1
|
Rodella P, Boreski D, Luz MAM, Gabriel EA, Takase LF, Chin CM. Taurine Neuroprotection and Neurogenesis Effect in Chronic Ethanol-Induced Rats. Nutrients 2024; 16:1973. [PMID: 38931326 PMCID: PMC11206532 DOI: 10.3390/nu16121973] [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: 05/31/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Taurine (2-aminoethanesulfonic acid) is a non-protein β-amino acid essential for cellular homeostasis, with antioxidant, anti-inflammatory, and cytoprotective properties that are crucial for life maintenance. This study aimed to evaluate the effects of taurine administration on hippocampal neurogenesis, neuronal preservation, or reverse damage in rats exposed to forced ethanol consumption in an animal model. Wistar rats were treated with ethanol (EtOH) for a 28-day period (5% in the 1st week, 10% in the 2nd week, and 20% in the 3rd and 4th weeks). Two taurine treatment protocols (300 mg/kg i.p.) were implemented: one during ethanol consumption to analyze neuroprotection, and another after ethanol consumption to assess the reversal of ethanol-induced damage. Overall, the results demonstrated that taurine treatment was effective in protecting against deficits induced by ethanol consumption in the dentate gyrus. The EtOH+TAU group showed a significant increase in cell proliferation (145.8%) and cell survival (54.0%) compared to the EtOH+Sal group. The results also indicated similar effects regarding the reversal of ethanol-induced damage 28 days after the cessation of ethanol consumption. The EtOH+TAU group exhibited a significant increase (41.3%) in the number of DCX-immunoreactive cells compared to the EtOH+Sal group. However, this amino acid did not induce neurogenesis in the tissues of healthy rats, implying that its activity may be contingent upon post-injury stimuli.
Collapse
Affiliation(s)
- Patricia Rodella
- Laboratory for Drug Design (LAPDESF), School of Pharmaceutical Sciences, University of São Paulo State (UNESP), Araraquara 14800-903, Brazil; (P.R.); (D.B.)
| | - Diogo Boreski
- Laboratory for Drug Design (LAPDESF), School of Pharmaceutical Sciences, University of São Paulo State (UNESP), Araraquara 14800-903, Brazil; (P.R.); (D.B.)
| | - Marcus Alexandre Mendes Luz
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, Brazil; (M.A.M.L.); (E.A.G.)
| | - Edmo Atique Gabriel
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, Brazil; (M.A.M.L.); (E.A.G.)
| | - Luiz Fernando Takase
- Morphology and Pathology Department, Federal University of São Paulo of São Carlos (UFSCar), São Carlos 13565-905, Brazil;
| | - Chung Man Chin
- Laboratory for Drug Design (LAPDESF), School of Pharmaceutical Sciences, University of São Paulo State (UNESP), Araraquara 14800-903, Brazil; (P.R.); (D.B.)
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, Brazil; (M.A.M.L.); (E.A.G.)
| |
Collapse
|
2
|
Wang QH, Wu RX, Ji JN, Zhang J, Niu SF, Tang BG, Miao BB, Liang ZB. Integrated Transcriptomics and Metabolomics Reveal Changes in Cell Homeostasis and Energy Metabolism in Trachinotus ovatus in Response to Acute Hypoxic Stress. Int J Mol Sci 2024; 25:1054. [PMID: 38256129 PMCID: PMC10815975 DOI: 10.3390/ijms25021054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Trachinotus ovatus is an economically important mariculture fish, and hypoxia has become a critical threat to this hypoxia-sensitive species. However, the molecular adaptation mechanism of T. ovatus liver to hypoxia remains unclear. In this study, we investigated the effects of acute hypoxic stress (1.5 ± 0.1 mg·L-1 for 6 h) and re-oxygenation (5.8 ± 0.3 mg·L-1 for 12 h) in T. ovatus liver at both the transcriptomic and metabolic levels to elucidate hypoxia adaptation mechanism. Integrated transcriptomics and metabolomics analyses identified 36 genes and seven metabolites as key molecules that were highly related to signal transduction, cell growth and death, carbohydrate metabolism, amino acid metabolism, and lipid metabolism, and all played key roles in hypoxia adaptation. Of these, the hub genes FOS and JUN were pivotal hypoxia adaptation biomarkers for regulating cell growth and death. During hypoxia, up-regulation of GADD45B and CDKN1A genes induced cell cycle arrest. Enhancing intrinsic and extrinsic pathways in combination with glutathione metabolism triggered apoptosis; meanwhile, anti-apoptosis mechanism was activated after hypoxia. Expression of genes related to glycolysis, gluconeogenesis, amino acid metabolism, fat mobilization, and fatty acid biosynthesis were up-regulated after acute hypoxic stress, promoting energy supply. After re-oxygenation for 12 h, continuous apoptosis favored cellular function and tissue repair. Shifting from anaerobic metabolism (glycolysis) during hypoxia to aerobic metabolism (fatty acid β-oxidation and TCA cycle) after re-oxygenation was an important energy metabolism adaptation mechanism. Hypoxia 6 h was a critical period for metabolism alteration and cellular homeostasis, and re-oxygenation intervention should be implemented in a timely way. This study thoroughly examined the molecular response mechanism of T. ovatus under acute hypoxic stress, which contributes to the molecular breeding of hypoxia-tolerant cultivars.
Collapse
Affiliation(s)
- Qing-Hua Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; (Q.-H.W.); (R.-X.W.); (J.-N.J.); (J.Z.); (B.-G.T.); (B.-B.M.); (Z.-B.L.)
| | - Ren-Xie Wu
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; (Q.-H.W.); (R.-X.W.); (J.-N.J.); (J.Z.); (B.-G.T.); (B.-B.M.); (Z.-B.L.)
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang 524025, China
| | - Jiao-Na Ji
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; (Q.-H.W.); (R.-X.W.); (J.-N.J.); (J.Z.); (B.-G.T.); (B.-B.M.); (Z.-B.L.)
| | - Jing Zhang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; (Q.-H.W.); (R.-X.W.); (J.-N.J.); (J.Z.); (B.-G.T.); (B.-B.M.); (Z.-B.L.)
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang 524025, China
| | - Su-Fang Niu
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; (Q.-H.W.); (R.-X.W.); (J.-N.J.); (J.Z.); (B.-G.T.); (B.-B.M.); (Z.-B.L.)
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang 524025, China
| | - Bao-Gui Tang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; (Q.-H.W.); (R.-X.W.); (J.-N.J.); (J.Z.); (B.-G.T.); (B.-B.M.); (Z.-B.L.)
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang 524025, China
| | - Ben-Ben Miao
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; (Q.-H.W.); (R.-X.W.); (J.-N.J.); (J.Z.); (B.-G.T.); (B.-B.M.); (Z.-B.L.)
| | - Zhen-Bang Liang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; (Q.-H.W.); (R.-X.W.); (J.-N.J.); (J.Z.); (B.-G.T.); (B.-B.M.); (Z.-B.L.)
| |
Collapse
|
3
|
Pradhan LK, Sahoo PK, Sarangi P, Chauhan NR, Das SK. Suppression of Chronic Unpredictable Stress-Persuaded Increased Monoamine Oxidase Activity by Taurine Promotes Significant Neuroprotection in Zebrafish Brain. Neurochem Res 2023; 48:82-95. [PMID: 36001190 DOI: 10.1007/s11064-022-03724-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 01/11/2023]
Abstract
Neuropsychiatric upshots following chronic exposure to unpredictable adverse stressors have been well documented in the literature. Considering the significant impact of chronic unpredictable stress (CUS), the literature is elusive regarding the neuroprotective efficacy of taurine against CUS-induced oxidative stress and chromatin condensation in the zebrafish brain. In this study, to ameliorate CUS-persuaded neurological outcomes, waterborne treatment of taurine as a prophylactic intervention was undertaken. Further, our approach also focused on the gross neurobehavioral response of zebrafish, oxidative stress indices and neuromorphology of the zebrafish brain following CUS exposure with taurine treatment. Because taurine provides significant neuroprotection against oxidative insult, the cytosolic level of monoamine oxidase (MAO) in the zebrafish brain following CUS exposure is worth investigating. Further, as heightened MAO activity is associated with augmented oxidative and chromatin condensation, the focus of this study was on whether taurine provides neuroprotection by downregulating MAO levels in the brain. Our findings show that CUS-persuaded altered neurobehavioral response was significantly rescued by taurine. Moreover, our findings firmly support the hypothesis that taurine acts as a radical neuroprotector by restoring glutathione biosynthesis in the zebrafish brain subsequent to CUS exposure. Additionally, the rising level of brain MAO following chronic exposure to CUS is ameliorated by taurine treatment. These findings strongly advocate the role of taurine as a natural MAO inhibitor through the neuroprotection it provides against CUS-instigated oxidative stress in zebrafish. However, the fundamental neuroprotective mechanism of such natural compounds needs to be elucidated to determine their neuroprotective efficacy against stress regimens.
Collapse
Affiliation(s)
- Lilesh Kumar Pradhan
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Kalinga Nagar, Bhubaneswar, 751003, India
| | - Pradyumna Kumar Sahoo
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Kalinga Nagar, Bhubaneswar, 751003, India
| | - Prerana Sarangi
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Kalinga Nagar, Bhubaneswar, 751003, India
| | - Nishant Ranjan Chauhan
- Infectious Disease Biology Division, Institute of Life Sciences, Bhubaneswar, 751023, India
| | - Saroj Kumar Das
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Kalinga Nagar, Bhubaneswar, 751003, India.
| |
Collapse
|
4
|
Rafiee Z, García-Serrano AM, Duarte JMN. Taurine Supplementation as a Neuroprotective Strategy upon Brain Dysfunction in Metabolic Syndrome and Diabetes. Nutrients 2022; 14:1292. [PMID: 35334949 PMCID: PMC8952284 DOI: 10.3390/nu14061292] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/07/2023] Open
Abstract
Obesity, type 2 diabetes, and their associated comorbidities impact brain metabolism and function and constitute risk factors for cognitive impairment. Alterations to taurine homeostasis can impact a number of biological processes, such as osmolarity control, calcium homeostasis, and inhibitory neurotransmission, and have been reported in both metabolic and neurodegenerative disorders. Models of neurodegenerative disorders show reduced brain taurine concentrations. On the other hand, models of insulin-dependent diabetes, insulin resistance, and diet-induced obesity display taurine accumulation in the hippocampus. Given the possible cytoprotective actions of taurine, such cerebral accumulation of taurine might constitute a compensatory mechanism that attempts to prevent neurodegeneration. The present article provides an overview of brain taurine homeostasis and reviews the mechanisms by which taurine can afford neuroprotection in individuals with obesity and diabetes. We conclude that further research is needed for understanding taurine homeostasis in metabolic disorders with an impact on brain function.
Collapse
Affiliation(s)
- Zeinab Rafiee
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22100 Lund, Sweden; (Z.R.); (A.M.G.-S.)
- Wallenberg Centre for Molecular Medicine, Lund University, 22100 Lund, Sweden
| | - Alba M. García-Serrano
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22100 Lund, Sweden; (Z.R.); (A.M.G.-S.)
- Wallenberg Centre for Molecular Medicine, Lund University, 22100 Lund, Sweden
| | - João M. N. Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22100 Lund, Sweden; (Z.R.); (A.M.G.-S.)
- Wallenberg Centre for Molecular Medicine, Lund University, 22100 Lund, Sweden
| |
Collapse
|
5
|
Amirshahrokhi K, Niapour A. Methylsulfonylmethane protects against ethanol-induced brain injury in mice through the inhibition of oxidative stress, proinflammatory mediators and apoptotic cell death. Int Immunopharmacol 2022; 106:108638. [PMID: 35203043 DOI: 10.1016/j.intimp.2022.108638] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/04/2022] [Accepted: 02/16/2022] [Indexed: 11/17/2022]
Abstract
Excessive ethanol consumption causes brain injury through oxidative stress, inflammation and apoptotic cell death. Methylsulfonylmethane (MSM) is a natural compound that has therapeutic effects on oxidative and inflammatory disorders. The aim of this study was to investigate the protective effect and underlying mechanisms of MSM on ethanol-induced brain injury in an experimental model. Male C57BL/6 mice were exposed to binge ethanol (5 g/kg/day, orally) and treated with MSM (200 and 400 mg/kg/day) concomitantly for 12 days. At the end of the experiment brain tissues were removed for histological and biochemical analysis. The results showed that MSM reduced ethanol-mediated oxidative stress by decreasing the levels of malondialdehyde (MDA) and carbonyl protein. The Nrf2/HO-1 pathway and the levels of cytoprotective antioxidants superoxide dismutase (SOD), catalase and glutathione (GSH) were increased by MSM in the brain tissue. MSM treatment reduced the ethanol-induced inflammatory factors including myeloperoxidase (MPO), iNOS/NO, cyclooxygenase (COX)-2, nuclear factor kappa B (NF-κB), NLRP3 inflammasome and proinflammatory cytokines including TNF-α, IL-1β, IL-6 and MCP-1. MSM also decreased the levels of pro-apoptotic caspase-3 and TUNEL positive cells while increased the level of anti-apoptotic Bcl-2 in the brain tissue. Our findings demonstrated that MSM protects against ethanol-induced brain injury by improving anti-oxidant defense mechanism and reducing ethanol-mediated inflammation and apoptosis. Therefore, MSM may be a potential protective approach for brain damage caused by high levels of alcohol.
Collapse
Affiliation(s)
- Keyvan Amirshahrokhi
- Department of Pharmacology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Ali Niapour
- Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
6
|
Differences Between Physiological and Pharmacological Actions of Taurine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1370:311-321. [DOI: 10.1007/978-3-030-93337-1_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
7
|
Wang X, Liu J, Hui X, Song Y. Metabolomics Applied to Cord Serum in Preeclampsia Newborns: Implications for Neonatal Outcomes. Front Pediatr 2022; 10:869381. [PMID: 35547553 PMCID: PMC9082809 DOI: 10.3389/fped.2022.869381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/11/2022] [Indexed: 11/17/2022] Open
Abstract
Preeclampsia (PE) is one of the leading causes of maternal and perinatal morbidity and mortality. However, it is still uncertain how PE affects neonate metabolism. We conducted an untargeted metabolomics analysis of cord blood to explore the metabolic changes in PE neonates. Umbilical cord serum samples from neonates with preeclampsia (n = 29) and non-preeclampsia (non-PE) (n = 32) pregnancies were analyzed using the UHPLC-QE-MS metabolomic platform. Different metabolites were screened, and pathway analysis was conducted. A subgroup analysis was performed among PE neonates to compare the metabolome between appropriate-for-gestational-age infants (n = 21) and small-for-gestational-age (SGA) infants (n = 8). A total of 159 different metabolites were detected in PE and non-PE neonates. Creatinine, N4-acetylcytidine, sphingomyelin (D18:1/16:0), pseudouridine, uric acid, and indolelactic acid were the most significant differential metabolites in the cord serum of PE neonates. Differential metabolite levels were elevated in PE neonates and were involved in the following metabolic pathways: glycine, serine, and threonine metabolism; sphingolipid, glyoxylate, and dicarboxylate metabolism; and arginine biosynthesis. In PE neonates, SGA neonates showed increased levels of hexacosanoyl carnitine and decreased abundance of 3-hydroxybutyric acid and 3-sulfinoalanine. Taurine-related metabolism and ketone body-related pathways were mainly affected. Based on the UHPLC-QE-MS metabolomics analysis, we identified the metabolic profiles of PE and SGA neonates. The abundance of metabolites related to certain amino acid, sphingolipid, and energy metabolism increased in the umbilical cord serum of PE neonates.
Collapse
Affiliation(s)
- Xiaoxu Wang
- Department of Obstetrics and Gynecology, National Clinical Research Centre for Obstetric and Gynecologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jieying Liu
- State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangyi Hui
- State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingna Song
- Department of Obstetrics and Gynecology, National Clinical Research Centre for Obstetric and Gynecologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
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
|
9
|
Liu J, Yuan J, Zhao J, Zhang L, Wang Q, Wang G. Serum metabolomic patterns in young patients with ischemic stroke: a case study. Metabolomics 2021; 17:24. [PMID: 33554271 DOI: 10.1007/s11306-021-01774-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 01/22/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Ischemic stroke is one of the leading causes of death and adult disability. The incidence of ischemic stroke continues to rise in young adults. This study aimed to provide a comprehensive evaluation of metabolic changes and explore possible mechanisms in young ischemic stroke patients without common risk factors. METHODS This study investigated serum metabolomics in 50 young patients with newly suffered ischemic stroke and 50 age-, sex-, and body mass index-matched healthy controls. Liquid chromatography coupled with a Waters Xevo TQ-S mass spectrometer with an electrospray ionization (ESI) source was used to analyze amino acid or bile acid, and free fatty acid or lipid was analyzed by liquid chromatography coupled with a Qtrap5500 mass spectrometer with an ESI source. The metabolomic data were analyzed by performing a multivariate statistical analysis. RESULTS A total of 197 metabolites, including amino acids, bile acids, free fatty acids, and lipids, were identified in all participants. Multivariate models showed significant differences in serum metabolomic patterns between young patients with ischemic stroke and healthy controls. The stroke patients had increased L-methionine, homocysteine, glutamine, uric acid, GCDCA, and PE (18:0/20:4, 16:0/22:5), and decreased levels of L-citrulline, taurine, PC (16:2/22:6, 16:2/20:5, 15:0/18:2), and SM (d18:1/23:0, d20:0/19:1, d18:1/22:0, d16:0/26:1, d16:0/18:0, d16:0/22:1, d18:1/19:1, d16:0/17:1, d16:1/24:1, d18:1/19:0). Based on the identified metabolites, the metabolic pathways of arginine biosynthesis, glycerophospholipid metabolism, and taurine and hypotaurine metabolism were significantly enriched in the young patients with ischemic stroke. CONCLUSIONS Serum metabolomic patterns were significantly different between young patients with ischemic stroke and healthy controls. Our study is beneficial in providing a further view into the pathophysiology of young patients with ischemic stroke.
Collapse
Affiliation(s)
- Jia Liu
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Junliang Yuan
- Department of Neurology, Peking University Sixth Hospital, Beijing, 100191, China
| | - Jingwei Zhao
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Lin Zhang
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Qiu Wang
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Guang Wang
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
10
|
Iezhitsa I, Agarwal R. New solutions for old challenges in glaucoma treatment: is taurine an option to consider? Neural Regen Res 2021; 16:967-971. [PMID: 33229737 PMCID: PMC8178787 DOI: 10.4103/1673-5374.297059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glaucoma is a range of progressive optic neuropathies characterized by progressive retinal ganglion cell loss and visual field defects. It is recognized as a leading cause of irreversible blindness affecting more than 70 million people worldwide. Currently, reduction of intraocular pressure, a widely recognized risk factor for glaucoma development, is the only pharmacological strategy for slowing down retinal ganglion cell loss and disease progression. However, retinal ganglion cell death and visual field loss have been observed in normotensive glaucoma, suggesting that the disease process is partially independent of intraocular pressure. Taurine is one of the agents that have attracted attention of researchers recently. Taurine has been shown to be involved in multiple cellular functions, including a central role as a neurotransmitter, as a trophic factor in the central nervous system development, as an osmolyte, as a neuromodulator, and as a neuroprotectant. It also plays a role in the maintenance of the structural integrity of the membranes and in the regulation of calcium transport and homeostasis. Taurine is known to prevent N-methyl-D-aspartic acid-induced excitotoxic injury to retinal ganglion cells. A recently published study clearly demonstrated that taurine prevents retinal neuronal apoptosis both in vivo and in vitro. Protective effect of taurine may be attributed to direct inhibition of apoptosis, an activation of brain derived neurotrophic factor-related neuroprotective mechanisms and reduction of retinal oxidative and nitrosative stresses. Further studies are needed to fully explore the potential of taurine as a neuroprotective agent, so that it can be applied in clinical practice, particularly for the treatment of glaucoma. The objective of current review was to summarize recent evidence on neuroprotective properties of taurine in glaucoma.
Collapse
Affiliation(s)
- Igor Iezhitsa
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia; Volgograd State Medical University, Research Centre for Innovative Medicines, Volgograd, Russian Federation
| | - Renu Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
11
|
Lv K, Yang C, Xiao R, Yang L, Liu T, Zhang R, Fan X. Dexmedetomidine attenuates ethanol-induced inhibition of hippocampal neurogenesis in neonatal mice. Toxicol Appl Pharmacol 2020; 390:114881. [PMID: 31954762 DOI: 10.1016/j.taap.2020.114881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND/AIMS Ethanol (EtOH) exposure during a period comparable to the third trimester in human results in obvious neurotoxicity in the developing hippocampus and persistent deficits in hippocampal neurogenesis. Dexmedetomidine (DEX), a highly selective α-2-adrenergic agonist has been demonstrated to restore the impaired neurogenesis and neuronal plasticity in the dentate gyrus (DG) that follows neurological insult. However, the protective roles of DEX in the EtOH-induced deficits of postnatal neurogenesis in the hippocampus are still unknown. METHODS Mice were pretreated with DEX prior to EtOH exposure to determine its protective effects on impaired postnatal hippocampal neurogenesis. Six-day-old neonatal mice were treated with DEX (125 μg/kg) or saline, followed by EtOH at a total of 5 g/kg or an equivalent volume of saline on P7. Immunohistochemistry and immunofluorescence were used to evaluate the neurogenesis and activated microglia in the DG. Quantitative real time PCR (qRT-PCR) was utilized to assess the expression of inflammatory factors in the hippocampus. RESULTS DEX pretreatment attenuated the inhibition of EtOH-mediated hippocampal neurogenesis and the reduction of hippocampal neural precursor cells (NPCs). We further confirmed that DEX pretreatment reversed the EtOH-induced microglia activation in the DG as well as the upregulation of the hippocampal TNFα, MCP-1, IL-6, and IL-1β mRNA levels. CONCLUSION Our findings indicate that DEX pretreatment protects against EtOH-mediated inhibition of hippocampal neurogenesis in postnatal mice and reverses EtOH-induced neuroinflammation via repressing microglia activation and the expression of inflammatory cytokines.
Collapse
Affiliation(s)
- Keyi Lv
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China
| | - Congwen Yang
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Rui Xiao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China
| | - Ling Yang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China
| | - Tianyao Liu
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China
| | - Ruiyu Zhang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
12
|
Ommati MM, Heidari R, Ghanbarinejad V, Abdoli N, Niknahad H. Taurine Treatment Provides Neuroprotection in a Mouse Model of Manganism. Biol Trace Elem Res 2019; 190:384-395. [PMID: 30357569 DOI: 10.1007/s12011-018-1552-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/15/2018] [Indexed: 12/11/2022]
Abstract
Manganese (Mn) is a trace element involved in many physiological processes. However, excessive Mn exposure leads to neurological complications. Although no precise mechanism(s) has been found for Mn-induced neurotoxicity, oxidative stress and mitochondrial injury seem to play a relevant role in this complication. On the other hand, there is no protective strategy against Mn neurotoxicity so far. Taurine is an amino acid with significant neuroprotective properties. The current study was designed to evaluate the effect of taurine supplementation and its potential mechanism(s) of action in a mouse model of manganism. Animals were treated with Mn (100 mg/kg, s.c) alone and/or in combination with taurine (50, 100, and 500 mg/kg, i.p, for eight consecutive days). Severe locomotor dysfunction along with a significant elevation in brain tissue biomarkers of oxidative stress was evident in Mn-exposed mice. On the other hand, it was revealed that mitochondrial indices of functionality were hampered in Mn-treated animals. Taurine supplementation (50, 100, and 500 mg/kg, i.p) alleviated Mn-induced locomotor deficit. Moreover, this amino acid mitigated oxidative stress biomarkers and preserved brain tissue mitochondrial indices of functionality. These data introduce taurine as a potential neuroprotective agent against Mn neurotoxicity. Antioxidative and mitochondria protecting effects of taurine might play a fundamental role in its neuroprotective properties against Mn toxicity.
Collapse
Affiliation(s)
- Mohammad Mehdi Ommati
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P. O. Box 158371345, Roknabad, Karafarin St., Shiraz, Fars, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P. O. Box 158371345, Roknabad, Karafarin St., Shiraz, Fars, Iran.
| | - Vahid Ghanbarinejad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P. O. Box 158371345, Roknabad, Karafarin St., Shiraz, Fars, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Narges Abdoli
- Iran Food and Drug Administration (IFDA), Ministry of Health, Tehran, Iran
| | - Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P. O. Box 158371345, Roknabad, Karafarin St., Shiraz, Fars, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
13
|
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
|
14
|
Li Y, Li S, Xie X, Xiu H, Liu X, Shao J, Zhang X. Neuroprotection by Taurine on HBCD-Induced Apoptosis in PC12 Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 1:95-106. [PMID: 28849447 DOI: 10.1007/978-94-024-1079-2_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Hexabromocyclododecane (HBCD) is a widely used brominated flame retardant (BFR). Because of their presence in human issues, including brain tissue, concern has been raised on their possible neurotoxicity. Presently, we explored the neuroprotection of taurine against HBCD-induced apoptotic damages in PC12 cells. Cells were pre-treated with taurine before HBCD exposure and the viability was assayed via the methyl-thiazolyl-tetrazolium (MTT) method. Apoptotic features were observed with Hoechst 33342 staining. Apoptotic ratio was measured using flow cytometry with Annexin V-FITC coupled propidium iodide (PI) double staining. The changes in the levels of Bcl-2 and Bax proteins were quantitated by the western blot. The activity of caspase-3 was tested and the results revealed that presence of HBCD decreased cell survival and led to apoptosis in the tested cells. Further, exposure of HBCD reduced protein expression of Bcl-2, increased expression in Bax protein and activity of caspase-3. Taurine attenuated HBCD-induced cell viability loss and cell apoptosis. Moreover, taurine significantly prevented from reducing Bcl-2 protein expression and elevating Bax protein expression and caspase-3 activity induced by HBCD. These results demonstrated that taurine can alleviate HBCD-induced apoptosis by altering Bcl-2 expression and Bax protein and Caspase-3 activity in PC12.
Collapse
Affiliation(s)
- Yachen Li
- School of Public Health, Dalian Medical University, No. 9 Western Section of Lushun South Road, Dalian, 116044, China.
| | - Shuangyue Li
- School of Public Health, Dalian Medical University, No. 9 Western Section of Lushun South Road, Dalian, 116044, China
| | - Xizhe Xie
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hang Xiu
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaohui Liu
- School of Public Health, Dalian Medical University, No. 9 Western Section of Lushun South Road, Dalian, 116044, China
| | - Jing Shao
- School of Public Health, Dalian Medical University, No. 9 Western Section of Lushun South Road, Dalian, 116044, China
| | - Xiuli Zhang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
15
|
Li S, Yang L, Zhang Y, Zhang C, Shao J, Liu X, Li Y, Piao F. Taurine Ameliorates Arsenic-Induced Apoptosis in the Hippocampus of Mice Through Intrinsic Pathway. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 1:183-192. [PMID: 28849454 DOI: 10.1007/978-94-024-1079-2_16] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Our group previously reported that arsenic (As) exposure induced apoptosis in hippocampus neurons. The aim of the present study was to clarify the protective capacity of taurine (Tau) on As-induced neuronal apoptosis and the related mechanism in mouse hippocampus. Mice were divided into: control group, Tau control group, As exposure group and Tau protective group, randomly. The apoptotic rate of mouse hippocampus was determined by TUNEL staining. The levels of Bcl-2 and Bax gene and protein were analyzed by real time RT-PCR and WB, respectively. Furthermore, cytochrome c (Cyt C) release, and the activity of caspase-8 and caspase-3 were also determined. The results showed that Tau treatment induced the decrease of TUNEL-positive cells, prohibited the disturbance of Bcl-2 and Bax expression, and inhibited Cyt C release and caspase-8 and caspase-3 activation significantly. The results indicated that Tau supplement markedly ameliorates As-induced apoptosis by mitochondria-related pathway in mouse hippocampus.
Collapse
Affiliation(s)
- Shuangyue Li
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
| | - Lijun Yang
- Dalian Center for Disease Control and Prevention, Dalian, China
| | - Yan Zhang
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
- Xunyi Center for Disease Control and Prevention, Xunyi, China
| | - Cong Zhang
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
| | - Jing Shao
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
| | - Xiaohui Liu
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
| | - Yachen Li
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
| | - Fengyuan Piao
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China.
| |
Collapse
|
16
|
Taranukhin AG, Saransaari P, Kiianmaa K, Gunnar T, Oja SS. Comparison of Toxicity of Taurine and GABA in Combination with Alcohol in 7-Day-Old Mice. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 2:1021-1033. [PMID: 28849519 DOI: 10.1007/978-94-024-1079-2_81] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Previously, we described the combined toxicity of taurine and alcohol, and assumed hypoglycemia to be one reason of this toxicity. To understand whether taurine-ethanol combined toxicity is exclusively connected to taurine or whether other inhibitory amino acids may have similar effects when combined with ethanol, we tested different doses of gamma-aminobutyric acid (GABA) in combination with ethanol in 7-day-old mice. The minimal dose of GABA in combination with 5 g/kg ethanol which could kill a mouse was 2 g/kg. GABA combined with ethanol at doses of 3 g/kg, 4 g/kg, 6 g/kg induced lethality of 30%, 90% and 100%, correspondingly. Taurine at the doses of 4 and 6 g/kg combined with ethanol induced death in 60 and 100% of mice. Ethanol (5 g/kg), taurine (6 g/kg), GABA (4 g/kg) administered alone and the combination of ethanol (5 g/kg) with taurine (3 g/kg) have no lethal effects. GABA (6 g/kg) applied alone induced 90% lethality. Taurine or GABA alone decreased blood glucose in a dose-depending manner. Ethanol potentiated GABA- and taurine-induced decrease in blood glucose and in some animals it dropped from 8.8 (intact) to a hypoglycemic level 3.1-3.3 mmol/L (GABA 4 g/kg, taurine 6 g/kg), but this may not be considered a single reason of death. We conclude that the combination of GABA and ethanol has a lethal effect and this is stronger than the combined toxicity of ethanol and taurine.
Collapse
Affiliation(s)
| | | | - Kalervo Kiianmaa
- Department of Alcohol, Drugs and Addiction, National Institute for Health and Welfare, Helsinki, Finland
| | - Teemu Gunnar
- Alcohol and Drug Analytics Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Simo S Oja
- Medical School, University of Tampere, Tampere, Finland
| |
Collapse
|
17
|
Fontana BD, Stefanello FV, Mezzomo NJ, Müller TE, Quadros VA, Parker MO, Rico EP, Rosemberg DB. Taurine modulates acute ethanol-induced social behavioral deficits and fear responses in adult zebrafish. J Psychiatr Res 2018; 104:176-182. [PMID: 30096615 DOI: 10.1016/j.jpsychires.2018.08.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/25/2018] [Accepted: 08/02/2018] [Indexed: 01/08/2023]
Abstract
Ethanol (EtOH) is a central nervous system (CNS) depressant drug that modifies various behavioral domains (i.e., sociability, aggressiveness, and memory) by promoting disinhibition of punished operant behavior and neurochemical changes. Taurine (TAU) is a β-amino sulfonic acid with pleiotropic roles in the brain. Although exogenous TAU is found in energy drinks and often mixed with alcohol in beverages, the putative risks of mixing TAU and EtOH are poorly explored. Here, we investigated whether TAU modulates social and fear responses by assessing shoaling behavior, preference for conspecifics, and antipredatory behavior of adult zebrafish acutely exposed to EtOH. Zebrafish shoals (4 fish per shoal) were exposed to water (control), TAU (42, 150, and 400 mg/L), 0.25% (v/v) EtOH alone or in association with TAU for 1 h, and their behaviors were analyzed at different time intervals (0-5 min, 30-35 min, and 55-60 min). The effects of TAU and EtOH were further tested in a social preference test and during exposure to a predator. Both EtOH and TAU co-treated fish showed a higher shoal dispersion, while TAU 400/EtOH group shoal area had a similar profile when compared to control. However, in the social preference test, TAU 400/EtOH impaired the seeking for conspecifics. Regarding fear-like behaviors, TAU-cotreated fish showed a prominent reduction in risk assessments when compared to EtOH alone. Overall, we demonstrate that TAU modulates EtOH-induced changes in different behavioral domains, suggesting a complex relationship between social and fear-like responses.
Collapse
Affiliation(s)
- Barbara D Fontana
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil.
| | - Flavia V Stefanello
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil
| | - Nathana J Mezzomo
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; Graduate Program in Pharmacology, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil
| | - Talise E Müller
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil
| | - Vanessa A Quadros
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil
| | - Matthew O Parker
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, UK; The International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA, 70458, USA
| | - Eduardo P Rico
- Graduate Program in Health Sciences, Laboratory of Neural Signaling and Psychopharmacology, Academic Unit of Health Sciences, University of Southern Santa Catarina (UNESC), 1105, Universitária Avenue, Bloco S, Sala 6, Criciúma, SC, 88806-000, Brazil
| | - Denis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA, 70458, USA.
| |
Collapse
|
18
|
Ahmadi N, Ghanbarinejad V, Ommati MM, Jamshidzadeh A, Heidari R. Taurine prevents mitochondrial membrane permeabilization and swelling upon interaction with manganese: Implication in the treatment of cirrhosis-associated central nervous system complications. J Biochem Mol Toxicol 2018; 32:e22216. [PMID: 30152904 DOI: 10.1002/jbt.22216] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 07/16/2018] [Accepted: 07/20/2018] [Indexed: 12/18/2022]
Abstract
Brain tissue manganese (Mn) accumulation is a cirrhosis-associated complication. Cellular mitochondria are among the potential targets for Mn-induced cytotoxicity. Taurine is one of the most abundant amino acids with high concentrations in human brain tissue. Several pharmacological properties including regulation of mitochondrial function are attributed to taurine. The current investigation was designed to evaluate the effect of taurine on Mn-induced mitochondrial impairment in isolated mice brain mitochondria. The brain mitochondria were exposed to increasing concentrations of Mn (0.1-10 mM). Taurine (0.1, 1, and 10 mM) was added as the protective agent. The severe collapse of mitochondrial membrane potential, decreased mitochondrial dehydrogenases activity, mitochondrial swelling, and depleted mitochondrial adenosine triphosphate (ATP) were evident in Mn-exposed mitochondria. It was found that taurine administration preserved mitochondrial ATP, prevented mitochondrial depolarization and swelling, and increased mitochondrial dehydrogenases activity. These data suggest mitochondrial protection as an underlying mechanism for the protective effects of taurine against Mn toxicity.
Collapse
Affiliation(s)
- Nahid Ahmadi
- Pharmaceutical Sciences Research Center, Department of Toxicology, Shiraz University of Medical Sciences, Shiraz, Fars, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Vahid Ghanbarinejad
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Mohammad Mehdi Ommati
- Pharmaceutical Sciences Research Center, Department of Toxicology, Shiraz University of Medical Sciences, Shiraz, Fars, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Akram Jamshidzadeh
- Pharmaceutical Sciences Research Center, Department of Toxicology, Shiraz University of Medical Sciences, Shiraz, Fars, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Department of Toxicology, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| |
Collapse
|
19
|
Wang G, Liu Q, Guo L, Zeng H, Ding C, Zhang W, Xu D, Wang X, Qiu J, Dong Q, Fan Z, Zhang Q, Pan J. Gut Microbiota and Relevant Metabolites Analysis in Alcohol Dependent Mice. Front Microbiol 2018; 9:1874. [PMID: 30158912 PMCID: PMC6104187 DOI: 10.3389/fmicb.2018.01874] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/25/2018] [Indexed: 12/21/2022] Open
Abstract
Alcohol abuse is a major public health crisis. Relative evidences supported that the gut microbiota (GM) played an important role in central nervous system (CNS) function, and the composition of them had changed after alcohol drinking. We sought to explore the changes of GM in alcohol dependence. In our study, the GM of mice with alcohol administration was detected through analyzed 16S rRNA gene sequencing and the fecal metabolites were analyzed by LC-MS. The microbial diversity was significantly higher in the alcohol administration group, the abundance of phylum Firmicutes and its class Clostridiales were elevated, meanwhile the abundance of Lachnospiraceae, Alistipes, and Odoribacter showed significant differences among the three groups. Based on LC-MS results, bile acid, secondary bile acid, serotonin and taurine level had varying degrees of changes in alcohol model. From paraffin sections, tissue damage was observed in liver and colon. These findings provide direct evidence that alcohol intake affects the composition of GM, enable a better understanding of the function of GM in the microbiota-gut-brain (MGB) axis, and give a new thought for alcohol addiction treatment.
Collapse
Affiliation(s)
- Guanhao Wang
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Qing Liu
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Liang Guo
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Haijuan Zeng
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Chengchao Ding
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Wentong Zhang
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Dongpo Xu
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Xiang Wang
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jingxuan Qiu
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Qingli Dong
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Ziquan Fan
- Thermo Fisher Scientific, Shanghai, China
| | - Qi Zhang
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jing Pan
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
20
|
Wallauer MM, Huf F, Tortorelli LS, Rahmeier FL, Carvalho FB, Meurer RT, da Cruz Fernandes M. Morphological changes in the cerebellum as a result of ethanol treatment and cigarette smoke exposure: A study on astrogliosis, apoptosis and Purkinje cells. Neurosci Lett 2018; 672:70-77. [DOI: 10.1016/j.neulet.2018.02.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/17/2018] [Accepted: 02/21/2018] [Indexed: 01/13/2023]
|
21
|
Paulucio D, Terra A, Santos CG, Cagy M, Velasques B, Ribeiro P, da Costa BM, Gongora M, Alvarenga R, Alonso L, Pompeu FAMS. Acute effect of Ethanol and Taurine on frontal cortex absolute beta power before and after exercise. PLoS One 2018. [PMID: 29538445 PMCID: PMC5851630 DOI: 10.1371/journal.pone.0194264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ethanol (ET) is a substance that modulates the Central Nervous System (CNS). Frequently, ET intake occurs combined with energy drinks, which contain taurine (TA), an important amino acid found in the body (i.e brain and muscles). Although TA administration has been used in the improvement of physical performance, the impact of TA, ET and exercise remains unknown. This study aimed to analyze the acute effect of 6g of Taurine (TA), 0.6 mL∙kg-1 of Ethanol (ET), and Taurine combined with Ethanol (TA+ET) ingestion on the electrocortical activity before and after a moderate intensity exercise in 9 subjects, 5 women (counterbalanced experimental design). In each of the 4 treatments (Placebo—PL, TA, ET and TA+ET), electroencephalography (EEG) tests were conducted in order to analyze changes in absolute beta power (ABP) in the frontal lobe in 3 moments: baseline (before ingestion), peak (before exercise) and post-exercise. In the PL treatment, the frontal areas showed decrease in ABP after exercise. However, in the ET+TA treatment, ABP values were greater after exercise, except for Fp1. The ET treatment had no effect on the Superior Frontal Gyrus area (F3, Fz and F4) and ABP decreased after exercise in Fp1 and Fp2. In the TA treatment, ABP increased after exercise, while it decreased at the peak moment in most of the frontal regions, except for Fp1, F3 and Fz. We concluded that after a moderate intensity exercise, a decrease in cortical activity occurs in placebo treatment. Moreover, we found a inhibitory effect of TA on cortical activity before exercise and a increased in cortical activity after exercise. A small ET dose is not enough to alter ABP in all regions of the frontal cortex and, in combination with TA, it showed an increase in the frontal cortex activity at the post-exercise moment.
Collapse
Affiliation(s)
- Dailson Paulucio
- Biometrics Laboratory, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Postgraduate in Physical Education, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of physiology in soccer, Botafogo de Futebol e Regatas, Rio de Janeiro, Brazil
| | - Augusto Terra
- Biometrics Laboratory, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Postgraduate in Physical Education, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Caleb G. Santos
- Biometrics Laboratory, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Army Biology Institute, Brazilian Army, Rua Francisco Manuel, Triagem, Rio de Janeiro, RJ, Brazil
| | - Mauricio Cagy
- Biomedical Engineering Program, COPPE, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruna Velasques
- Postgraduate in Physical Education, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Brain Mapping and Sensory Motor Integration Laboratory, Institute of Psychiatry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Ribeiro
- Biometrics Laboratory, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Postgraduate in Physical Education, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Brain Mapping and Sensory Motor Integration Laboratory, Institute of Psychiatry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| | - Bruno M. da Costa
- Biometrics Laboratory, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Postgraduate in Physical Education, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Neuroscience Laboratory of Exercise, Institute of Psychiatry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana Gongora
- Brain Mapping and Sensory Motor Integration Laboratory, Institute of Psychiatry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renato Alvarenga
- Biometrics Laboratory, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Postgraduate in Physical Education, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciano Alonso
- Biometrics Laboratory, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Postgraduate in Physical Education, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando A. M. S. Pompeu
- Biometrics Laboratory, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Postgraduate in Physical Education, School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Abad N, Rosenberg JT, Roussel T, Grice DC, Harrington MG, Grant SC. Metabolic assessment of a migraine model using relaxation-enhanced 1 H spectroscopy at ultrahigh field. Magn Reson Med 2018; 79:1266-1275. [PMID: 28921630 PMCID: PMC5775911 DOI: 10.1002/mrm.26811] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 04/10/2017] [Accepted: 06/02/2017] [Indexed: 01/08/2023]
Abstract
PURPOSE This study evaluates biochemical imbalances in a rat model that reflects dysfunctional pathways in migraine. The high sensitivity and spectral dispersion available to 1 H MRS at 21.1 T expands metabolic profiling in this migraine model to include lactate (Lac), taurine (Tau), aspartate, and Gly-a mixture of glycine, glutamine, and glutamate. METHODS Sprague-Dawley male rats were administered in situ an intraperitoneal injection of nitroglycerin (NTG) to induce the migraine analogue or saline as a control. A selective relaxation-enhanced MR spectroscopy sequence was used to target upfield metabolites from a 4-mm3 voxel for 2.5 h after injection. RESULTS Significant increases were evident for Lac as early as 10 min after NTG injection, peaking over 50% compared with baseline and control (normalized Lac/N-acetyl aspartate with NTG = 1.54 ± 0.65 versus with saline = 0.99 ± 0.08). Tau decreased progressively in controls over 2 h after injection, but remained elevated with NTG, peaking at 105 min after injection (normalized Tau/N-acetyl aspartate with NTG = 1.10 ± 0.18 versus with saline = 0.85 ± 0.14). Total creatine under NTG showed significant decreases with time and compared with saline; Gly demonstrated temporal increases for NTG. CONCLUSIONS These changes indicate an altered metabolic profile in the migraine analogue consistent with early changes in neural activity and/or vasodilation consistent with progressively enhanced neuroprotection and osmoregulation. Magn Reson Med 79:1266-1275, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Nastaren Abad
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida, USA
| | - Jens T Rosenberg
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida, USA
| | - Tangi Roussel
- Department of Chemical Physics, Weizmann Institute Science, Rehovot, Israel
| | - Dillon C Grice
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA
| | - Michael G Harrington
- Molecular Neurology Program, Huntington Medical Research Institutes, Pasadena, California, USA
| | - Samuel C Grant
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
23
|
Nor Arfuzir NN, Agarwal R, Iezhitsa I, Agarwal P, Sidek S, Ismail NM. Taurine protects against retinal and optic nerve damage induced by endothelin-1 in rats via antioxidant effects. Neural Regen Res 2018; 13:2014-2021. [PMID: 30233077 PMCID: PMC6183037 DOI: 10.4103/1673-5374.239450] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Endothelin-1 (ET-1), a potent vasoconstrictor, is involved in retinal vascular dysregulation and oxidative stress in glaucomatous eyes. Taurine (TAU), a naturally occurring free amino acid, is known for its neuroprotective and antioxidant properties. Hence, we evaluated its neuroprotective properties against ET-1 induced retinal and optic nerve damage. ET-1 was administered intravitreally to Sprague-Dawley rats and TAU was injected as pre-, co- or post-treatment. Animals were euthanized seven days post TAU injection. Retinae and optic nerve were examined for morphology, and were also processed for caspase-3 immunostaining. Retinal redox status was estimated by measuring retinal superoxide dismutase, catalase, glutathione, and malondialdehyde levels using enzyme-linked immuosorbent assay. Histopathological examination showed significantly improved retinal and optic nerve morphology in TAU-treated groups. Morphometric examination showed that TAU pre-treatment provided marked protection against ET-1 induced damage to retina and optic nerve. In accordance with the morphological observations, immunostaining for caspase showed a significantly lesser number of apoptotic retinal cells in the TAU pre-treatment group. The retinal oxidative stress was reduced in all TAU-treated groups, and particularly in the pre-treatment group. The findings suggest that treatment with TAU, particularly pre-treatment, prevents apoptosis of retinal cells induced by ET-1 and hence prevents the changes in the morphology of retina and optic nerve. The protective effect of TAU against ET-1 induced retinal and optic nerve damage is associated with reduced retinal oxidative stress.
Collapse
Affiliation(s)
- Natasha Najwa Nor Arfuzir
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA Sungai Buloh Campus, Selangor, Malaysia
| | - Renu Agarwal
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA Sungai Buloh Campus, Selangor, Malaysia
| | - Igor Iezhitsa
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA Sungai Buloh Campus, Selangor, Malaysia; Volgograd State Medical University, Research Institute of Pharmacology, Volgograd, Russia
| | - Puneet Agarwal
- Faculty of Medicine, International Medical University, IMU Clinical School, Seremban, Malaysia
| | - Sabrilhakim Sidek
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA Sungai Buloh Campus, Selangor, Malaysia
| | - Nafeeza Mohd Ismail
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA Sungai Buloh Campus, Selangor, Malaysia
| |
Collapse
|
24
|
Taurine ameliorated thyroid function in rats co-administered with chlorpyrifos and lead. Vet Res Commun 2016; 40:123-129. [DOI: 10.1007/s11259-016-9662-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 08/16/2016] [Indexed: 11/25/2022]
|
25
|
Iron Oxide Nanoparticles Induce Dopaminergic Damage: In vitro Pathways and In Vivo Imaging Reveals Mechanism of Neuronal Damage. Mol Neurobiol 2016; 52:913-26. [PMID: 26099304 DOI: 10.1007/s12035-015-9259-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Various iron-oxide nanoparticles have been in use for a long time as therapeutic and imaging agents and for supplemental delivery in cases of iron-deficiency. While all of these products have a specified size range of ∼ 40 nm and above, efforts are underway to produce smaller particles, down to ∼ 1 nm. Here, we show that after a 24-h exposure of SHSY-5Y human neuroblastoma cells to 10 μg/ml of 10 and 30 nm ferric oxide nanoparticles (Fe-NPs), cellular dopamine content was depleted by 68 and 52 %, respectively. Increases in activated tyrosine kinase c-Abl, a molecular switch induced by oxidative stress, and neuronal α-synuclein expression, a protein marker associated with neuronal injury, were also observed (55 and 38 % percent increases, respectively). Inhibition of cell-proliferation, significant reductions in the number of active mitochondria, and a dose-dependent increase in reactive oxygen species (ROS) were observed in neuronal cells. Additionally, using a rat in vitro blood-brain barrier (BBB) model, a dose-dependent increase in ROS accompanied by increased fluorescein efflux demonstrated compromised BBB integrity. To assess translational implications, in vivo Fe-NP-induced neurotoxicity was determined using in vivo MRI and post-mortem neurochemical and neuropathological correlates in adult male rats after exposure to 50 mg/kg of 10 nm Fe-NPs. Significant decrease in T 2 values was observed. Dynamic observations suggested transfer and retention of Fe-NPs from brain vasculature into brain ventricles. A significant decrease in striatal dopamine and its metabolites was also observed, and neuropathological correlates provided additional evidence of significant nerve cell body and dopaminergic terminal damage as well as damage to neuronal vasculature after exposure to 10 nm Fe-NPs. These data demonstrate a neurotoxic potential of very small size iron nanoparticles and suggest that use of these ferric oxide nanoparticles may result in neurotoxicity, thereby limiting their clinical application.
Collapse
|
26
|
Choline Ameliorates Deficits in Balance Caused by Acute Neonatal Ethanol Exposure. THE CEREBELLUM 2016; 14:413-20. [PMID: 26085462 DOI: 10.1007/s12311-015-0691-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Fetal alcohol spectrum disorder (FASD) is estimated to occur in 1 % of all live births. The developing cerebellum is vulnerable to the toxic effects of alcohol. People with FASD have cerebellar hypoplasia and developmental deficits associated with cerebellar injury. Choline is an essential nutrient, but many diets in the USA are choline deficient. In rats, choline given with or following alcohol exposure reduces many alcohol-induced neurobehavioral deficits but not those associated with cerebellar function. Our objective was to determine if choline supplementation prior to alcohol exposure would ameliorate the impact of ethanol on a cerebellar-associated behavioral test in mice. Pregnant C57Bl6/J mice were maintained on a choline-deficient diet from embryonic day 4.5. On postnatal day 1 (P1), pups were assigned to one of eight treatment groups: choline (C) or saline (S) pre-treatment from P1 to P5, ethanol (6 g/kg) or Intralipid(®) on P5, C and or S post-treatment from P6 to P20. On P30, balance and coordination were tested using the dowel crossing test. Overall, there was a significant effect of treatment and females crossed longer distances than males. Ethanol exposure significantly reduced the total distance crossed. Choline pre-treatment increased the distance crossed by males, and both pre- and post-treatment with choline significantly increased total distance crossed for females and males. There was no effect of choline on Intralipid®-exposed animals. This is the first study to show that choline ameliorates ethanol-induced effects on balance and coordination when given before ethanol exposure. Choline fortification of common foodstuffs may reduce the effects of alcohol.
Collapse
|
27
|
Oliveira SA, Chuffa LGA, Fioruci-Fontanelli BA, Lizarte Neto FS, Novais PC, Tirapelli LF, Oishi JC, Takase LF, Stefanini MA, Martinez M, Martinez FE. Apoptosis of Purkinje and granular cells of the cerebellum following chronic ethanol intake. THE CEREBELLUM 2015; 13:728-38. [PMID: 25129034 DOI: 10.1007/s12311-014-0591-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ethanol alters motricity, learning, cognition, and cellular metabolism in the cerebellum. We evaluated the effect of ethanol on apoptosis in Golgi, Purkinje, and granule cells of the cerebellum in adult rats. There were two groups of 20 rats: a control group that did not consume ethanol and an experimental group of UChA rats that consumed ethanol at 10% (<2 g ethanol/kg body weight/day). At 120 days old, rats were anesthetized and decapitated, and their cerebella were collected and fixed. Cerebellar sections were subjected to immunohistochemistry for terminal deoxynucleotide transferase dUTP nick end labeling (TUNEL), caspase-3, X-linked inhibitor of apoptosis protein (XIAP), and insulin-like growth factor 1-receptor (IGF-1R); real-time PCR (RT-PCR) to determine caspase-3, XIAP, and IGF-1R gene expression; and transmission electron microscopy (TEM). We identified fragmentation of DNA and an increase in caspase-3 protein and XIAP in Purkinje cells, whereas granule cells exhibited increased caspase-3 and XIAP. IGF-1R expression was unchanged. There was no significant difference in gene expression of caspase-3, XIAP, and IGF-1R. There were an increase in lipid droplets, a reduction in the cellular cytoplasm in electron-dense nuclei, and changes in the myelin sheath in the cerebellar cortex. In conclusion, our data demonstrated that ethanol induced apoptosis in the Purkinje and granule cells of the cerebellum of adult UChA rats.
Collapse
Affiliation(s)
- Suelen A Oliveira
- Graduate Program in General and Applied Biology, Institute of Bioscience, Univ. Estadual Paulista (UNESP), Botucatu, SP, 18618-970, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Neonatal taurine and alanine modulate anxiety-like behavior and decelerate cortical spreading depression in rats previously suckled under different litter sizes. Amino Acids 2015; 47:2437-45. [DOI: 10.1007/s00726-015-2036-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 06/17/2015] [Indexed: 11/26/2022]
|
29
|
Marzban H, Del Bigio MR, Alizadeh J, Ghavami S, Zachariah RM, Rastegar M. Cellular commitment in the developing cerebellum. Front Cell Neurosci 2015; 8:450. [PMID: 25628535 PMCID: PMC4290586 DOI: 10.3389/fncel.2014.00450] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 12/12/2014] [Indexed: 12/11/2022] Open
Abstract
The mammalian cerebellum is located in the posterior cranial fossa and is critical for motor coordination and non-motor functions including cognitive and emotional processes. The anatomical structure of cerebellum is distinct with a three-layered cortex. During development, neurogenesis and fate decisions of cerebellar primordium cells are orchestrated through tightly controlled molecular events involving multiple genetic pathways. In this review, we will highlight the anatomical structure of human and mouse cerebellum, the cellular composition of developing cerebellum, and the underlying gene expression programs involved in cell fate commitments in the cerebellum. A critical evaluation of the cell death literature suggests that apoptosis occurs in ~5% of cerebellar cells, most shortly after mitosis. Apoptosis and cellular autophagy likely play significant roles in cerebellar development, we provide a comprehensive discussion of their role in cerebellar development and organization. We also address the possible function of unfolded protein response in regulation of cerebellar neurogenesis. We discuss recent advancements in understanding the epigenetic signature of cerebellar compartments and possible connections between DNA methylation, microRNAs and cerebellar neurodegeneration. Finally, we discuss genetic diseases associated with cerebellar dysfunction and their role in the aging cerebellum.
Collapse
Affiliation(s)
- Hassan Marzban
- Department of Human Anatomy and Cell Science, University of Manitoba Winnipeg, MB, Canada
| | - Marc R Del Bigio
- Department of Human Anatomy and Cell Science, University of Manitoba Winnipeg, MB, Canada ; Department of Pathology, University of Manitoba Winnipeg, MB, Canada
| | - Javad Alizadeh
- Department of Human Anatomy and Cell Science, University of Manitoba Winnipeg, MB, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba Winnipeg, MB, Canada
| | - Robby M Zachariah
- Department of Biochemistry and Medical Genetics, University of Manitoba Winnipeg, MB, Canada ; Regenerative Medicine Program, University of Manitoba Winnipeg, MB, Canada
| | - Mojgan Rastegar
- Department of Biochemistry and Medical Genetics, University of Manitoba Winnipeg, MB, Canada ; Regenerative Medicine Program, University of Manitoba Winnipeg, MB, Canada
| |
Collapse
|
30
|
Taranukhin AG, Saransaari P, Kiianmaa K, Oja SS. Hypoglycemia is one possible mechanism in the combined toxicity of ethanol and taurine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 803:305-12. [PMID: 25833506 DOI: 10.1007/978-3-319-15126-7_24] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
31
|
Activation of liver X receptor is protective against ethanol-induced developmental impairment of Bergmann glia and Purkinje neurons in the mouse cerebellum. Mol Neurobiol 2013; 49:176-86. [PMID: 23900741 DOI: 10.1007/s12035-013-8510-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 07/08/2013] [Indexed: 10/26/2022]
Abstract
Cerebellar Purkinje cell and granule cell development are coordinated by Bergmann glia, and are particularly sensitive to ethanol (EtOH) exposure. The liver X receptor (LXR) plays important roles in Bergmann glial development. However, the effect of LXR activation on EtOH-mediated impairment of Bergmann glia and subsequently on Purkinje cell dendritogenesis remains undetermined. Therefore, using immunohistochemistry, quantitative real-time PCR and Western blot, we tested the possible protection of LXR agonist T0901317 (T0) on Bergmann glia and Purkinje cell dendritogenesis in mice exposed to ethanol. Results showed that a brief exposure of EtOH on postnatal day (PD 5) significantly decreased the average body weight of mice at PD 6 without alteration in the brain weight. In EtOH-exposed mice, the number of migrating granule cells in the molecular layer was significantly decreased, and this effect was attenuated by pretreatment of T0. EtOH exposure also resulted in the significant reduction of calbindin-labeled Purkinje cells, their maximum dendrite length, and impairment of Purkinje cell dendritogenesis. Furthermore, EtOH induced the activation of microglia in the Purkinje cell layer and impaired the development of Bergmann glia. However, pretreatment of T0 effectively blocked all of these responses. These responses were found to be mediated by the inhibition of upregulated levels of β-catenin and transcription factor LEF1 in the cerebellum. Overall, the results suggest that activating LXRs on postnatal mice exposed to EtOH is protective to Bergmann glia, and thus may play a critical role in preventing EtOH-induced defects during cerebellar development.
Collapse
|
32
|
Sadrian B, Wilson DA, Saito M. Long-lasting neural circuit dysfunction following developmental ethanol exposure. Brain Sci 2013; 3:704-27. [PMID: 24027632 PMCID: PMC3767176 DOI: 10.3390/brainsci3020704] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/10/2013] [Accepted: 04/23/2013] [Indexed: 01/14/2023] Open
Abstract
Fetal Alcohol Spectrum Disorder (FASD) is a general diagnosis for those exhibiting long-lasting neurobehavioral and cognitive deficiencies as a result of fetal alcohol exposure. It is among the most common causes of mental deficits today. Those impacted are left to rely on advances in our understanding of the nature of early alcohol-induced disorders toward human therapies. Research findings over the last decade have developed a model where ethanol-induced neurodegeneration impacts early neural circuit development, thereby perpetuating subsequent integration and plasticity in vulnerable brain regions. Here we review our current knowledge of FASD neuropathology based on discoveries of long-lasting neurophysiological effects of acute developmental ethanol exposure in animal models. We discuss the important balance between synaptic excitation and inhibition in normal neural network function, and relate the significance of that balance to human FASD as well as related disease states. Finally, we postulate that excitation/inhibition imbalance caused by early ethanol-induced neurodegeneration results in perturbed local and regional network signaling and therefore neurobehavioral pathology.
Collapse
Affiliation(s)
- Benjamin Sadrian
- Department of Child and Adolescent Psychiatry, New York University Langone School of Medicine, One Park Avenue, Eighth Floor, New York, NY 10128, USA; E-Mail:
- Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA; E-Mail:
| | - Donald A. Wilson
- Department of Child and Adolescent Psychiatry, New York University Langone School of Medicine, One Park Avenue, Eighth Floor, New York, NY 10128, USA; E-Mail:
- Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA; E-Mail:
| | - Mariko Saito
- Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA; E-Mail:
- Department of Psychiatry, New York University Langone School of Medicine, One Park Avenue, Eighth Floor, New York, NY 10128, USA
| |
Collapse
|
33
|
You JS, Kim SY, Park SY, Chang KJ. Dietary Taurine and Nutrient Intake and Dietary Quality by Alcohol Consumption Level in Korean Male College Students. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 776:121-7. [DOI: 10.1007/978-1-4614-6093-0_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
|
34
|
Taurine and Its Neuroprotective Role. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 775:19-27. [DOI: 10.1007/978-1-4614-6130-2_2] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
35
|
Lethality of Taurine and Alcohol Coadministration in Mice. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 776:29-38. [DOI: 10.1007/978-1-4614-6093-0_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
36
|
Ripps H, Shen W. Review: taurine: a "very essential" amino acid. Mol Vis 2012; 18:2673-86. [PMID: 23170060 PMCID: PMC3501277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 11/08/2012] [Indexed: 11/22/2022] Open
Abstract
Taurine is an organic osmolyte involved in cell volume regulation, and provides a substrate for the formation of bile salts. It plays a role in the modulation of intracellular free calcium concentration, and although it is one of the few amino acids not incorporated into proteins, taurine is one of the most abundant amino acids in the brain, retina, muscle tissue, and organs throughout the body. Taurine serves a wide variety of functions in the central nervous system, from development to cytoprotection, and taurine deficiency is associated with cardiomyopathy, renal dysfunction, developmental abnormalities, and severe damage to retinal neurons. All ocular tissues contain taurine, and quantitative analysis of ocular tissue extracts of the rat eye revealed that taurine was the most abundant amino acid in the retina, vitreous, lens, cornea, iris, and ciliary body. In the retina, taurine is critical for photoreceptor development and acts as a cytoprotectant against stress-related neuronal damage and other pathological conditions. Despite its many functional properties, however, the cellular and biochemical mechanisms mediating the actions of taurine are not fully known. Nevertheless, considering its broad distribution, its many cytoprotective attributes, and its functional significance in cell development, nutrition, and survival, taurine is undoubtedly one of the most essential substances in the body. Interestingly, taurine satisfies many of the criteria considered essential for inclusion in the inventory of neurotransmitters, but evidence of a taurine-specific receptor has yet to be identified in the vertebrate nervous system. In this report, we present a broad overview of the functional properties of taurine, some of the consequences of taurine deficiency, and the results of studies in animal models suggesting that taurine may play a therapeutic role in the management of epilepsy and diabetes.
Collapse
Affiliation(s)
- Harris Ripps
- Departments of Ophthalmology and Visual Science, Anatomy and Cell Biology, Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL 60612, USA.
| | | |
Collapse
|
37
|
Sayed RH, Salem HA, El-Sayeh BM. Potential protective effect of taurine against dibromoacetonitrile-induced neurotoxicity in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2012; 34:849-857. [PMID: 23021633 DOI: 10.1016/j.etap.2012.08.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Revised: 08/26/2012] [Accepted: 08/29/2012] [Indexed: 06/01/2023]
Abstract
Dibromoacetonitrile (DBAN) is a disinfection by-product of water chlorination. Epidemiological studies indicate that it might present a potential hazard to human health. The present study aimed to investigate the possible neurotoxicity of DBAN in rats and possible protection by taurine. Based on initial dose-response experiment, DBAN (60 mg/kg) was administrated orally for 7 days. DBAN administration significantly impaired behavior of rats. Further, DBAN produced significant decrease of monoamines, γ-aminobutyric acid (GABA), glutamate contents, acetylcholinestrase (AChE) and aspartate aminotransferase (AST) activities, in rat brain. On the other hand, a significant increase in malondialdehyde (MDA), nitric oxide (NO) contents and lactic dehydrogenase (LDH) activity was observed. Co-administration of taurine (200mg/kg, i.p.) with DBAN mitigated most tested parameters. In conclusion, the present study indicates that DBAN has the propensity to cause significant oxidative damage in rat brain. However, taurine has a promising role in attenuating the obtained hazardous effects of DBAN.
Collapse
Affiliation(s)
- Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | | | | |
Collapse
|
38
|
Taurine protects cerebellar neurons of the external granular layer against ethanol-induced apoptosis in 7-day-old mice. Amino Acids 2012; 43:1705-11. [DOI: 10.1007/s00726-012-1254-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 02/14/2012] [Indexed: 10/28/2022]
|
39
|
Buddhala C, Prentice H, Wu JY. Modes of Action of Taurine and Granulocyte Colony-stimulating Factor in Neuroprotection. ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.jecm.2011.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
40
|
In vivo monitoring of recovery from neurodegeneration in conditional transgenic SCA1 mice. Exp Neurol 2011; 232:290-8. [PMID: 21963649 DOI: 10.1016/j.expneurol.2011.09.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 07/29/2011] [Accepted: 09/12/2011] [Indexed: 11/24/2022]
Abstract
Reliable and objective markers of neuronal function and pathology that can directly assess the effects of neuroprotective treatments in the brain are urgently needed for clinical trials in neurodegenerative diseases. Here we assessed the sensitivity of high field proton magnetic resonance spectroscopy ((1)H MRS) to monitor reversal of neurodegeneration by taking advantage of a well characterized conditional mouse model of spinocerebellar ataxia type 1 (SCA1), where the cerebellar pathology and ataxic phenotype are reversible by doxycycline administration. Transgene expression was suppressed by feeding the mice with chow that contains doxycycline from 6 to 12 weeks of age in an early stage group and from 12 to 24 weeks in a mid-stage group. Cerebellar neurochemical profiles of treated and untreated conditional mice were measured at 9.4 tesla (T) before and after treatment and compared to those of wild type (WT) controls, as well as to histology measures (molecular layer thickness in the primary fissure and a global pathological severity score). Concentrations of N-acetylaspartate (NAA) and myo-inositol in the treated mice trended toward normalization to WT levels in both the early and mid-stage groups. The NAA-to-myo-inositol ratio was significantly different between the treated vs. untreated SCA1 mice and demonstrated partial reversal to WT values both at early and mid-stage, consistent with the histological measures. Taurine and total creatine levels were completely normalized in early and mid-stage treatment groups, respectively. The MRS markers were a more sensitive measure of treatment response than the histological measures from the same volume-of-interest in the early stage group. NAA, myo-inositol and taurine levels were significantly correlated with the histology measures in data combined from all groups. These data demonstrate that MRS markers reliably detect rescue from neuronal pathology and imply that the neurochemical levels measured by MRS accurately reflect treatment efficacy. Therefore this study presents an important step in validating MRS biomarkers as potential surrogate markers to evaluate therapeutics in pre-clinical and clinical trials in SCA1.
Collapse
|
41
|
Ramezani A, Goudarzi I, Lashkarbolouki T, Ghorbanian MT, Salmani ME, Abrari K. Neuroprotective effects of the 17β-estradiol against ethanol-induced neurotoxicity and oxidative stress in the developing male rat cerebellum: biochemical, histological and behavioral changes. Pharmacol Biochem Behav 2011; 100:144-51. [PMID: 21851833 DOI: 10.1016/j.pbb.2011.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Revised: 07/06/2011] [Accepted: 07/15/2011] [Indexed: 10/17/2022]
Abstract
During particular periods of central nervous system (CNS) development, exposure to ethanol can decrease regional brain growth and can result in selective loss of neurons. Unfortunately, there are few effective means of attenuating damage in the immature brain. In this study, the possible antioxidant and neuroprotective properties of 17β-estradiol against ethanol-induced neurotoxicity was investigated. 17β-estradiol (600 μg/kg) was injected subcutaneously in postnatal day (PD) 4 and 5, 30 min prior to intraperitoneal injection of ethanol (6g/kg) in rat pups. Ninety minutes after injection of ethanol, the activities of several antioxidant enzymes including superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (Gpx) in vermis of cerebellum were assayed. Thiobarbituric acid reactive substance (TBARS) levels were also measured as a marker of lipid peroxidation. Behavioral studies, including rotarod and locomotor activity tests were performed in PD 21-23 and histological study was performed after completion of behavioral measurements in postnatal day 23. The results of the present work demonstrated that ethanol could induce lipid peroxidation, increase TBARS levels and decrease glutathione peroxidase levels in pup cerebellum. We also observed that ethanol impaired performance on the rotarod and locomotor activities of rat pups. However, treatment with 17β-estradiol significantly attenuated motoric impairment, the lipid peroxidation process and restored the levels of antioxidants. Histological analysis also indicated that ethanol could decrease vermis Purkinje cell count and 17β-estradiol prevented this toxic effect. These results suggest that ethanol may induce lipid peroxidation in the rat pups cerebellum while treatment with 17β-estradiol improves motor deficits by protecting the cerebellum against ethanol toxicity.
Collapse
Affiliation(s)
- Azam Ramezani
- Faculty of Biology, Damghan University, Damghan, Iran
| | | | | | | | | | | |
Collapse
|
42
|
Zhou J, Li Y, Yan G, Bu Q, Lv L, Yang Y, Zhao J, Shao X, Deng Y, Zhu R, Zhao Y, Cen X. Protective Role of Taurine Against Morphine-Induced Neurotoxicity in C6 Cells via Inhibition of Oxidative Stress. Neurotox Res 2011; 20:334-42. [DOI: 10.1007/s12640-011-9247-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/05/2011] [Accepted: 05/06/2011] [Indexed: 01/15/2023]
|