1
|
Liu RX, Song DK, Zhang YY, Gong HX, Jin YC, Wang XS, Jiang YL, Yan YX, Lu BN, Wu YM, Wang M, Li XB, Zhang K, Liu SB. L-Cysteine: A promising nutritional supplement for alleviating anxiety disorders. Neuroscience 2024; 555:213-221. [PMID: 39089569 DOI: 10.1016/j.neuroscience.2024.07.038] [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: 04/30/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024]
Abstract
Anxiety disorders are prevalent chronic psychological disease with complex pathogenic mechanisms. Current anxiolytics have limited efficacy and numerous side effects in many anxiety patients, highlighting the urgent need for new therapies. Recent research has been focusing on nutritional supplements, particularly amino acids, as potential therapies for anxiety disorders. Among these, L-Cysteine plays a crucial role in various biological processes. L-Cysteine exhibits antioxidant properties that can enhance the antioxidant functions of the central nervous system (CNS). Furthermore, metabolites of L-cysteine, such as glutathione and hydrogen sulfide have been shown to alleviate anxiety through distinct molecular mechanisms. Long-term administration of L-Cysteine has anxiolytic, antidepressant, and memory-improving effects. L-Cysteine depletion can lead to increased oxidative stress in the brain. This review delves into the potential mechanisms of L-Cysteine and its main products, glutathione (GSH) and hydrogen sulfide (H2S) in the management of anxiety and related diseases.
Collapse
Affiliation(s)
- Rui-Xia Liu
- College of Life Sciences, Northwest University, Xi'an 710069, China; Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Da-Ke Song
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Ying-Ying Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Heng-Xin Gong
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Yu-Chen Jin
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xin-Shang Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China; State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Yong-Li Jiang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xinsi Road 1, Shaanxi, Xi'an 710038, China
| | - Yu-Xuan Yan
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Bei-Ning Lu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Yu-Mei Wu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Min Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xu-Bo Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Kun Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| | - Shui-Bing Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China; State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
2
|
Song X, Wang Y, Zou W, Wang Z, Cao W, Liang M, Li F, Zeng Q, Ren Z, Wang Y, Zheng K. Inhibition of mitophagy via the EIF2S1-ATF4-PRKN pathway contributes to viral encephalitis. J Adv Res 2024:S2090-1232(24)00326-6. [PMID: 39103048 DOI: 10.1016/j.jare.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024] Open
Abstract
INTRODUCTION Mitophagy, a selective form of autophagy responsible for maintaining mitochondrial homeostasis, regulates the antiviral immune response and acts as viral replication platforms to facilitate infection with various viruses. However, its precise role in herpes simplex virus 1 (HSV-1) infection and herpes simplex encephalitis (HSE) remains largely unknown. OBJECTIVES We aimed to investigate the regulation of mitophagy by HSV-1 neurotropic infection and its role in viral encephalitis, and to identify small compounds that regulate mitophagy to affect HSV-1 infection. METHODS The antiviral effects of compounds were investigated by Western blot, RT-PCR and plaque assay. The changes of Parkin (PRKN)-mediated mitophagy and Nuclear Factor kappa B (NFKB)-mediated neuroinflammation were examined by TEM, RT-qPCR, Western blot and ELISA. The therapeutic effect of taurine or PRKN-overexpression was confirmed in the HSE mouse model by evaluating survival rate, eye damage, neurodegenerative symptoms, immunohistochemistry analysis and histopathology. RESULTS HSV-1 infection caused the accumulation of damaged mitochondria in neuronal cells and in the brain tissue of HSE mice. Early HSV-1 infection led to mitophagy activation, followed by inhibition in the later viral infection. The HSV-1 proteins ICP34.5 or US11 deregulated the EIF2S1-ATF4 axis to suppress PRKN/Parkin mRNA expression, thereby impeding PRKN-dependent mitophagy. Consequently, inhibition of mitophagy by specific inhibitor midiv-1 promoted HSV-1 infection, whereas mitophagy activation by PRKN overexpression or agonists (CCCP and rotenone) attenuated HSV-1 infection and reduced the NF-κB-mediated neuroinflammation. Moreover, PRKN-overexpressing mice showed enhanced resistance to HSV-1 infection and ameliorated HSE pathogenesis. Furthermore, taurine, a differentially regulated gut microbial metabolite upon HSV-1 infection, acted as a mitophagy activator that transcriptionally promotes PRKN expression to stimulate mitophagy and to limit HSV-1 infection both in vitro and in vivo. CONCLUSION These results reveal the protective function of mitophagy in HSE pathogenesis and highlight mitophagy activation as a potential antiviral therapeutic strategy for HSV-1-related diseases.
Collapse
Affiliation(s)
- Xiaowei Song
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou 510632, China; Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou 511400, China
| | - Yiliang Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510440, China
| | - Weixiangmin Zou
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou 510632, China
| | - Zexu Wang
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou 510632, China
| | - Wenyan Cao
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou 510632, China
| | - Minting Liang
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou 510632, China
| | - Feng Li
- Infectious Diseases Institute, Guangzhou Eighth People's Hospital, Guangzhou 510440, China
| | - Qiongzhen Zeng
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou 510632, China
| | - Zhe Ren
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou 510632, China
| | - Yifei Wang
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou 510632, China.
| | - Kai Zheng
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China.
| |
Collapse
|
3
|
Marques LDS, Rocha YMD, Nascimento GAD, Santos SAAR, Vieira NCG, Moura LFWG, Alves DR, Silva WMBD, de Morais SM, de Oliveira KA, da Silva LMR, Sousa KKOD, Vieira-Neto AE, Coutinho HDM, Campos AR, Magalhães FEA. Potential of the Blue Calm® food supplement in the treatment of alcohol withdrawal-induced anxiety in adult zebrafish (Danio rerio). Neurochem Int 2024; 175:105706. [PMID: 38423391 DOI: 10.1016/j.neuint.2024.105706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/02/2024]
Abstract
Alcohol use disorder (AUD) is characterized by a set of behavioral, cognitive, nutritional, and physiological phenomena derived from the uncontrolled use of alcoholic beverages. There are cases in which AUD is associated with anxiety disorder, and when untreated, it requires careful pharmacotherapy. Blue Calm® (BC) is a food supplement indicated to aid restorative sleep, which has traces of medicinal plant extracts, as well as myo-inositol, magnesium bisglycinate, taurine, and L-tryptophan as its main chemical constituents. In this context, this study aimed to evaluate the potential of the BC in the treatment alcohol withdrawal-induced anxiety in adult zebrafish (aZF). Initially, BC was submitted to antioxidant activity against 2,2-diphenyl-1-picrylhydrazyl radical. Subsequently, the aZF (n = 6/group) were treated with BC (0.1 or 1 or 10 mg/mL; 20 μL; p.o.), and the sedative effect and acute toxicity (96 h) were evaluated. Then, the anxiolytic-like effect and the possible GABAergic mechanism were analyzed through the Light & Dark Test. Finally, BC action was evaluated for treating alcohol withdrawal-induced anxiety in aZF. Molecular docking was performed to evaluate the interaction of the major chemical constituents of BC with the GABAA receptor. BC showed antioxidant potential, a sedative effect, was not toxic, and all doses of BC had an anxiolytic-like effect and showed potential for the treatment of alcohol withdrawal-induced anxiety in aZF. In addition to the anxiolytic action, the main chemical constituents of BC were confirmed in the molecular docking, thus suggesting that BC is an anxiolytic that modulates the GABAergic system and has pharmacological potential for the treatment of alcohol withdrawal-induced anxiety.
Collapse
Affiliation(s)
- Luzia Débora S Marques
- Universidade Estadual do Ceará, Programa de Pós-Graduação em Nutrição e Saúde (PPGNS), Centro de Ciências da Saúde (CCS), Campus Do Itaperi, CEP 60.741-000, Fortaleza, CE, Brazil
| | - Yatagan M da Rocha
- Universidade Estadual do Ceará, Programa de Pós-Graduação em Nutrição e Saúde (PPGNS), Centro de Ciências da Saúde (CCS), Campus Do Itaperi, CEP 60.741-000, Fortaleza, CE, Brazil
| | - Gabriela A do Nascimento
- Universidade Estadual do Ceará, Programa de Pós-Graduação em Nutrição e Saúde (PPGNS), Centro de Ciências da Saúde (CCS), Campus Do Itaperi, CEP 60.741-000, Fortaleza, CE, Brazil
| | - Sacha Aubrey A R Santos
- Universidade de Fortaleza, Rede Nordeste de Biotecnologia (RENORBIO), Programa de Pós-Graduação em Ciências Médicas (PPGCM), Núcleo de Biologia Experimental (NUBEX), CEP 60.811-650, Fortaleza, Ceará, Brazil
| | - Natália Chaves G Vieira
- Universidade de Fortaleza, Rede Nordeste de Biotecnologia (RENORBIO), Programa de Pós-Graduação em Ciências Médicas (PPGCM), Núcleo de Biologia Experimental (NUBEX), CEP 60.811-650, Fortaleza, Ceará, Brazil
| | - Luiz Francisco Wemmenson G Moura
- Universidade de Fortaleza, Rede Nordeste de Biotecnologia (RENORBIO), Programa de Pós-Graduação em Ciências Médicas (PPGCM), Núcleo de Biologia Experimental (NUBEX), CEP 60.811-650, Fortaleza, Ceará, Brazil
| | - Daniela R Alves
- Universidade Estadual do Ceará, Programa de Pós-Graduação em Ciências Naturais (PPGCS), Centro de Ciências e Tecnologia (CCT), Laboratório de Análises Cromatográficas e Espectroscópicas (LACES), Campus do Itaperi, CEP 60714-903, Fortaleza, Ceará, Brazil
| | - Wildson Max B da Silva
- Universidade Estadual do Ceará, Programa de Pós-Graduação em Ciências Naturais (PPGCS), Centro de Ciências e Tecnologia (CCT), Laboratório de Análises Cromatográficas e Espectroscópicas (LACES), Campus do Itaperi, CEP 60714-903, Fortaleza, Ceará, Brazil
| | - Selene Maia de Morais
- Universidade Estadual do Ceará, Programa de Pós-Graduação em Ciências Naturais (PPGCS), Centro de Ciências e Tecnologia (CCT), Laboratório de Análises Cromatográficas e Espectroscópicas (LACES), Campus do Itaperi, CEP 60714-903, Fortaleza, Ceará, Brazil.
| | - Keciany A de Oliveira
- Universidade Estadual do Ceará, Programa de Pós-Graduação em Nutrição e Saúde (PPGNS), Centro de Ciências da Saúde (CCS), Campus Do Itaperi, CEP 60.741-000, Fortaleza, CE, Brazil
| | - Larissa Morais R da Silva
- Universidade Federal do Ceará, Programa de Pós-Graduação em Ciências e Tecnologia de Alimentos (PPGCTA), Laboratório de Microbiologia de Alimentos (LMA), Campos do Pici, CEP 60.356.000, Fortaleza, Ceará, Brazil.
| | - Kalina Kelma O de Sousa
- Universidade Estadual do Ceará, Laboratório de Bioprospecção de Produtos Naturais e Biotecnologia (LBPNB), Campus CECITEC, CEP 60.660-000, Tauá, Ceará, Brazil
| | - Antonio Eufrásio Vieira-Neto
- Universidade de Fortaleza, Rede Nordeste de Biotecnologia (RENORBIO), Programa de Pós-Graduação em Ciências Médicas (PPGCM), Núcleo de Biologia Experimental (NUBEX), CEP 60.811-650, Fortaleza, Ceará, Brazil
| | - Henrique Douglas Melo Coutinho
- Universidade Regional do Cariri - URCA, Programa de Pós-Graduação em Química Biológica (PPGQB), Laboratório de Microbiologia e Biologia Molecular (LMBM), CEP 63105-000, Crato, Ceará, Brazil.
| | - Adriana Rolim Campos
- Universidade de Fortaleza, Rede Nordeste de Biotecnologia (RENORBIO), Programa de Pós-Graduação em Ciências Médicas (PPGCM), Núcleo de Biologia Experimental (NUBEX), CEP 60.811-650, Fortaleza, Ceará, Brazil.
| | - Francisco Ernani Alves Magalhães
- Universidade Estadual do Ceará, Programa de Pós-Graduação em Nutrição e Saúde (PPGNS), Centro de Ciências da Saúde (CCS), Campus Do Itaperi, CEP 60.741-000, Fortaleza, CE, Brazil; Universidade de Fortaleza, Rede Nordeste de Biotecnologia (RENORBIO), Programa de Pós-Graduação em Ciências Médicas (PPGCM), Núcleo de Biologia Experimental (NUBEX), CEP 60.811-650, Fortaleza, Ceará, Brazil; Universidade Estadual do Ceará, Laboratório de Bioprospecção de Produtos Naturais e Biotecnologia (LBPNB), Campus CECITEC, CEP 60.660-000, Tauá, Ceará, Brazil.
| |
Collapse
|
4
|
Jangra A, Gola P, Singh J, Gond P, Ghosh S, Rachamalla M, Dey A, Iqbal D, Kamal M, Sachdeva P, Jha SK, Ojha S, Kumar D, Jha NK, Chopra H, Tan SC. Emergence of taurine as a therapeutic agent for neurological disorders. Neural Regen Res 2024; 19:62-68. [PMID: 37488845 PMCID: PMC10479846 DOI: 10.4103/1673-5374.374139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/01/2023] [Accepted: 03/28/2023] [Indexed: 07/26/2023] Open
Abstract
Taurine is a sulfur-containing, semi-essential amino acid that occurs naturally in the body. It alternates between inflammation and oxidative stress-mediated injury in various disease models. As part of its limiting functions, taurine also modulates endoplasmic reticulum stress, Ca2+ homeostasis, and neuronal activity at the molecular level. Taurine effectively protects against a number of neurological disorders, including stroke, epilepsy, cerebral ischemia, memory dysfunction, and spinal cord injury. Although various therapies are available, effective management of these disorders remains a global challenge. Approximately 30 million people are affected worldwide. The design of taurine formation could lead to potential drugs/supplements for the health maintenance and treatment of central nervous system disorders. The general neuroprotective effects of taurine and the various possible underlying mechanisms are discussed in this review. This article is a good resource for understanding the general effects of taurine on various diseases. Given the strong evidence for the neuropharmacological efficacy of taurine in various experimental paradigms, it is concluded that this molecule should be considered and further investigated as a potential candidate for neurotherapeutics, with emphasis on mechanism and clinical studies to determine efficacy.
Collapse
Affiliation(s)
- Ashok Jangra
- Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, Haryana, India
| | - Priyanka Gola
- Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, Haryana, India
| | - Jiten Singh
- Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, Haryana, India
| | - Pooja Gond
- Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, Haryana, India
| | - Swarnabha Ghosh
- Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, Haryana, India
| | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah, Saudi Arabia
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Dinesh Kumar
- Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, Haryana, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, India
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, India
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
5
|
Pyrzanowska J. Pharmacological activity of Aspalathus linearis extracts: pre-clinical research in view of prospective neuroprotection. Nutr Neurosci 2023; 26:384-402. [PMID: 35311618 DOI: 10.1080/1028415x.2022.2051955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Rooibos tea, a very popular everyday beverage made of Aspalathus linearis plant material and containing multiple polyphenolic compounds, reveals an expectation to positively affect various processes observed in the pathogenesis of neurodegenerative diseases as in the case of consumption of other polyphenol-abundant food products. METHODS This review is based on available data from pre-clinical in vitro and in vivo studies and presents a broad report on the pharmacological activity of the A. linearis extracts relevant for neurodegenerative diseases. RESULTS Flavonoids present in herbal infusions are absorbed from gastro-intestinal tract and may affect the central nervous system. The experimental investigations yield the results indicating to supporting role of A. linearis in the prevention of neurodegeneration, primarily owing to anti-oxidative and anti-inflammatory properties, anti-hyperglycaemic and anti-hyperlipidaemic effects as well as favourable impact on neurotransmission with following cognitive and behavioural after-math. DISCUSSION The multiple pharmacological activities and safety of Aspalathus linearis extracts are commented in the manuscript. The continuous rooibos tea consumption seems to be safe (despite anecdotal liver irritation); however, there is a risk of herbal-drug interactions.
Collapse
Affiliation(s)
- Justyna Pyrzanowska
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Warsaw, Poland
| |
Collapse
|
6
|
Ruminal Degradation of Taurine and Its Effects on Rumen Fermentation In Vitro. FERMENTATION-BASEL 2023. [DOI: 10.3390/fermentation9010043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Taurine accounts for approximately 0.1% of an animal’s body. It cannot be used for protein synthesis but plays a wide range of important roles in the animal body. Taurine does not exist in plants, while mammals can only synthesize 30–40% of the taurine they need. Supplementing taurine to beef cattle may be necessary to improve their nutrient utilization and health status. However, no data are available regarding the metabolism of taurine in the rumen. Two in vitro trials were conducted to investigate the ruminal degradability of taurine and its effects on rumen fermentation. In Trial 1, Tilley and Terry’s in vitro rumen fermentation technique was used for incubation. As treatments, two levels of taurine, i.e., 0 and 10 mg, were added into plastic tubes containing 0.4000 g of feed mixture with a calibrated volume of 50 mL. Three adult cattle fitted with rumen cannulas were used as the donors for rumen fluid. The incubation was carried out at 39 °C for 48 h. The results showed that the taurine degradability increased with incubation time (p < 0.001) while its 2 h-degradability reached 99%. Taurine decreased the 48 h-dry matter degradability (DMD) (p = 0.008) and increased the 24 h- and 48 h-pH (p = 0.005; p = 0.018), respectively. In Trial 2, the Hohenheim gas test was used for incubation. Four levels of taurine, i.e., 0, 5, 10 and 20 mg, were added into glass syringes containing 0.2000 g feed mixture with a calibrated volume of 100 mL as treatments. The rumen fluid donors were the same as in Trial 1. The incubation was carried out at 39 °C for 48 h. The results showed that taurine increased the 48 h-pH (p < 0.001) linearly, decreased the cumulative gas production (p < 0.001) and the total volatile fatty acids (VFA) concentration (p = 0.014), and quadratically affected the ammonia–nitrogen (p < 0.001) and microbial crude protein (MCP) concentrations (p < 0.001). It was concluded that taurine was highly degradable in rumen fermentation. Taurine inhibits ruminal fermentation by decreasing DMD, VFA and gas production while improving MCP synthesis on a dose-dependent basis.
Collapse
|
7
|
Giongo FK, Gallas-Lopes M, Benvenutti R, Sachett A, Bastos LM, Rosa AR, Herrmann AP. Effects of Taurine in Mice and Zebrafish Behavioral Assays With Translational Relevance to Schizophrenia. Int J Neuropsychopharmacol 2022; 26:125-136. [PMID: 36239455 PMCID: PMC9926054 DOI: 10.1093/ijnp/pyac073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/15/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Altered redox state and developmental abnormalities in glutamatergic and GABAergic transmission during development are linked to the behavioral changes associated with schizophrenia. As an amino acid that exerts antioxidant and inhibitory actions in the brain, taurine is a potential candidate to modulate biological targets relevant to this disorder. Here, we investigated in mice and zebrafish assays whether taurine prevents the behavioral changes induced by acute administration of MK-801 (dizocilpine), a glutamate N-methyl-D-aspartate (NMDA) receptor antagonist. METHODS C57BL/6 mice were i.p. administered with saline or taurine (50, 100, and 200 mg/kg) followed by MK-801 (0.15 mg/kg). Locomotor activity, social interaction, and prepulse inhibition of the acoustic startle reflex were then assessed in different sets of animals. Zebrafish were exposed to tank water or taurine (42, 150, and 400 mg/L) followed by MK-801 (5 µM); social preference and locomotor activity were evaluated in the same test. RESULTS MK-801 induced hyperlocomotion and disrupted sensorimotor gating in mice; in zebrafish, it reduced sociability and increased locomotion. Taurine was mostly devoid of effects and did not counteract NMDA antagonism in mice or zebrafish. DISCUSSION Contradicting previous clinical and preclinical data, taurine did not show antipsychotic-like effects in the present study. However, it still warrants consideration as a preventive intervention in animal models relevant to the prodromal phase of schizophrenia; further studies are thus necessary to evaluate whether and how taurine might benefit patients.
Collapse
Affiliation(s)
- Franciele Kich Giongo
- Laboratório de Neurobiologia e Psicofarmacologia Experimental (PsychoLab), Departamento de Farmacologia,Programa de Pós-Graduação em Farmacologia e Terapêutica
| | - Matheus Gallas-Lopes
- Laboratório de Neurobiologia e Psicofarmacologia Experimental (PsychoLab), Departamento de Farmacologia
| | | | | | - Leonardo Marensi Bastos
- Laboratório de Neurobiologia e Psicofarmacologia Experimental (PsychoLab), Departamento de Farmacologia
| | - Adriane Ribeiro Rosa
- Programa de Pós-Graduação em Farmacologia e Terapêutica,Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana Paula Herrmann
- Correspondence: Ana Paula Herrmann, PhD, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul. Rua Ramiro Barcelos 2600, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil ()
| |
Collapse
|
8
|
Ramírez-Guerrero S, Guardo-Maya S, Medina-Rincón GJ, Orrego-González EE, Cabezas-Pérez R, González-Reyes RE. Taurine and Astrocytes: A Homeostatic and Neuroprotective Relationship. Front Mol Neurosci 2022; 15:937789. [PMID: 35866158 PMCID: PMC9294388 DOI: 10.3389/fnmol.2022.937789] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/17/2022] [Indexed: 12/20/2022] Open
Abstract
Taurine is considered the most abundant free amino acid in the brain. Even though there are endogenous mechanisms for taurine production in neural cells, an exogenous supply of taurine is required to meet physiological needs. Taurine is required for optimal postnatal brain development; however, its brain concentration decreases with age. Synthesis of taurine in the central nervous system (CNS) occurs predominantly in astrocytes. A metabolic coupling between astrocytes and neurons has been reported, in which astrocytes provide neurons with hypotaurine as a substrate for taurine production. Taurine has antioxidative, osmoregulatory, and anti-inflammatory functions, among other cytoprotective properties. Astrocytes release taurine as a gliotransmitter, promoting both extracellular and intracellular effects in neurons. The extracellular effects include binding to neuronal GABAA and glycine receptors, with subsequent cellular hyperpolarization, and attenuation of N-methyl-D-aspartic acid (NMDA)-mediated glutamate excitotoxicity. Taurine intracellular effects are directed toward calcium homeostatic pathway, reducing calcium overload and thus preventing excitotoxicity, mitochondrial stress, and apoptosis. However, several physiological aspects of taurine remain unclear, such as the existence or not of a specific taurine receptor. Therefore, further research is needed not only in astrocytes and neurons, but also in other glial cells in order to fully comprehend taurine metabolism and function in the brain. Nonetheless, astrocyte’s role in taurine-induced neuroprotective functions should be considered as a promising therapeutic target of several neuroinflammatory, neurodegenerative and psychiatric diseases in the near future. This review provides an overview of the significant relationship between taurine and astrocytes, as well as its homeostatic and neuroprotective role in the nervous system.
Collapse
Affiliation(s)
- Sofía Ramírez-Guerrero
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Santiago Guardo-Maya
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Germán J. Medina-Rincón
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Eduardo E. Orrego-González
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Ricardo Cabezas-Pérez
- Grupo de Investigación en Ciencias Biomédicas GRINCIBIO, Facultad de Medicina, Universidad Antonio Nariño, Bogotá, Colombia
| | - Rodrigo E. González-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
- *Correspondence: Rodrigo E. González-Reyes,
| |
Collapse
|
9
|
Yu S, Chun E, Ji Y, Lee YJ, Jin M. Effects of red ginseng on gut, microbiota, and brain in a mouse model of post-infectious irritable bowel syndrome. J Ginseng Res 2021; 45:706-716. [PMID: 34764725 PMCID: PMC8569328 DOI: 10.1016/j.jgr.2021.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 01/21/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Background Irritable bowel syndrome (IBS), the most common functional gastrointestinal disorder, is characterized by chronic abdominal pain and bowel habit changes. Although diverse complicated etiologies are involved in its pathogenesis, a dysregulated gut–brain axis may be an important factor. Red ginseng (RG), a traditional herbal medicine, is proven to have anti-inflammatory effects and improve brain function; however, these effects have not been investigated in IBS. Methods Three-day intracolonic zymosan injections were used to induce post-infectious human IBS-like symptoms in mice. The animals were randomized to receive either phosphate-buffered saline (CG) or RG (30/100/300 mg/kg) for 10 days. Amitriptyline and sulfasalazine were used as positive controls. Macroscopic scoring was performed on day 4. Visceral pain and anxiety-like behaviors were assessed by colorectal distension and elevated plus maze and open field tests, respectively, on day 10. Next-generation sequencing of gut microbiota was performed, and biomarkers involved in gut–brain axis responses were analyzed. Results Compared to CG, RG significantly decreased the macroscopic score, frequency of visceral pain, and anxiety-like behavior in the IBS mice. These effects were comparable to those after sulfasalazine and amitriptyline treatments. Moreover, RG significantly increased the proliferation of beneficial microbes, including Lactobacillus johnsonii, Lactobacillus reuteri, and Parabacteroides goldsteinii. RG significantly suppressed expression of IL-1β and c-fos in the gut and prefrontal cortex, respectively. Further, it restored the plasma levels of corticosterone to within the normal range, accompanied by an increase in adrenocorticotropic hormone. Conclusion RG may be a potential therapeutic option for the management of human IBS.
Collapse
Affiliation(s)
- Seonhye Yu
- Department of Microbiology, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Eunho Chun
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Yeounjung Ji
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Young Joo Lee
- Department of Bioscience and Biotechnology, Sejong University, Seoul, Republic of Korea
| | - Mirim Jin
- Department of Microbiology, College of Medicine, Gachon University, Incheon, Republic of Korea.,Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| |
Collapse
|
10
|
Aslan Karakelle N, Dinçer S, Yar Sağlam AS. The effect of intracerebroventricular amyloid beta 1-42 application on cognitive functions in aged rats supplemented with taurine and the change of peroxisomal proteins in this process. Brain Res Bull 2021; 172:89-97. [PMID: 33892084 DOI: 10.1016/j.brainresbull.2021.04.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The aim of our study is to investigate the change of peroxisomal proteins in the neurodegenerative and oxidative process caused by the neurotoxicity of Aβ 1-42 in aged rats supplemented with taurine and to show the possible positive effects of taurine in this process. METHODS 30 Wistar albino rats were randomly divided into 5 groups as control, sham, Aβ 1-42, taurine, and Aβ 1-42+taurine. Taurine administration continued for 6 weeks (1000 mg/kg/day with drinking water). Stereotaxic surgery was applied to all groups (intracerebroventricular per lateral ventricle needle only or 5 μl, PBS, or Aβ 1-42). Spatial learning and memory performances of the animals were evaluated with Morris water maze and elevated plus maze. The levels of MDA and GSH were measured as oxidative stress parameters in the cerebral cortex and hippocampus. Expressions of CAT, PEX14, PMP70 of peroxisomal membrane proteins were indicated by Western blot analysis. RESULTS Our results showed that injection of Aβ 1-42 decreased the spatial learning and memory performance, cortex CAT and hippocampus PEX14, PMP70 and GSH levels, and increased cortex and hippocampus MDA levels (p < 0.05). Although the administration of taurine partially ameliorated the adverse effects of Aβ 1-42 injection, a significant difference was found only at the hippocampus GSH levels (p < 0.05). Also, taurine caused anxiety at this dose (p < 0.05). DISCUSSION In conclusion, decreased peroxisomal proteins and antioxidant capacity in neurodegenerative and oxidative processes induced by intracerebroventricular Aβ 1-42 injection showed that peroxisomes may play a role in this process and taurine supplementation may have positive effects especially in increasing antioxidant capacity.
Collapse
Affiliation(s)
- Nida Aslan Karakelle
- Department of Physiology, Lokman Hekim University Faculty of Medicine, Ankara, Turkey.
| | - Sibel Dinçer
- Department of Physiology, Gazi University Faculty of Medicine, Ankara, Turkey.
| | | |
Collapse
|
11
|
Pyrzanowska J, Joniec-Maciejak I, Blecharz-Klin K, Piechal A, Mirowska-Guzel D, Fecka I, Widy-Tyszkiewicz E. Aspalathus linearis infusion affects hole-board test behaviour and amino acid concentration in the brain. Neurosci Lett 2021; 747:135680. [PMID: 33529651 DOI: 10.1016/j.neulet.2021.135680] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/05/2021] [Accepted: 01/23/2021] [Indexed: 11/24/2022]
Abstract
Rooibos tea, brewed using Aspalathus linearis leaves, is a popular South African herbal infusion, but its everyday intake is not fully described in terms of the neuropsychopharmacological outcomes. The cell-protective activity of A. linearis is connected with the ability of reducing glycaemia, inflammation as well as oxidative stress. It was already shown that "fermented" rooibos herbal tea (FRHT), which is rich in phenolic compounds, improves the cognitive performance of rats in the water maze and impacts dopaminergic striatal transmission. The present research was taken to extend the knowledge about the feasible behavioural and neurochemical implications of sustained oral FRHT consumption. We hypothesized that it might affect brain amino acid content and thus induce behaviour and neuroprotection. FRHTs of different leaf to water ratios (1:100, 2:100 and 4:100), analysed by chromatographic methods as regards their flavonoid characteristics, were given to rats as only liquid for 3 months. Their behaviour was evaluated in the hole-board test (HBT). Brain amino acids concentration was analysed in the striatum, hippocampus and prefrontal cortex by HPLC-ECD. The rats drinking rooibos tea presented increased motor activity defined as time spent on moving in the HBT. Their exploration measured by head-dipping and rearing was enhanced. Longer time of the testing-box central zone occupation indicated to reduction in anxiety-related behaviour. Excitatory amino acids (aspartate and glutamate) content was decreased in the striatum of animals drinking the infusions whereas taurine level was increased both in the striatum and hippocampus. In conclusion we suggest that long-term FRHT intake affects exploration and anxiety-related behaviour of the rats as well as exerts biochemical outcomes in the brain that support the neuroprotective impact of rooibos tea.
Collapse
Affiliation(s)
- Justyna Pyrzanowska
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1b, 02-097, Warsaw, Poland.
| | - Ilona Joniec-Maciejak
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1b, 02-097, Warsaw, Poland
| | - Kamilla Blecharz-Klin
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1b, 02-097, Warsaw, Poland
| | - Agnieszka Piechal
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1b, 02-097, Warsaw, Poland
| | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1b, 02-097, Warsaw, Poland
| | - Izabela Fecka
- Department of Pharmacognosy and Herbal Medicines, Wroclaw Medical University, Borowska 211, 50-556, Wroclaw, Poland
| | - Ewa Widy-Tyszkiewicz
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1b, 02-097, Warsaw, Poland
| |
Collapse
|