inflammation and brain injury in the serum of female albino rats. Ecancermedicalscience 2023; 17:1499. [PMID: 36816786 PMCID: PMC9937074 DOI: 10.3332/ecancer.2023.1499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Indexed: 01/24/2023] Open
Abstract
Etoposide (ETP) is one of the leading antitumour agents in cancer chemotherapy. Many studies have reported on ETP-induced peripheral neuropathy; however, few reports have focused on its brain toxicity. The current research investigates the protective potential of taurine, piracetam and vinpocetine on serum biomarkers associated with inflammation and brain injury induced by ETP in a rodent model. A total of 30 female albino rats were equally divided into five groups; the 1st and 2nd groups were the control and ETP-treated groups, respectively, while the 3rd, 4th and 5th groups were ETP-treated rats cotreated with taurine, piracetam and vinpocetine, respectively. Administration of ETP reduced body weight significantly, enhanced production of serum proinflammatory cytokines including tumour necrosis factor-alpha, interleukin-1 beta (IL-1β) and IL-6 and decreased glutathione serum levels. Moreover, ETP treatment resulted in upregulation of glial fibrillary acidic protein expression and histopathological alterations in the rats' brain compared to the control group. Co-treatment with taurine, piracetam and vinpocetine counteracted ETP-induced brain injury and altered serum biomarkers levels. We concluded that co-treatment with vinpocetine could serve as a complementary therapeutic agent in reducing brain injury and toxicity induced by ETP.
Collapse
Affiliation(s)
- Arwa Salam Mohammed
- Department of Pharmacology and Toxicology, College of Pharmacy, Hawler Medical University, Erbil 44001, Iraq
| | - Ansam N Al-Hassani
- Department of Pharmacology and Toxicology, College of Pharmacy, Hawler Medical University, Erbil 44001, Iraq
| | - Rafal Abdulrazaq Alrawi
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil 44001, Iraq
| | - Rawaz D Tawfeeq
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil 44001, Iraq
| |
Collapse
|
7
|
Ben-Azu B, Adebayo OG, Jarikre TA, Oyovwi MO, Edje KE, Omogbiya IA, Eduviere AT, Moke EG, Chijioke BS, Odili OS, Omondiabge OP, Oyovbaire A, Esuku DT, Ozah EO, Japhet K. Taurine, an essential β-amino acid insulates against ketamine-induced experimental psychosis by enhancement of cholinergic neurotransmission, inhibition of oxidative/nitrergic imbalances, and suppression of COX-2/iNOS immunoreactions in mice. Metab Brain Dis 2022; 37:2807-2826. [PMID: 36057735 DOI: 10.1007/s11011-022-01075-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 08/23/2022] [Indexed: 12/22/2022]
Abstract
Cholinergic, oxidative, nitrergic alterations, and neuroinflammation are some key neuropathological features common in schizophrenia disease. They involve complex biological processes that alter normal behavior. The present treatments used in the management of the disorder remain ineffective together with some serious side effects as one of their setbacks. Taurine is a naturally occurring essential β-amino acid reported to elicit antipsychotic property in first episode psychosis in clinical setting, thus require preclinical investigation. Hence, we set out to investigate the effects of taurine in the prevention and reversal of ketamine-induced psychotic-like behaviors and the associated putative neurobiological mechanisms underlying its effects. Adult male Swiss mice were sheared into three separate cohorts of experiments (n = 7): drug alone, preventive and reversal studies. Treatments consisted of saline (10 mL/kg/p.o./day), taurine (50 and 100 mg/kg/p.o./day) and risperidone (0.5 mg/kg/p.o./day) with concomitant ketamine (20 mg/kg/i.p./day) injections between days 8-14, or 14 days entirely. Behavioral hyperactivity, despair, cognitive impairment, and catalepsy were measured. Brain oxidative/nitrergic imbalance, immunoreactivity (COX-2 and iNOS), and cholinergic markers were determined in the striatum, prefrontal-cortex, and hippocampus. Taurine abates ketamine-mediated psychotic-like episodes without cataleptogenic potential. Taurine attenuated ketamine-induced decrease in glutathione, superoxide-dismutase and catalase levels in the striatum, prefrontal-cortex and hippocampus. Also, taurine prevented and reversed ketamine-mediated elevation of malondialdehyde, nitrite contents, acetylcholinesterase activity, and suppressed COX-2 and iNOS expressions in a brain-region dependent manner. Conclusively, taurine insulates against ketamine-mediated psychotic phenotype by normalizing brain central cholinergic neurotransmissions, oxidative, nitrergic and suppression of immunoreactive proteins in mice brains.
Collapse
Affiliation(s)
- Benneth Ben-Azu
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria.
| | - Olusegun G Adebayo
- Neurophysiology Unit, Department of Physiology, Faculty of Basic Medical Sciences, PAMO University of Medical Sciences, Port-Harcourt, River State, Nigeria
| | - Thiophilus Aghogho Jarikre
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Mega O Oyovwi
- Department of Basic Medical Science, Achievers University, Owo, Ondo State, Nigeria
| | - Kesiena Emmanuel Edje
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Itivere Adrian Omogbiya
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Anthony T Eduviere
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Emuesiri Goodies Moke
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Bienose S Chijioke
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Onyebuchi S Odili
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Osemudiame P Omondiabge
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Aghogho Oyovbaire
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Daniel T Esuku
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Esther O Ozah
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Kelvin Japhet
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| |
Collapse
|
8
|
Nascimento TS, Pinto DV, Dias RP, Raposo RS, Nunes PIG, Roque CR, Santos FA, Andrade GM, Viana JL, Fostier AH, Sussulini A, Alvarez-Leite JI, Fontes-Ribeiro C, Malva JO, Oriá RB. Chronic Methylmercury Intoxication Induces Systemic Inflammation, Behavioral, and Hippocampal Amino Acid Changes in C57BL6J Adult Mice. Int J Mol Sci 2022; 23:13837. [PMID: 36430321 PMCID: PMC9697706 DOI: 10.3390/ijms232213837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 11/12/2022] Open
Abstract
Methylmercury (MeHg) is highly toxic to the human brain. Although much is known about MeHg neurotoxic effects, less is known about how chronic MeHg affects hippocampal amino acids and other neurochemical markers in adult mice. In this study, we evaluated the MeHg effects on systemic lipids and inflammation, hippocampal oxidative stress, amino acid levels, neuroinflammation, and behavior in adult male mice. Challenged mice received MeHg in drinking water (2 mg/L) for 30 days. We assessed weight gain, total plasma cholesterol (TC), triglycerides (TG), endotoxin, and TNF levels. Hippocampal myeloperoxidase (MPO), malondialdehyde (MDA), acetylcholinesterase (AChE), amino acid levels, and cytokine transcripts were evaluated. Mice underwent open field, object recognition, Y, and Barnes maze tests. MeHg-intoxicated mice had higher weight gain and increased the TG and TC plasma levels. Elevated circulating TNF and LPS confirmed systemic inflammation. Higher levels of MPO and MDA and a reduction in IL-4 transcripts were found in the hippocampus. MeHg-intoxication led to increased GABA and glycine, reduced hippocampal taurine levels, delayed acquisition in the Barnes maze, and poor locomotor activity. No significant changes were found in AChE activity and object recognition. Altogether, our findings highlight chronic MeHg-induced effects that may have long-term mental health consequences in prolonged exposed human populations.
Collapse
Affiliation(s)
- Tyciane S. Nascimento
- Neuroscience and Behavior Laboratory, Drug Research and Development Center, Federal University of Ceará, Fortaleza 60430-275, Brazil
| | - Daniel V. Pinto
- Laboratory of Tissue Healing, Ontogeny, and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil
| | - Ronaldo P. Dias
- Laboratory of Tissue Healing, Ontogeny, and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil
| | - Ramon S. Raposo
- Experimental Biology Core, Health Sciences Center, University of Fortaleza, Fortaleza 60812-020, Brazil
| | - Paulo Iury G. Nunes
- Natural Products Laboratory, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza 60430-270, Brazil
| | - Cássia R. Roque
- Laboratory of Tissue Healing, Ontogeny, and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil
| | - Flávia A. Santos
- Natural Products Laboratory, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza 60430-270, Brazil
| | - Geanne M. Andrade
- Neuroscience and Behavior Laboratory, Drug Research and Development Center, Federal University of Ceará, Fortaleza 60430-275, Brazil
| | - José Lucas Viana
- Department of Analytical Chemistry, Institute of Chemistry, University of Campinas—UNICAMP, Campinas 13083-862, Brazil
| | - Anne H. Fostier
- Department of Analytical Chemistry, Institute of Chemistry, University of Campinas—UNICAMP, Campinas 13083-862, Brazil
| | - Alessandra Sussulini
- Department of Analytical Chemistry, Institute of Chemistry, University of Campinas—UNICAMP, Campinas 13083-862, Brazil
| | - Jacqueline I. Alvarez-Leite
- Laboratory of Atherosclerosis and Nutritional Biochemistry, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Carlos Fontes-Ribeiro
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Institute of Pharmacology and Experimental Therapeutics and Center for Innovative Biomedicine and Biotechnology (CIBB), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - João O. Malva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Institute of Pharmacology and Experimental Therapeutics and Center for Innovative Biomedicine and Biotechnology (CIBB), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Reinaldo B. Oriá
- Laboratory of Tissue Healing, Ontogeny, and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil
| |
Collapse
|
9
|
Effects of splenectomy on skin inflammation and psoriasis-like phenotype of imiquimod-treated mice. Sci Rep 2022; 12:14738. [PMID: 36042262 PMCID: PMC9427736 DOI: 10.1038/s41598-022-18900-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/22/2022] [Indexed: 02/08/2023] Open
Abstract
Imiquimod (IMQ) is widely used as animal model of psoriasis, a chronic inflammatory skin disorder. Although topical application of IMQ to back skin causes splenomegaly in mice, how the spleen affects the psoriasis-like phenotype of IMQ-treated mice remains unclear. In this study, we analyzed the cellular composition of spleen and measured metabolites in blood of IMQ-treated mice. We also investigated whether splenectomy influences the degree of skin inflammation and pathology in IMQ-treated mice. Flow cytometry showed that the numbers of CD11b+Ly6c+ neutrophils, Ter119+ proerythroblasts, B220+ B cells, F4/80+ macrophages, and CD11c+ dendritic cells in the spleen were significantly higher in IMQ-treated mice compared to control mice. An untargeted metabolomics analysis of blood identified 14 metabolites, including taurine and 2,6-dihydroxybenzoic acid, whose levels distinguished the two groups. The composition of cells in the spleen and blood metabolites positively correlated with the weight of the spleen. However, splenectomy did not affect IMQ-induced psoriasis-like phenotypes compared with sham-operated mice, although splenectomy increased the expression of interleukin-17A mRNA in the skin of IMQ-treated mice. These data suggest that the spleen does not play a direct role in the development of psoriasis-like phenotype on skin of IMQ-treated mice, though IMQ causes splenomegaly.
Collapse
|
10
|
Pérez-Hernández E, Pastrana-Carballo JJ, Gómez-Chávez F, Gupta RC, Pérez-Hernández N. A Key Metabolic Regulator of Bone and Cartilage Health. Endocrinol Metab (Seoul) 2022; 37:559-574. [PMID: 35938304 PMCID: PMC9449101 DOI: 10.3803/enm.2022.1443] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/07/2022] [Indexed: 11/17/2022] Open
Abstract
Taurine, a cysteine-derived zwitterionic sulfonic acid, is a common ingredient in energy drinks and is naturally found in fish and other seafood. In humans, taurine is produced mainly in the liver, and it can also be obtained from food. In target tissues, such as the retina, heart, and skeletal muscle, it functions as an essential antioxidant, osmolyte, and antiapoptotic agent. Taurine is also involved in energy metabolism and calcium homeostasis. Taurine plays a considerable role in bone growth and development, and high-profile reports have demonstrated the importance of its metabolism for bone health. However, these reports have not been collated for more than 10 years. Therefore, this review focuses on taurine-bone interactions and covers recently discovered aspects of taurine's effects on osteoblastogenesis, osteoclastogenesis, bone structure, and bone pathologies (e.g., osteoporosis and fracture healing), with due attention to the taurine-cartilage relationship.
Collapse
Affiliation(s)
- Elizabeth Pérez-Hernández
- Medical Unit of High Specialty of Traumatology, Orthopedics and Rehabilitation “Dr. Victorio de la Fuente Narváez”, Mexican Social Security Institute, Mexico City, Mexico
| | | | - Fernando Gómez-Chávez
- National School of Medicine and Homeopathy, National Polytechnic Institute, Mexico City, Mexico
| | - Ramesh C. Gupta
- School of Agricultural Sciences and Rural Development (SASRD) Nagaland University, Medziphema, India
- Ramesh C. Gupta. School of Agricultural Sciences and Rural Development (SASRD) Nagaland University, Medziphema-797106, India Tel: +91-3862-247102, Fax: +91-3862-247113, E-mail:
| | - Nury Pérez-Hernández
- National School of Medicine and Homeopathy, National Polytechnic Institute, Mexico City, Mexico
- Corresponding authors: Nury Pérez-Hernández. National School of Medicine and Homeopathy, National Polytechnic Institute, 07320, Mexico City, Mexico Tel: +52-5729-6000 ext. 55537, Fax: +52-5729-6000, E-mail:
| |
Collapse
|
11
|
Duszka K. Versatile Triad Alliance: Bile Acid, Taurine and Microbiota. Cells 2022; 11:2337. [PMID: 35954180 PMCID: PMC9367564 DOI: 10.3390/cells11152337] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/21/2022] [Accepted: 07/24/2022] [Indexed: 11/21/2022] Open
Abstract
Taurine is the most abundant free amino acid in the body, and is mainly derived from the diet, but can also be produced endogenously from cysteine. It plays multiple essential roles in the body, including development, energy production, osmoregulation, prevention of oxidative stress, and inflammation. Taurine is also crucial as a molecule used to conjugate bile acids (BAs). In the gastrointestinal tract, BAs deconjugation by enteric bacteria results in high levels of unconjugated BAs and free taurine. Depending on conjugation status and other bacterial modifications, BAs constitute a pool of related but highly diverse molecules, each with different properties concerning solubility and toxicity, capacity to activate or inhibit receptors of BAs, and direct and indirect impact on microbiota and the host, whereas free taurine has a largely protective impact on the host, serves as a source of energy for microbiota, regulates bacterial colonization and defends from pathogens. Several remarkable examples of the interaction between taurine and gut microbiota have recently been described. This review will introduce the necessary background information and lay out the latest discoveries in the interaction of the co-reliant triad of BAs, taurine, and microbiota.
Collapse
Affiliation(s)
- Kalina Duszka
- Department of Nutritional Sciences, University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
12
|
Liu W, Wang Q, Chang J, Bhetuwal A, Bhattarai N, Ni X. Circulatory Metabolomics Reveals the Association of the Metabolites With Clinical Features in the Patients With Intrahepatic Cholestasis of Pregnancy. Front Physiol 2022; 13:848508. [PMID: 35899031 PMCID: PMC9309339 DOI: 10.3389/fphys.2022.848508] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Intrahepatic cholestasis of pregnancy (ICP) is associated with an increased risk of adverse pregnancy to the mother and fetus. As yet, the metabolic profiles and the association of the clinical features remain obscure. Methods: Fifty-seven healthy pregnant women and 52 patients with ICP were recruited in this study. Plasma samples were collected from pregnancies who received prenatal care between 30 and 36 weeks. Untargeted metabolomics to portray the metabolic profiles were performed by LC/MS. Multivariate combined with the univariate analysis was performed to screen out differential metabolites between the ICP and control groups. A de-biased sparse partial correlation (DSPC) network analysis of differential metabolites was conducted to explore the potential mutual regulation among metabolites on the basis of de-sparsified graphical lasso modeling. The pathway analysis was carried out using MetaboAnalyst. Linear regression and Pearson correlation analysis was applied to analyze correlations of bile acid levels, metabolites, newborn weights, and pregnancy outcomes in ICP patients. Results: Conspicuous metabolic changes and choreographed metabolic profiles were disclosed: 125 annotated metabolites and 18 metabolic pathways were disturbed in ICP patients. DSPC networks indicated dense interactions among amino acids and their derivatives, bile acids, carbohydrates, and organic acids. The levels of total bile acid (TBA) were increased in ICP patients with meconium-stained amniotic fluid (MSAF) compared with those without MSAF. An abnormal tryptophan metabolism, elevated long chain saturated fatty acids and estrone sulfate levels, and a low-antioxidant capacity were relevant to increased bile acid levels. Newborn weights were significantly associated with the levels of bile acids and some metabolites of amino acids. Conclusion: Our study revealed the metabolomic profiles in circulation and the correlation of the metabolites with clinical features in ICP patients. Our data suggest that disturbances in metabolic pathways might be associated with adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Wenhu Liu
- Department of Gynecology and Obstetrics, International Collaborative Research Center for Medical Metabolomics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
- School of Pharmacy, School of Basic Medical Sciences and Forensic Medical, North Sichuan Medical College, Nanchong, China
| | - Qiang Wang
- Department of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, China
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jinxia Chang
- School of Pharmacy, School of Basic Medical Sciences and Forensic Medical, North Sichuan Medical College, Nanchong, China
| | - Anup Bhetuwal
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Nisha Bhattarai
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xin Ni
- Department of Gynecology and Obstetrics, International Collaborative Research Center for Medical Metabolomics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
- *Correspondence: Xin Ni,
| |
Collapse
|
13
|
Rubio-Casillas A, Gupta RC, Redwa EM, Uversky VN, Badierah R. Early taurine administration as a means for halting the cytokine storm progression in COVID-19 patients. EXPLORATION OF MEDICINE 2022. [DOI: 10.37349/emed.2022.00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Around the world, more than 6.2 million individuals have died as a result of coronavirus disease 2019 (COVID-19). According to a recent survey conducted among immunologists, epidemiologists, and virologists, this disease is expected to become endemic. This implies that the disease could have a continuous presence and/or normal frequency in the population. Pharmacological interventions to prevent infection, as well as to treat the patients at an early phase of illness to avoid hospitalization are essential additions to the vaccines. Taurine is known to inhibit the generation of all inflammatory mediators linked to the cytokine storm. It can also protect against lung injury by suppressing increased oxidants production and promoting the resolution of the inflammatory process. Neutrophil lactoferrin degranulation stimulated by taurine may have antiviral effects against SARS-CoV-2, limiting viral replication. It is hypothesized that if taurine is administered early in the onset of COVID-19 disease, it may stop the cytokine storm from progressing, lowering morbidity and mortality.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- 1Autlán Regional Hospital, Health Secretariat, Autlán, Jalisco 48900, Mexico 2Biology Laboratory, Autlán Regional High School, University of Guadalajara, Autlán, Jalisco 48900, Mexico
| | - Ramesh C. Gupta
- 3School of Agricultural Sciences and Rural Development, Nagaland University, Medziphema 797004, India
| | - Elrashdy M. Redwa
- 4Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia 5Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria 21934, Egypt
| | - Vladimir N. Uversky
- 6Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Raied Badierah
- 7Medical Laboratory, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
14
|
Newman SD, Schnakenberg Martin AM, Raymond D, Cheng H, Wilson L, Barnes S, O’Donnell BF. The relationship between cannabis use and taurine: A MRS and metabolomics study. PLoS One 2022; 17:e0269280. [PMID: 35653401 PMCID: PMC9162360 DOI: 10.1371/journal.pone.0269280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/18/2022] [Indexed: 02/07/2023] Open
Abstract
Taurine is an essential amino acid. It has been shown to be neuroprotective including protecting against the neurotoxic effects of glutamate. The goal of the current study was to examine the relationship between CB use and taurine measured in brain using magnetic resonance spectroscopy (MRS), and peripherally from a urine sample. Two experiments are presented. The first is a reanalysis of published data that examined taurine and glutamate in the dorsal anterior cingulate of a CB user group and non-user group using MRS. The second experiment, in a separate CB user group, used metabolomics analysis to measure taurine levels in urine. Because body composition has been associated with the pharmacokinetics of cannabis and taurine levels, a moderation model was examined with body composition included as the covariate. The MRS study found taurine levels were correlated with glutamate in both groups and taurine was correlated with frequency of CB use in the CB user group. The moderation model demonstrated significant effects of CB use and BMI; the interaction was marginally significant with lower BMI individuals showing a positive relationship between CB use and taurine. A similar finding was observed for the urine analysis. Both CB use and weight, as well as the interaction were significant. In this case, individuals with higher weight showed an association between CB use and taurine levels. This study shows the feasibility and potential importance of examining the relationship between taurine and CB use as it may shed light on a mechanism that underlies the neuroprotective effects of CB.
Collapse
Affiliation(s)
- Sharlene D. Newman
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, Alabama, United States of America
- * E-mail:
| | - Ashley M. Schnakenberg Martin
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Psychology Service, VA Connecticut Healthcare System, West Haven, Connecticut, United States of America
| | - David Raymond
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, United States of America
| | - Hu Cheng
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, United States of America
| | - Landon Wilson
- Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Stephen Barnes
- Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Brian F. O’Donnell
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, United States of America
- Program in Neuroscience, Indiana University, Bloomington, Indiana, United States of America
| |
Collapse
|
15
|
Yang JY, Zhang TT, Yu ZL, Wang CC, Zhao YC, Wang YM, Xue CH. Taurine Alleviates Trimethylamine N-Oxide-Induced Atherosclerosis by Regulating Bile Acid Metabolism in ApoE -/- Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:5738-5747. [PMID: 35486890 DOI: 10.1021/acs.jafc.2c01376] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Trimethylamine N-oxide (TMAO) widely exists in seafood and is associated with the atherosclerosis progress, but dietary seafood reduced the cardiovascular risk. This intimates that there may be some ingredients in seafood to offset the cardiovascular risk caused by TMAO. Taurine is a marker ingredient in seafood. Thus, this study determined the influences of taurine on TMAO-induced atherosclerosis in apolipoprotein-E-deficient mice. The results showed that dietary taurine significantly reduced the TMAO-induced atherosclerotic lesion area. Further studies found that taurine increased the hepatic- and serum-conjugated bile acid/unconjugated bile acid ratio via increasing hepatic gene expression of conjugated bile acid synthesis. Meanwhile, taurine changed TMAO-induced abnormal bile acid profiles in the gallbladder. Moreover, taurine increased bile acid deconjugation by enhancing the genera Ruminiclostridium level and increased excretion of fecal neutral sterols. Additionally, taurine attenuated inflammation in the serum and artery. These results indicate that taurine alleviated TMAO-induced atherosclerosis via regulating bile acid metabolism.
Collapse
Affiliation(s)
- Jin-Yue Yang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Tian-Tian Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Zhu-Lin Yu
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Cheng-Cheng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Ying-Cai Zhao
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
- Laboratory of Marine Drugs & Biological Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong 266237, People's Republic of China
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
- Laboratory of Marine Drugs & Biological Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong 266237, People's Republic of China
| |
Collapse
|
16
|
Xia Y, Huang X, Mo L, Wang C, Fan W, Huang H. TMT-based proteomics analysis of the cerebral cortex of TauT knockout rats. Proteome Sci 2022; 20:6. [PMID: 35468821 PMCID: PMC9040245 DOI: 10.1186/s12953-022-00189-z] [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: 10/08/2021] [Accepted: 04/13/2022] [Indexed: 11/16/2022] Open
Abstract
Background Taurine serves a variety of nutritional and physiological roles, and it is mostly transported in cells via taurine transporter (TauT). The effect of taurine transporter in cerebral cortex is still unknown. We employed TMT label-based proteomics to find differences in proteins in the cerebral cortex of TauT knockout rats in this investigation. The goal of this research was to see how TauT deletion affected protein alterations in brain tissue and to see if there was a new research area for TauT. Methods The cerebral cortex of TauT knockout rats and wild-type control rats were analyzed using TMT-based proteomics, and differentially expressed proteins were analyzed by bioinformatics analysis means such as GO and KEGG, the association between the proteins was found by PPI, and biologically significant and interesting proteins were selected for verification by WB and immunohistochemistry. Results There were total of 8275 proteins found, but only 35 differentially expressed proteins were identified (27 up-regulated and 8 down-regulated), and gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to predict the biological pathways and functional classification of the proteins. The results show that these differentially expressed proteins are mainly enriched in lysine degradation, cell cycle, chronic myeloid leukemia, and longevity regulating pathways-multiple species, renal cell carcinoma, pathways in cancer, etc. To verify the proteomic data, we analyzed the expression of Annexin6 and Pik3r2 by western blotting and immunofluorescence. The results are consistent with proteomics, which proves the reliability of our proteomics data. Conclusion Through TMT-based proteomics, we have a comprehensive understanding of the effect of TauT knockout on the changes of other proteins in the cerebral cortex, providing new evidence for further understanding the function of TauT.
Collapse
Affiliation(s)
- Yiming Xia
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Xiaoling Huang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Lidong Mo
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Chen Wang
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Weijia Fan
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Huiling Huang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China. .,Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China.
| |
Collapse
|
17
|
Wilde C, Mitgau J, Suchý T, Schoeneberg T, Liebscher I. Translating the Force - mechano-sensing GPCRs. Am J Physiol Cell Physiol 2022; 322:C1047-C1060. [PMID: 35417266 DOI: 10.1152/ajpcell.00465.2021] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Incorporating mechanical cues into cellular responses allows us to experience our direct environment. Specialized cells can perceive and discriminate between different physical properties such as level of vibration, temperature, or pressure. Mechanical forces are abundant signals that also shape general cellular responses such as cytoskeletal rearrangement, differentiation, or migration and contribute to tissue development and function. The molecular structures that perceive and transduce mechanical forces are specialized cytoskeletal proteins, cell junction molecules, and membrane proteins such as ion channels and metabotropic receptors. G protein-coupled receptors (GPCRs) have attracted attention as metabotropic force receptors as they are among the most important drug targets. This review summarizes the function of mechano-sensitive GPCRs, specifically, the angiotensin II type 1 receptor and adrenergic, apelin, histamine, parathyroid hormone 1, and orphan receptors, focusing particularly on the advanced knowledge gained from adhesion-type GPCRs. We distinguish between shear stress and cell swelling/stretch as the two major types of mechano-activation of these receptors and contemplate the potential contribution of the force-from-lipid and force-from-tether models that have previously been suggested for ion channels.
Collapse
Affiliation(s)
- Caroline Wilde
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Jakob Mitgau
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Tomás Suchý
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Torsten Schoeneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| |
Collapse
|
18
|
Bai X, Bian Z. MicroRNA-21 Is a Versatile Regulator and Potential Treatment Target in Central Nervous System Disorders. Front Mol Neurosci 2022; 15:842288. [PMID: 35173580 PMCID: PMC8841607 DOI: 10.3389/fnmol.2022.842288] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of endogenous, non-coding, single-stranded RNAs with a length of approximately 22 nucleotides that are found in eukaryotes. miRNAs are involved in the regulation of cell differentiation, proliferation, invasion, apoptosis, and metabolism by regulating the expression of their target genes. Emerging studies have suggested that various miRNAs play key roles in the pathogenesis of central nervous system (CNS) disorders and may be viable therapeutic targets. In particular, miR-21 has prominently emerged as a focus of increasing research on the mechanisms of its involvement in CNS disorders. Herein, we reviewed recent studies on the critical roles of miR-21, including its dysregulated expression and target genes, in the regulation of pathophysiological processes of CNS disorders, with a special focus on apoptosis and inflammation. Collectively, miR-21 is a versatile regulator in the progression of CNS disorders and could be a promising biomarker and therapeutic target for these diseases. An in-depth understanding of the mechanisms by which miR-21 affects the pathogenesis of CNS disorders could pave the way for miR-21 to serve as a therapeutic target for these conditions.
Collapse
Affiliation(s)
- Xue Bai
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhigang Bian
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Zhigang Bian,
| |
Collapse
|
19
|
Gregor A, Pignitter M, Trajanoski S, Auernigg-Haselmaier S, Somoza V, König J, Duszka K. Microbial contribution to the caloric restriction-triggered regulation of the intestinal levels of glutathione transferases, taurine, and bile acid. Gut Microbes 2022; 13:1992236. [PMID: 34693866 PMCID: PMC8547879 DOI: 10.1080/19490976.2021.1992236] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Recently we showed that caloric restriction (CR) triggers an increase in the levels of free taurine, taurine-conjugated bile acids (BA), and other taurine conjugates in intestinal mucosa while decreasing glutathione (GSH) levels in wild-type male mice. In the current project, we decided to investigate whether the microbiota is involved in the response to CR by depleting gut bacteria. The antibiotics treatment diminished CR-specific increase in the levels of free taurine and its conjugates as well as upregulated expression and activity of GSH transferases (GST) in the intestinal mucosa. Further, it diminished a CR-related increase in BAs levels in the liver, plasma, and intestinal mucosa. Transplant of microbiota from CR mice to ad libitum fed mice triggered CR-like changes in MGST1 expression, levels of taurine and taurine conjugates in the mucosa of the ileum. We show for the first time, that microbiota contributes to the intestinal response to CR-triggered changes in BA, taurine, and GST levels.
Collapse
Affiliation(s)
- András Gregor
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Marc Pignitter
- Department of Physiological Chemistry, University of Vienna, Vienna, Austria
| | - Slave Trajanoski
- Core Facility Computational Bioanalytics, Medical University of Graz, Graz, Austria
| | | | - Veronika Somoza
- Department of Physiological Chemistry, University of Vienna, Vienna, Austria,Leibniz-Institut for Food Systems Biology, Technical University of Munich, Munich, Germany
| | - Jürgen König
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Kalina Duszka
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria,CONTACT Kalina Duszka Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| |
Collapse
|
20
|
Mani V, Mohd Azahan NS, Ramasamy K, Lim SM, Abdul Majeed AB. Mahanimbine Improved Aging-Related Memory Deficits in Mice through Enhanced Cholinergic Transmission and Suppressed Oxidative Stress, Amyloid Levels, and Neuroinflammation. Brain Sci 2021; 12:12. [PMID: 35053756 PMCID: PMC8773876 DOI: 10.3390/brainsci12010012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/14/2022] Open
Abstract
Murraya koenigii leaves contain mahanimbine, a carbazole alkaloid, reported with improving cholinergic neuronal transmission and reducing neuroinflammation in the CNS. The current research investigated the effects of mahanimbine on age-related memory deficits, oxidative stress, cholinergic dysfunction, amyloid formation, and neuroinflammation in aged mice (16 months old). Mahanimbine was administered (1 and 2 mg/kg, p.o.) daily to groups of aged mice for 30 days. The Morris water maze (MWM) task was performed to study spatial learning (escape latency (EL) and swimming distance (SD)) and memory (probe test). The levels of malondialdehyde (MDA), glutathione (GSH), acetylcholine (ACh), acetylcholinesterase (AChE), β-amyloid (Aβ1-40 and Aβ1-42), β-secretase (BACE-1), as well as neuroinflammation markers (total cyclooxygenase (COX) and COX-2 expression), were measured from the isolated brain. Mahanimbine reduced the EL time and SD in the MWM test. From the probe trial, the mahanimbine-treated group spent more time in the targeted quadrant related to the age-matched control, which indicated the enhancement of memory retention. From the biochemical tests, the treatment decreased MDA, AChE, Aβ1-40, and Aβ1-42, BACE-1, total COX activity, and COX-2 expression. It also raised the brain GSH and ACh levels in aged mice compared to age-matched control. These results have supported the reversal of memory dysfunctions by mahanimbine in aged mice and hypothesized that it could be a potential target to treat age-related neurodegenerative disease.
Collapse
Affiliation(s)
- Vasudevan Mani
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| | - Nur Syamimi Mohd Azahan
- Collaborative Drug Discovery Research (CDDR) Group and Brain Degeneration and Therapeutics Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam 42300, Malaysia; (N.S.M.A.); (K.R.); (S.M.L.); (A.B.A.M.)
| | - Kalavathy Ramasamy
- Collaborative Drug Discovery Research (CDDR) Group and Brain Degeneration and Therapeutics Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam 42300, Malaysia; (N.S.M.A.); (K.R.); (S.M.L.); (A.B.A.M.)
| | - Siong Meng Lim
- Collaborative Drug Discovery Research (CDDR) Group and Brain Degeneration and Therapeutics Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam 42300, Malaysia; (N.S.M.A.); (K.R.); (S.M.L.); (A.B.A.M.)
| | - Abu Bakar Abdul Majeed
- Collaborative Drug Discovery Research (CDDR) Group and Brain Degeneration and Therapeutics Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam 42300, Malaysia; (N.S.M.A.); (K.R.); (S.M.L.); (A.B.A.M.)
| |
Collapse
|
21
|
Neganova M, Liu J, Aleksandrova Y, Klochkov S, Fan R. Therapeutic Influence on Important Targets Associated with Chronic Inflammation and Oxidative Stress in Cancer Treatment. Cancers (Basel) 2021; 13:6062. [PMID: 34885171 PMCID: PMC8657135 DOI: 10.3390/cancers13236062] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/28/2021] [Accepted: 11/28/2021] [Indexed: 01/17/2023] Open
Abstract
Chronic inflammation and oxidative stress are the interconnected pathological processes, which lead to cancer initiation and progression. The growing level of oxidative and inflammatory damage was shown to increase cancer severity and contribute to tumor spread. The overproduction of reactive oxygen species (ROS), which is associated with the reduced capacity of the endogenous cell defense mechanisms and/or metabolic imbalance, is the main contributor to oxidative stress. An abnormal level of ROS was defined as a predisposing factor for the cell transformation that could trigger pro-oncogenic signaling pathways, induce changes in gene expression, and facilitate accumulation of mutations, DNA damage, and genomic instability. Additionally, the activation of transcription factors caused by a prolonged oxidative stress, including NF-κB, p53, HIF1α, etc., leads to the expression of several genes responsible for inflammation. The resulting hyperactivation of inflammatory mediators, including TNFα, TGF-β, interleukins, and prostaglandins can contribute to the development of neoplasia. Pro-inflammatory cytokines were shown to trigger adaptive reactions and the acquisition of resistance by tumor cells to apoptosis, while promoting proliferation, invasion, and angiogenesis. Moreover, the chronic inflammatory response leads to the excessive production of free radicals, which further aggravate the initiated reactions. This review summarizes the recent data and progress in the discovery of mechanisms that associate oxidative stress and chronic inflammation with cancer onset and metastasis. In addition, the review provides insights for the development of therapeutic approaches and the discovery of natural substances that will be able to simultaneously inhibit several key oncological and inflammation-related targets.
Collapse
Affiliation(s)
- Margarita Neganova
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China; (M.N.); (J.L.)
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China; (M.N.); (J.L.)
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yulia Aleksandrova
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Sergey Klochkov
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Ruitai Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China; (M.N.); (J.L.)
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
22
|
Wang K, Zhang B, Tian T, Zhang B, Shi G, Zhang C, Li G, Huang M. Taurine protects dopaminergic neurons in paraquat-induced Parkinson's disease mouse model through PI3K/Akt signaling pathways. Amino Acids 2021; 54:1-11. [PMID: 34837554 DOI: 10.1007/s00726-021-03104-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/24/2021] [Indexed: 01/19/2023]
Abstract
Taurine (Tau) is one of the most abundant amino acids in the brain and regulates physiological functions in the central nervous system, including anti-inflammatory effects. There is growing evidence that microglia-mediated neuro-inflammatory responses are an integral part of Parkinson's disease (PD) onset and progression. Among the many factors regulating the inflammatory response, phosphatidylinositol-3 kinase (PI3K) is susceptible to activation by a variety of cytokines and physicochemical factors, and subsequently recruits signaling proteins containing the pleckstrin homology structural domain to further regulate protein kinase B (AKT) expression involved in the regulation of the intracellular immune response and inflammatory response. Therefore, we established a PD mouse model using paraquat (PQ) intraperitoneal injection staining to explore the mechanism of Tau action on PI3K/AKT signaling pathway. Our study showed that PD mice with Tau intervention recovered motor and non-motor functions to some extent, and the number of dopaminergic (DAc) neurons in the substantia nigra and the level of dopamine (DA) secretion in the striatum were also significantly increased compared with the PQ-dyed group, and the protein content of PI3K and PDK-1 and the phosphorylation level of AKT were reduced in parallel with the reduction in the expression of microglia and related inflammatory factors. In conclusion, our results suggest that Tau may regulate microglia-mediated inflammatory responses through inhibition of the PI3K/AKT pathway in the midbrain of PD mice, thereby reducing DAc neurons damage.
Collapse
Affiliation(s)
- Kaidong Wang
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China.,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Baofu Zhang
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China.,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Tian Tian
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China.,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Bingyang Zhang
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China.,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Ge Shi
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China.,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Chunhui Zhang
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China.,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Guoliang Li
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China.,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Min Huang
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China. .,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China.
| |
Collapse
|
23
|
Surai PF, Earle-Payne K, Kidd MT. Taurine as a Natural Antioxidant: From Direct Antioxidant Effects to Protective Action in Various Toxicological Models. Antioxidants (Basel) 2021; 10:1876. [PMID: 34942978 PMCID: PMC8698923 DOI: 10.3390/antiox10121876] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 12/18/2022] Open
Abstract
Natural antioxidants have received tremendous attention over the last 3 decades. At the same time, the attitude to free radicals is slowly changing, and their signalling role in adaptation to stress has recently received a lot of attention. Among many different antioxidants in the body, taurine (Tau), a sulphur-containing non-proteinogenic β-amino acid, is shown to have a special place as an important natural modulator of the antioxidant defence networks. Indeed, Tau is synthesised in most mammals and birds, and the Tau requirement is met by both synthesis and food/feed supply. From the analysis of recent data, it could be concluded that the direct antioxidant effect of Tau due to scavenging free radicals is limited and could be expected only in a few mammalian/avian tissues (e.g., heart and eye) with comparatively high (>15-20 mM) Tau concentrations. The stabilising effects of Tau on mitochondria, a prime site of free radical formation, are characterised and deserve more attention. Tau deficiency has been shown to compromise the electron transport chain in mitochondria and significantly increase free radical production. It seems likely that by maintaining the optimal Tau status of mitochondria, it is possible to control free radical production. Tau's antioxidant protective action is of great importance in various stress conditions in human life, and is related to commercial animal and poultry production. In various in vitro and in vivo toxicological models, Tau showed AO protective effects. The membrane-stabilizing effects, inhibiting effects on ROS-producing enzymes, as well as the indirect AO effects of Tau via redox balance maintenance associated with the modulation of various transcription factors (e.g., Nrf2 and NF-κB) and vitagenes could also contribute to its protective action in stress conditions, and thus deserve more attention.
Collapse
Affiliation(s)
- Peter F. Surai
- Vitagene and Health Research Centre, Bristol BS4 2RS, UK
- Department of Microbiology and Biochemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria
- Biochemistry and Physiology Department, Saint-Petersburg State University of Veterinary Medicine, 196084 St. Petersburg, Russia
- Department of Animal Nutrition, Faculty of Agricultural and Environmental Sciences, Szent Istvan University, H-2103 Gödöllo, Hungary
| | - Katie Earle-Payne
- NHS Greater Glasgow and Clyde, Renfrewshire Health and Social Care Centre, 10 Ferry Road, Renfrew PA4 8RU, UK;
| | - Michael T. Kidd
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
| |
Collapse
|
24
|
Parga JA, Rodriguez-Perez AI, Garcia-Garrote M, Rodriguez-Pallares J, Labandeira-Garcia JL. NRF2 Activation and Downstream Effects: Focus on Parkinson's Disease and Brain Angiotensin. Antioxidants (Basel) 2021; 10:antiox10111649. [PMID: 34829520 PMCID: PMC8614768 DOI: 10.3390/antiox10111649] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) are signalling molecules used to regulate cellular metabolism and homeostasis. However, excessive ROS production causes oxidative stress, one of the main mechanisms associated with the origin and progression of neurodegenerative disorders such as Parkinson's disease. NRF2 (Nuclear Factor-Erythroid 2 Like 2) is a transcription factor that orchestrates the cellular response to oxidative stress. The regulation of NRF2 signalling has been shown to be a promising strategy to modulate the progression of the neurodegeneration associated to Parkinson's disease. The NRF2 pathway has been shown to be affected in patients with this disease, and activation of NRF2 has neuroprotective effects in preclinical models, demonstrating the therapeutic potential of this pathway. In this review, we highlight recent advances regarding the regulation of NRF2, including the effect of Angiotensin II as an endogenous signalling molecule able to regulate ROS production and oxidative stress in dopaminergic neurons. The genes regulated and the downstream effects of activation, with special focus on Kruppel Like Factor 9 (KLF9) transcription factor, provide clues about the mechanisms involved in the neurodegenerative process as well as future therapeutic approaches.
Collapse
Affiliation(s)
- Juan A. Parga
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
- Correspondence: (J.A.P.); (J.L.L.-G.)
| | - Ana I. Rodriguez-Perez
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
| | - Maria Garcia-Garrote
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
| | - Jannette Rodriguez-Pallares
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
| | - Jose L. Labandeira-Garcia
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
- Correspondence: (J.A.P.); (J.L.L.-G.)
| |
Collapse
|
25
|
Likhitweerawong N, Thonusin C, Boonchooduang N, Louthrenoo O, Nookaew I, Chattipakorn N, Chattipakorn SC. Profiles of urine and blood metabolomics in autism spectrum disorders. Metab Brain Dis 2021; 36:1641-1671. [PMID: 34338974 PMCID: PMC8502415 DOI: 10.1007/s11011-021-00788-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 07/01/2021] [Indexed: 01/06/2023]
Abstract
Early diagnosis and treatment for autism spectrum disorder (ASD) pose challenges. The current diagnostic approach for ASD is mainly clinical assessment of patient behaviors. Biomarkers-based identification of ASD would be useful for pediatricians. Currently, there is no specific treatment for ASD, and evidence for the efficacy of alternative treatments remains inconclusive. The prevalence of ASD is increasing, and it is becoming more urgent to find the pathogenesis of such disorder. Metabolomic studies have been used to deeply investigate the alteration of metabolic pathways, including those associated with ASD. Metabolomics is a promising tool for identifying potential biomarkers and possible pathogenesis of ASD. This review comprehensively summarizes and discusses the abnormal metabolic pathways in ASD children, as indicated by evidence from metabolomic studies in urine and blood. In addition, the targeted interventions that could correct the metabolomic profiles relating to the improvement of autistic behaviors in affected animals and humans have been included. The results revealed that the possible underlying pathophysiology of ASD were alterations of amino acids, reactive oxidative stress, neurotransmitters, and microbiota-gut-brain axis. The potential common pathways shared by animal and human studies related to the improvement of ASD symptoms after pharmacological interventions were mammalian-microbial co-metabolite, purine metabolism, and fatty acid oxidation. The content of this review may contribute to novel biomarkers for the early diagnosis of ASD and possible therapeutic paradigms.
Collapse
Affiliation(s)
- Narueporn Likhitweerawong
- Division of Growth and Development, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chanisa Thonusin
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, 110 Inthawarorot Road, Sriphum, Muang, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nonglak Boonchooduang
- Division of Growth and Development, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Orawan Louthrenoo
- Division of Growth and Development, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Intawat Nookaew
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Arkanasa, USA
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, 110 Inthawarorot Road, Sriphum, Muang, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C. Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, 110 Inthawarorot Road, Sriphum, Muang, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
26
|
The Role of Taurine in Mitochondria Health: More Than Just an Antioxidant. Molecules 2021; 26:molecules26164913. [PMID: 34443494 PMCID: PMC8400259 DOI: 10.3390/molecules26164913] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/08/2021] [Accepted: 08/10/2021] [Indexed: 12/21/2022] Open
Abstract
Taurine is a naturally occurring sulfur-containing amino acid that is found abundantly in excitatory tissues, such as the heart, brain, retina and skeletal muscles. Taurine was first isolated in the 1800s, but not much was known about this molecule until the 1990s. In 1985, taurine was first approved as the treatment among heart failure patients in Japan. Accumulating studies have shown that taurine supplementation also protects against pathologies associated with mitochondrial defects, such as aging, mitochondrial diseases, metabolic syndrome, cancer, cardiovascular diseases and neurological disorders. In this review, we will provide a general overview on the mitochondria biology and the consequence of mitochondrial defects in pathologies. Then, we will discuss the antioxidant action of taurine, particularly in relation to the maintenance of mitochondria function. We will also describe several reported studies on the current use of taurine supplementation in several mitochondria-associated pathologies in humans.
Collapse
|
27
|
Baliou S, Adamaki M, Ioannou P, Pappa A, Panayiotidis MI, Spandidos DA, Christodoulou I, Kyriakopoulos AM, Zoumpourlis V. Protective role of taurine against oxidative stress (Review). Mol Med Rep 2021; 24:605. [PMID: 34184084 PMCID: PMC8240184 DOI: 10.3892/mmr.2021.12242] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/03/2021] [Indexed: 12/14/2022] Open
Abstract
Taurine is a fundamental mediator of homeostasis that exerts multiple roles to confer protection against oxidant stress. The development of hypertension, muscle/neuro‑associated disorders, hepatic cirrhosis, cardiac dysfunction and ischemia/reperfusion are examples of some injuries that are linked with oxidative stress. The present review gives a comprehensive description of all the underlying mechanisms of taurine, with the aim to explain its anti‑oxidant actions. Taurine is regarded as a cytoprotective molecule due to its ability to sustain normal electron transport chain, maintain glutathione stores, upregulate anti‑oxidant responses, increase membrane stability, eliminate inflammation and prevent calcium accumulation. In parallel, the synergistic effect of taurine with other potential therapeutic modalities in multiple disorders are highlighted. Apart from the results derived from research findings, the current review bridges the gap between bench and bedside, providing mechanistic insights into the biological activity of taurine that supports its potential therapeutic efficacy in clinic. In the future, further clinical studies are required to support the ameliorative effect of taurine against oxidative stress.
Collapse
Affiliation(s)
- Stella Baliou
- National Hellenic Research Foundation, 11635 Athens, Greece
| | - Maria Adamaki
- National Hellenic Research Foundation, 11635 Athens, Greece
| | - Petros Ioannou
- Department of Internal Medicine and Infectious Diseases, University Hospital of Heraklion, 71110 Heraklion, Greece
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Mihalis I. Panayiotidis
- Department of Cancer Genetics, Therapeutics and Ultrastructural Pathology, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
- The Cyprus School of Molecular Medicine, 2371 Nicosia, Cyprus
| | - Demetrios A. Spandidos
- Department of Internal Medicine and Infectious Diseases, University Hospital of Heraklion, 71110 Heraklion, Greece
| | | | | | | |
Collapse
|
28
|
Modulation of vigabatrin induced cerebellar injury: the role of caspase-3 and RIPK1/RIPK3-regulated cell death pathways. J Mol Histol 2021; 52:781-798. [PMID: 34046766 DOI: 10.1007/s10735-021-09984-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/24/2021] [Indexed: 01/11/2023]
Abstract
Vigabatrin is the drug of choice in resistant epilepsy and infantile spasms. Ataxia, tremors, and abnormal gait have been frequently reported following its use indicating cerebellar involvement. This study aimed, for the first time, to investigate the involvement of necroptosis and apoptosis in the VG-induced cerebellar cell loss and the possible protective role of combined omega-3 and vitamin B12 supplementation. Fifty Sprague-Dawley adult male rats (160-200 g) were divided into equal five groups: the control group received normal saline, VG200 and VG400 groups received VG (200 mg or 400 mg/kg, respectively), VG200 + OB and VG400 + OB groups received combined VG (200 mg or 400 mg/kg, respectively), vitamin B12 (1 mg/kg), and omega-3 (1 g/kg). All medications were given daily by gavage for four weeks. Histopathological changes were examined in H&E and luxol fast blue (LFB) stained sections. Immunohistochemical staining for caspase-3 and receptor-interacting serine/threonine-protein kinase-1 (RIPK1) as well as quantitative real-time polymerase chain reaction (qRT-PCR) for myelin basic protein (MBP), caspase-3, and receptor-interacting serine/threonine-protein kinase-3 (RIPK3) genes were performed. VG caused a decrease in the granular layer thickness and Purkinje cell number, vacuolations, demyelination, suppression of MBP gene expression, and induction of caspases-3, RIPK1, and RIPK3 in a dose-related manner. Combined supplementation with B12 and omega-3 improved the cerebellar histology, increased MBP, and decreased apoptotic and necroptotic markers. In conclusion, VG-induced neuronal cell loss is dose-dependent and related to both apoptosis and necroptosis. This could either be ameliorated (in low-dose VG) or reduced (in high-dose VG) by combined supplementation with B12 and omega-3.
Collapse
|
29
|
Gonzalez-Vazquez A, Aguilar-Peralta AK, Tomas-Sanchez C, Blanco-Alvarez VM, Martinez-Fong D, Gonzalez-Barrios JA, Treviño S, Millán-Perez Peña L, Alatriste V, Soto-Rodriguez G, Brambila E, Leon-Chavez BA. Taurine Increases Zinc Preconditioning-Induced Prevention of Nitrosative Stress, Metabolic Alterations, and Motor Deficits in Young Rats following Intrauterine Ischemia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6696538. [PMID: 34040692 PMCID: PMC8121588 DOI: 10.1155/2021/6696538] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/12/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022]
Abstract
Oxygen deprivation in newborns leads to hypoxic-ischemic encephalopathy, whose hallmarks are oxidative/nitrosative stress, energetic metabolism alterations, nutrient deficiency, and motor behavior disability. Zinc and taurine are known to protect against hypoxic-ischemic brain damage in adults and neonates. However, the combined effect of prophylactic zinc administration and therapeutic taurine treatment on intrauterine ischemia- (IUI-) induced cerebral damage remains unknown. The present work evaluated this issue in male pups subjected to transient IUI (10 min) at E17 and whose mothers received zinc from E1 to E16 and taurine from E17 to postnatal day 15 (PND15) via drinking water. We assessed motor alterations, nitrosative stress, lipid peroxidation, and the antioxidant system comprised of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx). Enzymes of neuronal energetic pathways, such as aspartate aminotransferase (AST), alanine aminotransferase (ALT), and lactate dehydrogenase (LDH), were also evaluated. The hierarchization score of the protective effect of pharmacological strategies (HSPEPS) was used to select the most effective treatment. Compared with the IUI group, zinc, alone or combined with taurine, improved motor behavior and reduced nitrosative stress by increasing SOD, CAT, and GPx activities and decreasing the GSSG/GSH ratio in the cerebral cortex and hippocampus. Taurine alone increased the AST/ALT, LDH/ALT, and AST/LDH ratios in the cerebral cortex, showing improvement of the neural bioenergetics system. This result suggests that taurine improves pyruvate, lactate, and glutamate metabolism, thus decreasing IUI-caused cerebral damage and relieving motor behavior impairment. Our results showed that taurine alone or in combination with zinc provides neuroprotection in the IUI rat model.
Collapse
Affiliation(s)
- Alejandro Gonzalez-Vazquez
- Facultad de Ciencias Químicas, Benemérita, Universidad Autónoma de Puebla, 14 sur y Av. San Claudio, Puebla, 72570 Puebla, Mexico
| | - Ana-Karina Aguilar-Peralta
- Facultad de Ciencias Químicas, Benemérita, Universidad Autónoma de Puebla, 14 sur y Av. San Claudio, Puebla, 72570 Puebla, Mexico
| | - Constantino Tomas-Sanchez
- Facultad de Ciencias Químicas, Benemérita, Universidad Autónoma de Puebla, 14 sur y Av. San Claudio, Puebla, 72570 Puebla, Mexico
| | - Victor-Manuel Blanco-Alvarez
- Facultad de enfermería, Benemérita Universidad Autónoma de Puebla, 27 sur 1304, Col. Volcanes, Puebla, 72410 Puebla, Mexico
- Facultad de Medicina, Benemérita Universidad Autónoma de Puebla, 13 sur 2702, Col. Volcanes, Puebla, 72410 Puebla, Mexico
| | - Daniel Martinez-Fong
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Apartado Postal 14-740, 07000 México, DF, Mexico
| | - Juan-Antonio Gonzalez-Barrios
- Laboratorio de Medicina Genómica, Hospital Regional 1° de Octubre, ISSSTE, Avenida, Instituto Politécnico Nacional #1669, 07760 México DF, Mexico
| | - Samuel Treviño
- Facultad de Ciencias Químicas, Benemérita, Universidad Autónoma de Puebla, 14 sur y Av. San Claudio, Puebla, 72570 Puebla, Mexico
| | - Lourdes Millán-Perez Peña
- Centro de Química, ICUAP, Benemérita Universidad Autónoma de Puebla, 14 sur y Av. San Claudio, Puebla, 72570 Puebla, Mexico
| | - Victorino Alatriste
- Facultad de Ciencias Químicas, Benemérita, Universidad Autónoma de Puebla, 14 sur y Av. San Claudio, Puebla, 72570 Puebla, Mexico
| | - Guadalupe Soto-Rodriguez
- Facultad de Medicina, Benemérita Universidad Autónoma de Puebla, 13 sur 2702, Col. Volcanes, Puebla, 72410 Puebla, Mexico
| | - Eduardo Brambila
- Facultad de Ciencias Químicas, Benemérita, Universidad Autónoma de Puebla, 14 sur y Av. San Claudio, Puebla, 72570 Puebla, Mexico
| | - Bertha Alicia Leon-Chavez
- Facultad de Ciencias Químicas, Benemérita, Universidad Autónoma de Puebla, 14 sur y Av. San Claudio, Puebla, 72570 Puebla, Mexico
| |
Collapse
|
30
|
Pamies D, Sartori C, Schvartz D, González-Ruiz V, Pellerin L, Nunes C, Tavel D, Maillard V, Boccard J, Rudaz S, Sanchez JC, Zurich MG. Neuroinflammatory Response to TNFα and IL1β Cytokines Is Accompanied by an Increase in Glycolysis in Human Astrocytes In Vitro. Int J Mol Sci 2021; 22:4065. [PMID: 33920048 PMCID: PMC8071021 DOI: 10.3390/ijms22084065] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 01/11/2023] Open
Abstract
Astrogliosis has been abundantly studied in rodents but relatively poorly in human cells due to limited access to the brain. Astrocytes play important roles in cerebral energy metabolism, and are also key players in neuroinflammation. Astroglial metabolic and inflammatory changes as a function of age have been reported, leading to the hypothesis that mitochondrial metabolism and inflammatory responses are interconnected in supporting a functional switch of astrocytes from neurotrophic to neurotoxic. This study aimed to explore the metabolic changes occurring in astrocytes during their activation. Astrocytes were derived from human ReN cell neural progenitors and characterized. They were activated by exposure to tumor necrosis factor alpha (TNFα) or interleukin 1β (IL1β) for 24 h. Astrocyte reaction and associated energy metabolic changes were assessed by immunostaining, gene expression, proteomics, metabolomics and extracellular flux analyses. ReN-derived astrocytes reactivity was observed by the modifications of genes and proteins linked to inflammation (cytokines, nuclear factor-kappa B (NFκB), signal transducers and activators of transcription (STATs)) and immune pathways (major histocompatibility complex (MHC) class I). Increased NFκB1, NFκB2 and STAT1 expression, together with decreased STAT3 expression, suggest an activation towards the detrimental pathway. Strong modifications of astrocyte cytoskeleton were observed, including a glial fibrillary acidic protein (GFAP) decrease. Astrogliosis was accompanied by changes in energy metabolism characterized by increased glycolysis and lactate release. Increased glycolysis is reported for the first time during human astrocyte activation. Astrocyte activation is strongly tied to energy metabolism, and a possible association between NFκB signaling and/or MHC class I pathway and glycolysis is suggested.
Collapse
Affiliation(s)
- David Pamies
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland; (D.P.); (C.S.); (L.P.); (C.N.); (D.T.); (V.M.)
- Swiss Centre for Applied Human Toxicology (SCAHT), 4055 Basel, Switzerland; (D.S.); (V.G.-R.); (J.B.); (S.R.); (J.-C.S.)
| | - Chiara Sartori
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland; (D.P.); (C.S.); (L.P.); (C.N.); (D.T.); (V.M.)
| | - Domitille Schvartz
- Swiss Centre for Applied Human Toxicology (SCAHT), 4055 Basel, Switzerland; (D.S.); (V.G.-R.); (J.B.); (S.R.); (J.-C.S.)
- Translational Biomarker Group, Department of Internal Medicine Specialties, University of Geneva, CH-1211 Genève, Switzerland
| | - Víctor González-Ruiz
- Swiss Centre for Applied Human Toxicology (SCAHT), 4055 Basel, Switzerland; (D.S.); (V.G.-R.); (J.B.); (S.R.); (J.-C.S.)
- Analytical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland and School of Pharmaceutical Sciences, University of Geneva, CH-1211 Genève, Switzerland
| | - Luc Pellerin
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland; (D.P.); (C.S.); (L.P.); (C.N.); (D.T.); (V.M.)
- INSERM U1082, Faculté de Médecine et de Pharmacie, Université de Poitiers, F-86021 Poitiers, France
| | - Carolina Nunes
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland; (D.P.); (C.S.); (L.P.); (C.N.); (D.T.); (V.M.)
- Swiss Centre for Applied Human Toxicology (SCAHT), 4055 Basel, Switzerland; (D.S.); (V.G.-R.); (J.B.); (S.R.); (J.-C.S.)
| | - Denise Tavel
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland; (D.P.); (C.S.); (L.P.); (C.N.); (D.T.); (V.M.)
- Swiss Centre for Applied Human Toxicology (SCAHT), 4055 Basel, Switzerland; (D.S.); (V.G.-R.); (J.B.); (S.R.); (J.-C.S.)
| | - Vanille Maillard
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland; (D.P.); (C.S.); (L.P.); (C.N.); (D.T.); (V.M.)
- Swiss Centre for Applied Human Toxicology (SCAHT), 4055 Basel, Switzerland; (D.S.); (V.G.-R.); (J.B.); (S.R.); (J.-C.S.)
| | - Julien Boccard
- Swiss Centre for Applied Human Toxicology (SCAHT), 4055 Basel, Switzerland; (D.S.); (V.G.-R.); (J.B.); (S.R.); (J.-C.S.)
- Analytical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland and School of Pharmaceutical Sciences, University of Geneva, CH-1211 Genève, Switzerland
| | - Serge Rudaz
- Swiss Centre for Applied Human Toxicology (SCAHT), 4055 Basel, Switzerland; (D.S.); (V.G.-R.); (J.B.); (S.R.); (J.-C.S.)
- Analytical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland and School of Pharmaceutical Sciences, University of Geneva, CH-1211 Genève, Switzerland
| | - Jean-Charles Sanchez
- Swiss Centre for Applied Human Toxicology (SCAHT), 4055 Basel, Switzerland; (D.S.); (V.G.-R.); (J.B.); (S.R.); (J.-C.S.)
- Translational Biomarker Group, Department of Internal Medicine Specialties, University of Geneva, CH-1211 Genève, Switzerland
| | - Marie-Gabrielle Zurich
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland; (D.P.); (C.S.); (L.P.); (C.N.); (D.T.); (V.M.)
- Swiss Centre for Applied Human Toxicology (SCAHT), 4055 Basel, Switzerland; (D.S.); (V.G.-R.); (J.B.); (S.R.); (J.-C.S.)
| |
Collapse
|
31
|
Robinson JL, Yanes JA, Reid MA, Murphy JE, Busler JN, Mumford PW, Young KC, Pietrzkowski ZJ, Nemzer BV, Hunter JM, Beck DT. Neurophysiological Effects of Whole Coffee Cherry Extract in Older Adults with Subjective Cognitive Impairment: A Randomized, Double-Blind, Placebo-Controlled, Cross-Over Pilot Study. Antioxidants (Basel) 2021; 10:144. [PMID: 33498314 PMCID: PMC7909261 DOI: 10.3390/antiox10020144] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/10/2021] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Bioactive plant-based compounds have shown promise as protective agents across multiple domains including improvements in neurological and psychological measures. Methodological challenges have limited our understanding of the neurophysiological changes associated with polyphenol-rich supplements such as whole coffee cherry extract (WCCE). In the current study, we (1) compared 100 mg of WCCE to a placebo using an acute, randomized, double-blind, within-subject, cross-over design, and we (2) conducted a phytochemical analysis of WCCE. The primary objective of the study was to determine the neurophysiological and behavioral changes that resulted from the acute administration of WCCE. We hypothesized that WCCE would increase brain-derived neurotrophic factor (BDNF) and glutamate levels while also increasing neurofunctional measures in cognitive brain regions. Furthermore, we expected there to be increased behavioral performance associated with WCCE, as measured by reaction time and accuracy. Participants underwent four neuroimaging scans (pre- and post-WCCE and placebo) to assess neurofunctional/metabolic outcomes using functional magnetic resonance imaging and magnetic resonance spectroscopy. The results suggest that polyphenol-rich WCCE is associated with decreased reaction time and may protect against cognitive errors on tasks of working memory and response inhibition. Behavioral findings were concomitant with neurofunctional changes in structures involved in decision-making and attention. Specifically, we found increased functional connectivity between the anterior cingulate and regions involved in sensory and decision-making networks. Additionally, we observed increased BDNF and an increased glutamate/gamma-aminobutyric acid (GABA) ratio following WCCE administration. These results suggest that WCCE is associated with acute neurophysiological changes supportive of faster reaction times and increased, sustained attention.
Collapse
Affiliation(s)
- Jennifer L. Robinson
- Department of Psychology, Auburn University, Auburn, AL 36849, USA; (J.A.Y.); (J.E.M.); (J.N.B.)
- Auburn University MRI Research Center, Auburn University, Auburn, AL 36849, USA;
- Alabama Advanced Imaging Consortium, Auburn University, Auburn, AL 36849, USA
- Initiative for the Center for Neuroscience, Auburn University, Auburn, AL 36849, USA;
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL 36849, USA
| | - Julio A. Yanes
- Department of Psychology, Auburn University, Auburn, AL 36849, USA; (J.A.Y.); (J.E.M.); (J.N.B.)
- Auburn University MRI Research Center, Auburn University, Auburn, AL 36849, USA;
- Alabama Advanced Imaging Consortium, Auburn University, Auburn, AL 36849, USA
- Initiative for the Center for Neuroscience, Auburn University, Auburn, AL 36849, USA;
| | - Meredith A. Reid
- Auburn University MRI Research Center, Auburn University, Auburn, AL 36849, USA;
- Alabama Advanced Imaging Consortium, Auburn University, Auburn, AL 36849, USA
- Initiative for the Center for Neuroscience, Auburn University, Auburn, AL 36849, USA;
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL 36849, USA
| | - Jerry E. Murphy
- Department of Psychology, Auburn University, Auburn, AL 36849, USA; (J.A.Y.); (J.E.M.); (J.N.B.)
| | - Jessica N. Busler
- Department of Psychology, Auburn University, Auburn, AL 36849, USA; (J.A.Y.); (J.E.M.); (J.N.B.)
- Auburn University MRI Research Center, Auburn University, Auburn, AL 36849, USA;
- Alabama Advanced Imaging Consortium, Auburn University, Auburn, AL 36849, USA
- Initiative for the Center for Neuroscience, Auburn University, Auburn, AL 36849, USA;
| | - Petey W. Mumford
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA; (P.W.M.); (K.C.Y.)
| | - Kaelin C. Young
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA; (P.W.M.); (K.C.Y.)
- Edward Via College of Osteopathic Medicine, Auburn, AL 36830, USA
| | | | - Boris V. Nemzer
- VDF FutureCeuticals, Inc., 2692 N. State Route 1-17, Momence, IL 60954, USA; (B.V.N.); (J.M.H.)
| | - John M. Hunter
- VDF FutureCeuticals, Inc., 2692 N. State Route 1-17, Momence, IL 60954, USA; (B.V.N.); (J.M.H.)
| | - Darren T. Beck
- Initiative for the Center for Neuroscience, Auburn University, Auburn, AL 36849, USA;
- Edward Via College of Osteopathic Medicine, Auburn, AL 36830, USA
| |
Collapse
|
32
|
Thirupathi A, Pinho RA, Baker JS, István B, Gu Y. Taurine Reverses Oxidative Damages and Restores the Muscle Function in Overuse of Exercised Muscle. Front Physiol 2020; 11:582449. [PMID: 33192592 PMCID: PMC7649292 DOI: 10.3389/fphys.2020.582449] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
Exercise-induced oxidative stress is linked with the expression level of endogenous antioxidants, but these antioxidants cannot overcome all oxidative stress-related damages in the cells, particularly when cells are under physiological stress. Sometimes, compounds are needed for cellular function, which are produced/activated within the cells, and these compounds can be synthesized by performing exercise, especially high-performance exercise. Taurine is a sulfur-containing amino acid used for various physiological functions. However, its synthesis and accumulation under the oxidative environment may be compromised. Recently, we have shown that taurine level is increased during exercise performance with a decrease in oxidative damage in overused muscles. Other studies have also shown that short-term supplementation with taurine increased physiological performance during severe work intensities, suggesting the role of taurine in improving muscle performance during exercise. However, its precursor cysteine is used in the synthesis of other compounds like GSH and Coenzyme A, which are important for regulating the redox system and energy homeostasis. It is, therefore, important to understand whether taurine synthesis within the cells can blunt the activity of other compounds that are beneficial in preventing oxidative damage during intense exercise. Furthermore, it is important to understand whether taurine supplementation can prevent the conditions observed in the physiological stress of muscles. This review discusses how taurine synthesis could alter exercise-induced ROS generation and the relationship between the physiological stress of muscle and subsequent improvements in exercise performance.
Collapse
Affiliation(s)
| | - Ricardo A Pinho
- Laboratory of Exercise Biochemistry in Health, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Julien S Baker
- Department of Sport, Physical Education and Health, Hong Kong Baptist University, Hong Kong, China
| | - Bíró István
- Faculty of Engineering, University of Szeged, Szeged, Hungary
| | - Yaodong Gu
- Faculty of Sports Science, Ningbo University, Ningbo, China
| |
Collapse
|
33
|
Zhang F, Qi N, Zeng Y, Bao M, Chen Y, Liao J, Wei L, Cao D, Huang S, Luo Q, Jiang Y, Mo Z. The Endogenous Alterations of the Gut Microbiota and Feces Metabolites Alleviate Oxidative Damage in the Brain of LanCL1 Knockout Mice. Front Microbiol 2020; 11:557342. [PMID: 33117306 PMCID: PMC7575697 DOI: 10.3389/fmicb.2020.557342] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/12/2020] [Indexed: 12/26/2022] Open
Abstract
Altered composition of the gut microbiota has been observed in many neurodegenerative diseases. LanCL1 has been proven to protect neurons and reduce oxidative stress. The present study was designed to investigate alterations of the gut microbiota in LanCL1 knockout mice and to study the interactions between gut bacteria and the brain. Wild-type and LanCL1 knockout mice on a normal chow diet were evaluated at 4 and 8-9 weeks of age. 16s rRNA sequence and untargeted metabolomics analyses were performed to investigate changes in the gut microbiota and feces metabolites. Real-time polymerase chain reaction analysis, AB-PAS staining, and a TUNEL assay were performed to detect alterations in the gut and brain of knockout mice. The serum cytokines of 9-week-old knockout mice, which were detected by a multiplex cytokine assay, were significantly increased. In the central nervous system, there was no increase of antioxidant defense genes even though there was only low activity of glutathione S-transferase in the brain of 8-week-old knockout mice. Interestingly, the gut tight junctions, zonula occludens-1 and occludin, also displayed a downregulated expression level in 8-week-old knockout mice. On the contrary, the production of mucus increased in 8-week-old knockout mice. Moreover, the compositions of the gut microbiota and feces metabolites markedly changed in 8-week-old knockout mice but not in 4-week-old mice. Linear discriminant analysis and t-tests identified Akkermansia as a specific abundant bacteria in knockout mice. Quite a few feces metabolites that have protective effects on the brain were reduced in 8-week-old knockout mice. However, N-acetylsphingosine was the most significant downregulated feces metabolite, which may cause the postponement of neuronal apoptosis. To further investigate the effect of the gut microbiota, antibiotics treatment was given to both types of mice from 5 to 11 weeks of age. After treatment, a significant increase of oxidative damage in the brain of knockout mice was observed, which may have been alleviated by the gut microbiota before. In conclusion, alterations of the gut microbiota and feces metabolites alleviated oxidative damage to the brain of LanCL1 knockout mice, revealing that an endogenous feedback loop mechanism of the microbiota-gut-brain axis maintains systemic homeostasis.
Collapse
Affiliation(s)
- Fangxing Zhang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, China
- Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Nana Qi
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
| | - Yanyu Zeng
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
| | - Mengying Bao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
| | - Yang Chen
- Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinling Liao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
| | - Luyun Wei
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, China
| | - Dehao Cao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, China
| | - Shengzhu Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, China
| | - Qianqian Luo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, China
| | - Yonghua Jiang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
| |
Collapse
|
34
|
Qaradakhi T, Gadanec LK, McSweeney KR, Abraham JR, Apostolopoulos V, Zulli A. The Anti-Inflammatory Effect of Taurine on Cardiovascular Disease. Nutrients 2020; 12:E2847. [PMID: 32957558 PMCID: PMC7551180 DOI: 10.3390/nu12092847] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/02/2020] [Accepted: 09/12/2020] [Indexed: 12/12/2022] Open
Abstract
Taurine is a non-protein amino acid that is expressed in the majority of animal tissues. With its unique sulfonic acid makeup, taurine influences cellular functions, including osmoregulation, antioxidation, ion movement modulation, and conjugation of bile acids. Taurine exerts anti-inflammatory effects that improve diabetes and has shown benefits to the cardiovascular system, possibly by inhibition of the renin angiotensin system. The beneficial effects of taurine are reviewed.
Collapse
Affiliation(s)
- Tawar Qaradakhi
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia; (L.K.G.); (K.R.M.); (J.R.A.); (V.A.); (A.Z.)
| | | | | | | | | | | |
Collapse
|
35
|
Yuceli S, Yazici GN, Mammadov R, Suleyman H, Kaya M, Ozdogan S. The Effect of Rutin on Experimental Traumatic Brain Injury and Edema in Rats. In Vivo 2020; 34:2453-2460. [PMID: 32871772 DOI: 10.21873/invivo.12060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/12/2020] [Accepted: 06/16/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND/AIM The aim of the study was to evaluate the effect of rutin, which is a vitamin P1 flavonoid with anti-inflammatory and anti-edema effects, on traumatic brain injury (TBI) and edema in rats. MATERIALS AND METHODS Rats were divided into 3 groups as sham group without brain trauma (SG), brain trauma without medication (BT) group and Rutin treated brain trauma (RBT) group. Fifty mg/kg rutin was administered to the RBT group once a day for three days. On the fourth day, rats were sacrificed. Extracted brain tissues were examined biochemically and histopathologically. RESULTS We found that the levels of malondialdehyde, nuclear factor-kappa B and tumor necrosis factor-alpha decreased, and those of total glutathione increased significantly. Furthermore, rutin administration reduced pyramidal neuron degeneration and poly-morpho-nuclear-leucocyte accumulation due to trauma in brain tissue, while eliminating edema. CONCLUSION Rutin might be effective in the treatment of TBI and TBI-related brain edema.
Collapse
Affiliation(s)
- Sahin Yuceli
- Department of Neurosurgery, Neon Hospital, Erzincan, Turkey
| | - Gulce Naz Yazici
- Department of Histology and Embryology, Erzincan Binali Yildirim University School of Medicine, Erzincan, Turkey
| | - Renad Mammadov
- Department of Pharmacology, Erzincan Binali Yildirim University School of Medicine, Erzincan, Turkey
| | - Halis Suleyman
- Department of Pharmacology, Erzincan Binali Yildirim University School of Medicine, Erzincan, Turkey
| | - Mustafa Kaya
- Department of Neurosurgery, Sakarya Training and Research Hospital, Sakarya, Turkey
| | - Selcuk Ozdogan
- Department of Neurosurgery, Beykent University School of Medicine, Istanbul, Turkey
| |
Collapse
|
36
|
Mousavi K, Niknahad H, Ghalamfarsa A, Mohammadi H, Azarpira N, Ommati MM, Heidari R. Taurine mitigates cirrhosis-associated heart injury through mitochondrial-dependent and antioxidative mechanisms. Clin Exp Hepatol 2020; 6:207-219. [PMID: 33145427 PMCID: PMC7592093 DOI: 10.5114/ceh.2020.99513] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/09/2020] [Indexed: 12/15/2022] Open
Abstract
Cirrhosis-induced heart injury and cardiomyopathy is a serious consequence of this disease. It has been shown that bile duct ligated (BDL) animals could serve as an appropriate experimental model to investigate heart tissue injury in cirrhosis. The accumulation of cytotoxic chemicals (e.g., bile acids) could also adversely affect the heart tissue. Oxidative stress and mitochondrial impairment are the most prominent mechanisms of bile acid cytotoxicity. Taurine (Tau) is the most abundant non-protein amino acid in the human body. The cardioprotective effects of this amino acid have repeatedly been investigated. In the current study, it was examined whether mitochondrial dysfunction and oxidative stress are involved in the pathogenesis of cirrhosis-induced heart injury. Rats underwent BDL surgery. BDL animals received Tau (50, 100, and 500 mg/kg, i.p.) for 42 consecutive days. A significant increase in oxidative stress biomarkers was detected in the heart tissue of BDL animals. Moreover, it was found that heart tissue mitochondrial indices of functionality were deteriorated in the BDL group. Tau treatment significantly decreased oxidative stress and improved mitochondrial function in the heart tissue of cirrhotic animals. These data provide clues for the involvement of mitochondrial impairment and oxidative stress in the pathogenesis of heart injury in BDL rats. On the other hand, Tau supplementation could serve as an effective ancillary treatment against BDL-associated heart injury. Mitochondrial regulating and antioxidative properties of Tau might play a fundamental role in its mechanism of protective effects in the heart tissue of BDL animals.
Collapse
Affiliation(s)
- Khadijeh Mousavi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Niknahad
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amin Ghalamfarsa
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamidreza Mohammadi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
37
|
Josson Akkara P, Sabina EP. A biochemical approach to the anti-inflammatory, antioxidant and antiapoptotic potential of beta-carotene as a protective agent against bromobenzene-induced hepatotoxicity in female Wistar albino rats. J Appl Biomed 2020; 18:87-95. [PMID: 34907730 DOI: 10.32725/jab.2020.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 07/28/2020] [Indexed: 11/05/2022] Open
Abstract
Bromobenzene is a compound which has contributed much in understanding the mechanisms involved in xenobiotic hepatotoxicity induced by drugs and environment pollutants. In the present study, the protective and ameliorative effect of beta-carotene was investigated against bromobenzene-induced hepatotoxicity and compared with silymarin, a standard hepatoprotective reference drug. Beta-carotene (10 mg/kg b.w. p.o.) was administered to the rats for 9 days before intragastric intubation of bromobenzene (10 mmol/kg b.w.). Liver marker enzymes (aspartate transaminase, alanine transaminase and alkaline phosphatase), total protein content, bilirubin, total cholesterol, high-density lipoproteins, triglycerides, antioxidant status (reduced glutathione, superoxide dismutase, catalase, glutathione-S-transferase and glutathione peroxidase) were assessed along with histopathological analysis. ELISA was performed for analysing the levels of cytokines such as TNF-α, IL-1β and IL-6 in serum and in the liver. Caspase-3, COX-2 and NF-κB were evaluated by Western blotting. Administration of bromobenzene resulted in elevated levels of liver marker enzymes, bilirubin, lipid peroxidation and cytokines but deterioration in total protein content, antioxidant levels and histopathological conditions. Pre-treatment with beta-carotene not only significantly decreased the levels of liver markers, lipid peroxidation and cytokines but also improved histo-architecture and increased antioxidant levels minimising oxidative stress, and reduced factors contributing to apoptosis. This significant reversal of the biochemical changes on pre-treatment with beta-carotene in comparison with rats administered with bromobenzene clearly demonstrates that beta-carotene possesses promising hepatoprotective effect through its antioxidant, anti-inflammatory and antiapoptotic activity and hence is suggested as a potential therapeutic agent for protection from bromobenzene.
Collapse
Affiliation(s)
- Priya Josson Akkara
- Vellore Institute of Technology, School of Bio Sciences and Technology, Vellore, India.,Kristu Jayanti College (Autonomous), Bengaluru, India
| | - Evan Prince Sabina
- Vellore Institute of Technology, School of Bio Sciences and Technology, Vellore, India
| |
Collapse
|
38
|
Mersman B, Zaidi W, Syed NI, Xu F. Taurine Promotes Neurite Outgrowth and Synapse Development of Both Vertebrate and Invertebrate Central Neurons. Front Synaptic Neurosci 2020; 12:29. [PMID: 32792935 PMCID: PMC7387692 DOI: 10.3389/fnsyn.2020.00029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/24/2020] [Indexed: 12/13/2022] Open
Abstract
Taurine is a sulfur-containing amino acid that is widely expressed throughout the human brain, heart, retina, and muscle tissues. Taurine deficiency is associated with cardiomyopathy, renal dysfunction, abnormalities of the developing nervous system, and epilepsy which suggests a role specific to excitable tissues. Like vertebrates, invertebrates maintain high levels of taurine during embryonic and larval development, which decline during aging, indicating a potential developmental role. Notwithstanding its extensive presence throughout, taurine’s precise role/s during early brain development, function, and repair remains largely unknown in both vertebrate and invertebrate. Here, we investigated whether taurine affects neurite outgrowth, synapse formation, and synaptic transmission between postnatal day 0 rat cortical neurons in vitro, whereas its synaptogenic role was tested more directly using the Lymnaea soma-soma synapse model. We provide direct evidence that when applied at physiological concentrations, taurine exerts a significant neurotrophic effect on neuritic outgrowth and thickness of neurites as well as the expression of synaptic puncta as revealed by immunostaining of presynaptic synaptophysin and postsynaptic PSD95 proteins in rat cortical neurons, indicating direct involvement in synapse development. To demonstrate taurine’s direct effects on neurons in the absence of glia and other confounding factors, we next exploited individually identified pre- and postsynaptic neurons from the mollusk Lymnaea stagnalis. We found that taurine increased both the incidence of synapse formation (percent of cells that form synapses) and the efficacy of synaptic transmission between the paired neurons. This effect was comparable, but not additive, to Lymnaea trophic factor-induced synaptogenesis. This study thus provides direct morphological and functional evidence that taurine plays an important role in neurite outgrowth, synaptogenesis, and synaptic transmission during the early stages of brain development and that this role is conserved across both vertebrate and invertebrate species.
Collapse
Affiliation(s)
- Brittany Mersman
- Department of Biology, College of Arts and Sciences, Saint Louis University, St. Louis, MO, United States.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, United States
| | - Wali Zaidi
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Naweed I Syed
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Fenglian Xu
- Department of Biology, College of Arts and Sciences, Saint Louis University, St. Louis, MO, United States.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, United States
| |
Collapse
|
39
|
Seo M, Anderson G. Gut-Amygdala Interactions in Autism Spectrum Disorders: Developmental Roles via regulating Mitochondria, Exosomes, Immunity and microRNAs. Curr Pharm Des 2020; 25:4344-4356. [PMID: 31692435 DOI: 10.2174/1381612825666191105102545] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 11/01/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Autism Spectrum Disorders (ASD) have long been conceived as developmental disorder. A growing body of data highlights a role for alterations in the gut in the pathoetiology and/or pathophysiology of ASD. Recent work shows alterations in the gut microbiome to have a significant impact on amygdala development in infancy, suggesting that the alterations in the gut microbiome may act to modulate not only amygdala development but how the amygdala modulates the development of the frontal cortex and other brain regions. METHODS This article reviews wide bodies of data pertaining to the developmental roles of the maternal and foetal gut and immune systems in the regulation of offspring brain development. RESULTS A number of processes seem to be important in mediating how genetic, epigenetic and environmental factors interact in early development to regulate such gut-mediated changes in the amygdala, wider brain functioning and inter-area connectivity, including via regulation of microRNA (miR)-451, 14-3-3 proteins, cytochrome P450 (CYP)1B1 and the melatonergic pathways. As well as a decrease in the activity of monoamine oxidase, heightened levels of in miR-451 and CYP1B1, coupled to decreased 14-3-3 act to inhibit the synthesis of N-acetylserotonin and melatonin, contributing to the hyperserotonemia that is often evident in ASD, with consequences for mitochondria functioning and the content of released exosomes. These same factors are likely to play a role in regulating placental changes that underpin the association of ASD with preeclampsia and other perinatal risk factors, including exposure to heavy metals and air pollutants. Such alterations in placental and gut processes act to change the amygdala-driven biological underpinnings of affect-cognitive and affect-sensory interactions in the brain. CONCLUSION Such a perspective readily incorporates previously disparate bodies of data in ASD, including the role of the mu-opioid receptor, dopamine signaling and dopamine receptors, as well as the changes occurring to oxytocin and taurine levels. This has a number of treatment implications, the most readily applicable being the utilization of sodium butyrate and melatonin.
Collapse
Affiliation(s)
- Moonsang Seo
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - George Anderson
- CRC Scotland & London, Eccleston Square, London, United Kingdom
| |
Collapse
|
40
|
Expedition into Taurine Biology: Structural Insights and Therapeutic Perspective of Taurine in Neurodegenerative Diseases. Biomolecules 2020; 10:biom10060863. [PMID: 32516961 PMCID: PMC7355587 DOI: 10.3390/biom10060863] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/28/2020] [Accepted: 06/02/2020] [Indexed: 12/18/2022] Open
Abstract
Neurodegenerative diseases (NDs) are characterized by the accumulation of misfolded proteins. The hallmarks of protein aggregation in NDs proceed with impairment in the mitochondrial function, besides causing an enhancement in endoplasmic reticulum (ER) stress, neuroinflammation and synaptic loss. As accumulation of misfolded proteins hampers normal neuronal functions, it triggers ER stress, which leads to the activation of downstream effectors formulating events along the signaling cascade—referred to as unfolded protein response (UPRER) —thereby controlling cellular gene expression. The absence of disease-modifying therapeutic targets in different NDs, and the exponential increase in the number of cases, makes it critical to explore new approaches to treating these devastating diseases. In one such approach, osmolytes (low molecular weight substances), such as taurine have been found to promote protein folding under stress conditions, thereby averting aggregation of the misfolded proteins. Maintaining the structural integrity of the protein, taurine-mediated resumption of protein folding prompts a shift in folding homeostasis more towards functionality than towards aggregation and degradation. Together, taurine enacts protection in NDs by causing misfolded proteins to refold, so as to regain their stability and functionality. The present study provides recent and useful insights into understanding the progression of NDs, besides summarizing the genetics of NDs in correlation with mitochondrial dysfunction, ER stress, neuroinflammation and synaptic loss. It also highlights the structural and functional aspects of taurine in imparting protection against the aggregation/misfolding of proteins, thereby shifting the focus more towards the development of effective therapeutic modules that could avert the development of NDs.
Collapse
|
41
|
Salman M, Tabassum H, Parvez S. Tannic Acid Provides Neuroprotective Effects Against Traumatic Brain Injury Through the PGC-1α/Nrf2/HO-1 Pathway. Mol Neurobiol 2020; 57:2870-2885. [PMID: 32399817 DOI: 10.1007/s12035-020-01924-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/22/2020] [Indexed: 12/30/2022]
Abstract
The present research was conducted to elucidate a possible molecular mechanism related to neuromodulatory effects of tannic acid (TA) supplementation against traumatic brain injury (TBI) in a rodent model. Oxidative damage and neuroinflammation play a critical role in TBI and lead to behavioral alterations and neuronal dysfunction and death. These changes suggest a potential avenue in neurotherapeutic intervention. The aim of the present study was to investigate the neuroprotective effects of TA and potential mechanism of these effects in a controlled cortical impact injury model of TBI in Wistar rats that were treated with TA (50 mg/kg body weight. i.p.) before 30 min and 6 and 18 h after TBI. TBI-induced rats were examined after 24 h for behavioral dysfunction, Nissl stain, lipid peroxidation rate, glutathione level, activities of antioxidant enzymes (catalase, glutathione S-transferase, glutathione peroxidase, and superoxide dismutase), the expression level of 4-hydroxynonenal, pro-inflammatory cytokines such as tumor necrosis factor alpha and interleukin-1 beta, as well as brain edema and immunoreactivity of glial fibrillary acidic protein. Results indicated that TA supplementation significantly modulated above mentioned alterations. Moreover, TA treatment effectively upregulated the protein expression of peroxisome proliferator-activated receptor gamma co-activator 1 alpha (PGC-1α) and nuclear factor-E2-related factor-2 (Nrf2) as well as mitochondrial transcription factor A and heme oxygenase-1 (HO-1) following TBI. Overall, our results suggest that TA effectively ameliorates the behavioral alterations, oxidative damage, mitochondrial impairment, and inflammation against TBI that may be attributed to activation of PGC-1α/Nrf-2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Mohd Salman
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Heena Tabassum
- Division of Biomedical Sciences, Indian Council of Medical Research, Ministry of Health and Family Welfare, Govt. of India, V. Ramalingaswamy Bhawan, P.O. Box No. 4911, New Delhi, 110029, India.
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
42
|
Herzog N, Laager R, Thommen E, Widmer M, Vincent AM, Keller A, Becker C, Beck K, Perrig S, Bernasconi L, Neyer P, Marsch S, Schuetz P, Sutter R, Tisljar K, Hunziker S. Association of Taurine with In-Hospital Mortality in Patients after Out-of-Hospital Cardiac Arrest: Results from the Prospective, Observational COMMUNICATE Study. J Clin Med 2020; 9:jcm9051405. [PMID: 32397548 PMCID: PMC7290691 DOI: 10.3390/jcm9051405] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/01/2020] [Accepted: 05/05/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Studies have suggested that taurine may have neuro- and cardio-protective functions, but there is little research looking at taurine levels in patients after out-of-hospital cardiac arrest (OHCA). Our aim was to evaluate the association of taurine with mortality and neurological deficits in a well-defined cohort of OHCA patients. Methods: We prospectively measured serum taurine concentration in OHCA patients upon admission to the intensive care unit (ICU) of the University Hospital Basel (Switzerland). We analyzed the association of taurine levels and in-hospital mortality (primary endpoint). We further evaluated neurological outcomes assessed by the cerebral performance category scale. We calculated logistic regression analyses and report odds ratios (OR) and 95% confidence intervals (CI). We calculated different predefined multivariable regression models including demographic variables, comorbidities, initial vital signs, initial blood markers and resuscitation measures. We assessed discrimination by means of area under the receiver operating curve (ROC). Results: Of 240 included patients, 130 (54.2%) survived until hospital discharge and 110 (45.8%) had a favorable neurological outcome. Taurine levels were significantly associated with higher in-hospital mortality (adjusted OR 4.12 (95%CI 1.22 to 13.91), p = 0.02). In addition, a significant association between taurine concentration and a poor neurological outcome was observed (adjusted OR of 3.71 (95%CI 1.13 to 12.25), p = 0.03). Area under the curve (AUC) suggested only low discrimination for both endpoints (0.57 and 0.57, respectively). Conclusion: Admission taurine levels are associated with mortality and neurological outcomes in OHCA patients and may help in the risk assessment of this vulnerable population. Further studies are needed to assess whether therapeutic modulation of taurine may improve clinical outcomes after cardiac arrest.
Collapse
Affiliation(s)
- Naemi Herzog
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Rahel Laager
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Emanuel Thommen
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Madlaina Widmer
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Alessia M. Vincent
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Annalena Keller
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Christoph Becker
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
- Faculty of Medicine, University of Basel, 4031 Basel, Switzerland; (S.M.); (P.S.); (R.S.)
- Emergency Department, University Hospital Basel, 4031 Basel, Switzerland
| | - Katharina Beck
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Sebastian Perrig
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Kantonsspital Aarau, 5000 Aarau, Switzerland; (L.B.); (P.N.)
| | - Peter Neyer
- Institute of Laboratory Medicine, Kantonsspital Aarau, 5000 Aarau, Switzerland; (L.B.); (P.N.)
| | - Stephan Marsch
- Faculty of Medicine, University of Basel, 4031 Basel, Switzerland; (S.M.); (P.S.); (R.S.)
- Department of Intensive Care, University Hospital Basel, 4031 Basel, Switzerland;
| | - Philipp Schuetz
- Faculty of Medicine, University of Basel, 4031 Basel, Switzerland; (S.M.); (P.S.); (R.S.)
- Department of Internal Medicine, Kantonsspital Aarau, 5000 Aarau, Switzerland
| | - Raoul Sutter
- Faculty of Medicine, University of Basel, 4031 Basel, Switzerland; (S.M.); (P.S.); (R.S.)
- Department of Intensive Care, University Hospital Basel, 4031 Basel, Switzerland;
- Department of Neurology, University Hospital Basel, 4031 Basel, Switzerland
| | - Kai Tisljar
- Department of Intensive Care, University Hospital Basel, 4031 Basel, Switzerland;
| | - Sabina Hunziker
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
- Faculty of Medicine, University of Basel, 4031 Basel, Switzerland; (S.M.); (P.S.); (R.S.)
- Department of Intensive Care, University Hospital Basel, 4031 Basel, Switzerland;
- Correspondence: ; Tel.: +41-61-265-25-25
| |
Collapse
|
43
|
Chatterjee I, Lu R, Zhang Y, Zhang J, Dai Y, Xia Y, Sun J. Vitamin D receptor promotes healthy microbial metabolites and microbiome. Sci Rep 2020; 10:7340. [PMID: 32355205 PMCID: PMC7192915 DOI: 10.1038/s41598-020-64226-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/03/2020] [Indexed: 02/06/2023] Open
Abstract
Microbiota derived metabolites act as chemical messengers that elicit a profound impact on host physiology. Vitamin D receptor (VDR) is a key genetic factor for shaping the host microbiome. However, it remains unclear how microbial metabolites are altered in the absence of VDR. We investigated metabolites from mice with tissue-specific deletion of VDR in intestinal epithelial cells or myeloid cells. Conditional VDR deletion severely changed metabolites specifically produced from carbohydrate, protein, lipid, and bile acid metabolism. Eighty-four out of 765 biochemicals were significantly altered due to the Vdr status, and 530 significant changes were due to the high-fat diet intervention. The impact of diet was more prominent due to loss of VDR as indicated by the differences in metabolites generated from energy expenditure, tri-carboxylic acid cycle, tocopherol, polyamine metabolism, and bile acids. The effect of HFD was more pronounced in female mice after VDR deletion. Interestingly, the expression levels of farnesoid X receptor in liver and intestine were significantly increased after intestinal epithelial VDR deletion and were further increased by the high-fat diet. Our study highlights the gender differences, tissue specificity, and potential gut-liver-microbiome axis mediated by VDR that might trigger downstream metabolic disorders.
Collapse
Affiliation(s)
- Ishita Chatterjee
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | - Rong Lu
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | - Yongguo Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | - Jilei Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | - Yang Dai
- Department of Bioengineering, University of Illinois at Chicago, Chicago, USA
| | - Yinglin Xia
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, USA.
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, USA.
| |
Collapse
|
44
|
Winiarska-Mieczan A, Baranowska-Wójcik E, Kwiecień M, Grela ER, Szwajgier D, Kwiatkowska K, Kiczorowska B. The Role of Dietary Antioxidants in the Pathogenesis of Neurodegenerative Diseases and Their Impact on Cerebral Oxidoreductive Balance. Nutrients 2020; 12:nu12020435. [PMID: 32046360 PMCID: PMC7071337 DOI: 10.3390/nu12020435] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 01/26/2020] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases are progressive diseases of the nervous system that lead to neuron loss or functional disorders. Neurodegenerative diseases require long-term, sometimes life-long pharmacological treatment, which increases the risk of adverse effects and a negative impact of pharmaceuticals on the patients’ general condition. One of the main problems related to the treatment of this type of condition is the limited ability to deliver drugs to the brain due to their poor solubility, low bioavailability, and the effects of the blood-brain barrier. Given the above, one of the main objectives of contemporary scientific research focuses on the prevention of neurodegenerative diseases. As disorders related to the competence of the antioxidative system are a marker in all diseases of this type, the primary prophylactics should entail the use of exogenous antioxidants, particularly ones that can be used over extended periods, regardless of the patient’s age, and that are easily available, e.g., as part of a diet or as diet supplements. The paper analyzes the significance of the oxidoreductive balance in the pathogenesis of neurodegenerative diseases. Based on information published globally in the last 10 years, an analysis is also provided with regard to the impact of exogenous antioxidants on brain functions with respect to the prevention of this type of diseases.
Collapse
Affiliation(s)
- Anna Winiarska-Mieczan
- Department of Bromatology and Food Physiology, University of Life Sciences in Lublin, 20-950 Lublin, Poland; (M.K.); (E.R.G.); (K.K.); (B.K.)
- Correspondence: ; Tel.: +48-81-445-67-44; Fax: +48-81-53-335-49
| | - Ewa Baranowska-Wójcik
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, 20-950 Lublin, Poland; (E.B.-W.); (D.S.)
| | - Małgorzata Kwiecień
- Department of Bromatology and Food Physiology, University of Life Sciences in Lublin, 20-950 Lublin, Poland; (M.K.); (E.R.G.); (K.K.); (B.K.)
| | - Eugeniusz R. Grela
- Department of Bromatology and Food Physiology, University of Life Sciences in Lublin, 20-950 Lublin, Poland; (M.K.); (E.R.G.); (K.K.); (B.K.)
| | - Dominik Szwajgier
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, 20-950 Lublin, Poland; (E.B.-W.); (D.S.)
| | - Katarzyna Kwiatkowska
- Department of Bromatology and Food Physiology, University of Life Sciences in Lublin, 20-950 Lublin, Poland; (M.K.); (E.R.G.); (K.K.); (B.K.)
| | - Bożena Kiczorowska
- Department of Bromatology and Food Physiology, University of Life Sciences in Lublin, 20-950 Lublin, Poland; (M.K.); (E.R.G.); (K.K.); (B.K.)
| |
Collapse
|
45
|
Supinski GS, Wang L, Schroder EA, Callahan LAP. Taurine administration ablates sepsis induced diaphragm weakness. Respir Physiol Neurobiol 2019; 271:103289. [PMID: 31505275 DOI: 10.1016/j.resp.2019.103289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/23/2019] [Accepted: 09/05/2019] [Indexed: 12/15/2022]
Abstract
Infection induced diaphragm weakness is a major contributor to death and prolonged mechanical ventilation in critically ill patients. Infection induced muscle dysfunction is associated with activation of muscle proteolytic enzymes, and taurine is known to suppress proteolysis. We therefore postulated that taurine administration may prevent infection induced diaphragm dysfunction. The purpose of this study was to test this hypothesis using a clinically relevant animal model of infection, i.e. cecal ligation puncture induced sepsis (CLP). Studies were performed on (n = 5-7 mice/group): (a) sham operated controls, (b) animals with sepsis induced by CLP, (c) sham operated animals given taurine (75 mg/kg/d, intraperitoneally), and (d) CLP animals given taurine. At intervals after surgery animals were euthanized, diaphragm force generation measured in vitro, and diaphragm calpain, caspase and proteasomal activity determined. CLP elicited a large reduction in diaphragm specific force generation at 24 h (1-150 Hz, p < 0.001) and taurine significantly attenuated CLP induced diaphragm weakness at all stimulation frequencies (p < 0.001). CLP induced significant increases in diaphragm calpain, caspase and proteasomal activity; taurine administration prevented increases in the activity of all three pathways. In additional time course experiments, diaphragm force generation remained at control levels over 72 h in CLP animals treated with daily taurine administration, while CLP animals demonstrated severe, sustained reductions in diaphragm strength (p < 0.01 for all time points). Our results indicate that taurine administration prevents infection induced diaphragm weakness and reduces activation of three major proteolytic pathways. Because this agent is has been shown to be safe, non-toxic when administered to humans, taurine may have a role in treating infection induced diaphragm weakness. Future clinical studies will be needed to assess this possibility.
Collapse
Affiliation(s)
- Gerald S Supinski
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kentucky, Lexington, KY, United States
| | - Lin Wang
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kentucky, Lexington, KY, United States
| | - Elizabeth A Schroder
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kentucky, Lexington, KY, United States
| | - Leigh Ann P Callahan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
46
|
Jakaria M, Azam S, Haque ME, Jo SH, Uddin MS, Kim IS, Choi DK. Taurine and its analogs in neurological disorders: Focus on therapeutic potential and molecular mechanisms. Redox Biol 2019; 24:101223. [PMID: 31141786 PMCID: PMC6536745 DOI: 10.1016/j.redox.2019.101223] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/21/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022] Open
Abstract
Taurine is a sulfur-containing amino acid and known as semi-essential in mammals and is produced chiefly by the liver and kidney. It presents in different organs, including retina, brain, heart and placenta and demonstrates extensive physiological activities within the body. In the several disease models, it attenuates inflammation- and oxidative stress-mediated injuries. Taurine also modulates ER stress, Ca2+ homeostasis and neuronal activity at the molecular level as part of its broader roles. Different cellular processes such as energy metabolism, gene expression, osmosis and quality control of protein are regulated by taurine. In addition, taurine displays potential ameliorating effects against different neurological disorders such as neurodegenerative diseases, stroke, epilepsy and diabetic neuropathy and protects against injuries and toxicities of the nervous system. Several findings demonstrate its therapeutic role against neurodevelopmental disorders, including Angelman syndrome, Fragile X syndrome, sleep-wake disorders, neural tube defects and attention-deficit hyperactivity disorder. Considering current biopharmaceutical limitations, developing novel delivery approaches and new derivatives and precursors of taurine may be an attractive option for treating neurological disorders. Herein, we present an overview on the therapeutic potential of taurine against neurological disorders and highlight clinical studies and its molecular mechanistic roles. This article also addresses the neuropharmacological potential of taurine analogs.
Collapse
Affiliation(s)
- Md Jakaria
- Department of Applied Life Sciences and Integrated Bioscience, Graduate School, Konkuk University, Chungju, South Korea
| | - Shofiul Azam
- Department of Applied Life Sciences and Integrated Bioscience, Graduate School, Konkuk University, Chungju, South Korea
| | - Md Ezazul Haque
- Department of Applied Life Sciences and Integrated Bioscience, Graduate School, Konkuk University, Chungju, South Korea
| | - Song-Hee Jo
- Department of Applied Life Sciences and Integrated Bioscience, Graduate School, Konkuk University, Chungju, South Korea
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - In-Su Kim
- Department of Applied Life Sciences and Integrated Bioscience, Graduate School, Konkuk University, Chungju, South Korea; Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Sciences, and Research Institute of Inflammatory Diseases (RID), Konkuk University, Chungju, South Korea
| | - Dong-Kug Choi
- Department of Applied Life Sciences and Integrated Bioscience, Graduate School, Konkuk University, Chungju, South Korea; Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Sciences, and Research Institute of Inflammatory Diseases (RID), Konkuk University, Chungju, South Korea.
| |
Collapse
|