1
|
Šardzíková S, Gajewska M, Gałka N, Štefánek M, Baláž A, Garaiová M, Holič R, Świderek W, Šoltys K. Can longer lifespan be associated with gut microbiota involvement in lipid metabolism? FEMS Microbiol Ecol 2024; 100:fiae135. [PMID: 39354675 PMCID: PMC11503954 DOI: 10.1093/femsec/fiae135] [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: 12/01/2023] [Revised: 08/25/2024] [Accepted: 09/30/2024] [Indexed: 10/03/2024] Open
Abstract
Biological aging is linked to altered body composition and reduced neuroactive steroid hormones like dehydroepiandrosterone sulfate (DHEAS), which can stimulate the GABA signaling pathway via gut microbiota. Our study examined the association of gut microbiota with lifespan in mice through comprehensive analysis of its composition and functional involvement in cholesterol sulfate, a precursor of DHEAS, metabolism. We used 16S rRNA and metagenomic sequencing, followed by metabolic pathway prediction and thin layer chromatography and MALDI-TOF cholesterol sulfate identification. Significant increases in bacteria such as Bacteroides, typical for long-lived and Odoribacter and Colidextribacter, specific for short-lived mice were detected. Furthermore, for males (Rikenella and Alloprevotella) and females (Lactobacillus and Bacteroides), specific bacterial groups emerged as predictors (AUC = 1), highlighting sex-specific patterns. Long-lived mice showed a strong correlation of Bacteroides (0.918) with lipid and steroid hormone metabolism, while a negative correlation of GABAergic synapse with body weight (-0.589). We found that several Bacteroides species harboring the sulfotransferase gene and gene cluster for sulfonate donor synthesis are involved in converting cholesterol to cholesterol sulfate, significantly higher in the feces of long-lived individuals. Overall, we suggest that increased involvement of gut bacteria, mainly Bacteroides spp., in cholesterol sulfate synthesis could ameliorate aging through lipid metabolism.
Collapse
Affiliation(s)
- Sára Šardzíková
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia
| | - Marta Gajewska
- Institute of Animal Sciences, Department of Animal Genetics and Conservation, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Norbert Gałka
- Institute of Animal Sciences, Department of Animal Genetics and Conservation, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Matúš Štefánek
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia
| | - Andrej Baláž
- Faculty of Mathematics, Physics and Informatics, Comenius University in Bratislava, 842 48 Bratislava, Slovakia
| | - Martina Garaiová
- Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska Cesta 9, 84005 Bratislava, Slovakia
| | - Roman Holič
- Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska Cesta 9, 84005 Bratislava, Slovakia
| | - Wiesław Świderek
- Institute of Animal Sciences, Department of Animal Genetics and Conservation, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Katarína Šoltys
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia
| |
Collapse
|
2
|
Superti F, Russo R. Alpha-Lipoic Acid: Biological Mechanisms and Health Benefits. Antioxidants (Basel) 2024; 13:1228. [PMID: 39456481 PMCID: PMC11505271 DOI: 10.3390/antiox13101228] [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: 08/19/2024] [Revised: 09/26/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Alpha-lipoic acid (ALA) is a bioactive molecule with significant health effects. The biological action of ALA has been ascribed to the characteristic antioxidant properties of the oxidized form (ALA) and its reduced counterpart the dihydrolipoic acid (DHLA) system. The ALA/DHLA combination represents an ideal antioxidant since it can quench radicals, is able to chelate metals, is amphiphilic, and has no major adverse effects. This unique system is able to scavenge reactive oxygen species, exerting a major effect on tissue levels of reduced forms of other antioxidants, including glutathione. For this reason, ALA is also known as the "antioxidant of antioxidants". This review analyzes the antioxidant, anti-inflammatory, and neuroprotective effects of ALA and discusses its applications as an ameliorative tool for chronic diseases and those associated with oxidative stress. Results from in vitro and in vivo studies demonstrated that ALA modulates various oxidative stress pathways suggesting its application, alone or in combination with other functional substances, as a useful support in numerous conditions, in which the balance oxidant-antioxidant is disrupted, such as neurodegenerative disorders. Based on several successful clinical studies, it has been also established that oral ALA supplements are clinically useful in relieving the complications of diabetes and other disorders including cardiovascular diseases and nerve discomforts suggesting that ALA can be considered a useful approach to improving our health.
Collapse
Affiliation(s)
- Fabiana Superti
- Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, RM, Italy;
- Association for Research on Integrative Oncology Therapies, (ARTOI) Foundation, Via Ludovico Micara, 73, 00165 Rome, RM, Italy
| | - Rosario Russo
- Giellepi S.p.A., Via G. Verdi, 41/Q, 20831 Seregno, MB, Italy
| |
Collapse
|
3
|
Gómez-Fernández D, Romero-González A, Suárez-Rivero JM, Cilleros-Holgado P, Álvarez-Córdoba M, Piñero-Pérez R, Romero-Domínguez JM, Reche-López D, López-Cabrera A, Ibáñez-Mico S, Castro de Oliveira M, Rodríguez-Sacristán A, González-Granero S, García-Verdugo JM, Sánchez-Alcázar JA. A Multi-Target Pharmacological Correction of a Lipoyltransferase LIPT1 Gene Mutation in Patient-Derived Cellular Models. Antioxidants (Basel) 2024; 13:1023. [PMID: 39199267 PMCID: PMC11351668 DOI: 10.3390/antiox13081023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/12/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
Mutations in the lipoyltransferase 1 (LIPT1) gene are rare inborn errors of metabolism leading to a fatal condition characterized by lipoylation defects of the 2-ketoacid dehydrogenase complexes causing early-onset seizures, psychomotor retardation, abnormal muscle tone, severe lactic acidosis, and increased urine lactate, ketoglutarate, and 2-oxoacid levels. In this article, we characterized the disease pathophysiology using fibroblasts and induced neurons derived from a patient bearing a compound heterozygous mutation in LIPT1. A Western blot analysis revealed a reduced expression of LIPT1 and absent expression of lipoylated pyruvate dehydrogenase E2 (PDH E2) and alpha-ketoglutarate dehydrogenase E2 (α-KGDH E2) subunits. Accordingly, activities of PDH and α-KGDH were markedly reduced, associated with cell bioenergetics failure, iron accumulation, and lipid peroxidation. In addition, using a pharmacological screening, we identified a cocktail of antioxidants and mitochondrial boosting agents consisting of pantothenate, nicotinamide, vitamin E, thiamine, biotin, and α-lipoic acid, which is capable of rescuing LIPT1 pathophysiology, increasing the LIPT1 expression and lipoylation of mitochondrial proteins, improving cell bioenergetics, and eliminating iron overload and lipid peroxidation. Furthermore, our data suggest that the beneficial effect of the treatment is mainly mediated by SIRT3 activation. In conclusion, we have identified a promising therapeutic approach for correcting LIPT1 mutations.
Collapse
Affiliation(s)
- David Gómez-Fernández
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (D.G.-F.); (A.R.-G.); (J.M.S.-R.); (P.C.-H.); (M.Á.-C.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (A.L.-C.)
| | - Ana Romero-González
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (D.G.-F.); (A.R.-G.); (J.M.S.-R.); (P.C.-H.); (M.Á.-C.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (A.L.-C.)
| | - Juan M. Suárez-Rivero
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (D.G.-F.); (A.R.-G.); (J.M.S.-R.); (P.C.-H.); (M.Á.-C.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (A.L.-C.)
| | - Paula Cilleros-Holgado
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (D.G.-F.); (A.R.-G.); (J.M.S.-R.); (P.C.-H.); (M.Á.-C.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (A.L.-C.)
| | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (D.G.-F.); (A.R.-G.); (J.M.S.-R.); (P.C.-H.); (M.Á.-C.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (A.L.-C.)
| | - Rocío Piñero-Pérez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (D.G.-F.); (A.R.-G.); (J.M.S.-R.); (P.C.-H.); (M.Á.-C.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (A.L.-C.)
| | - José Manuel Romero-Domínguez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (D.G.-F.); (A.R.-G.); (J.M.S.-R.); (P.C.-H.); (M.Á.-C.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (A.L.-C.)
| | - Diana Reche-López
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (D.G.-F.); (A.R.-G.); (J.M.S.-R.); (P.C.-H.); (M.Á.-C.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (A.L.-C.)
| | - Alejandra López-Cabrera
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (D.G.-F.); (A.R.-G.); (J.M.S.-R.); (P.C.-H.); (M.Á.-C.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (A.L.-C.)
| | - Salvador Ibáñez-Mico
- Hospital Clínico Universitario Virgen de la Arrixaca, Servicio de Neuropediatría, 30120 Murcia, Spain;
| | - Marta Castro de Oliveira
- Neuropediatria, Neurolinkia, C. Jardín de la Isla, 8, Local 4 y 5, 41014 Sevilla, Spain;
- FEA Pediatría, Centro Universitario Hospitalar de Faro, R. Leão Penedo, 8000-386 Faro, Portugal
- Neuropediatria, Servicio de Pediatría, Hospital Universitario Virgen Macarena, 41009 Sevilla, Spain;
| | - Andrés Rodríguez-Sacristán
- Neuropediatria, Servicio de Pediatría, Hospital Universitario Virgen Macarena, 41009 Sevilla, Spain;
- Departamento de Farmacología, Radiología y Pediatría de la Facultad de Medicina de la Universidad de Sevilla, 41009 Sevilla, Spain
| | - Susana González-Granero
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED-ISCIII, 46980 Valencia, Spain; (S.G.-G.); (J.M.G.-V.)
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED-ISCIII, 46980 Valencia, Spain; (S.G.-G.); (J.M.G.-V.)
| | - José A. Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (D.G.-F.); (A.R.-G.); (J.M.S.-R.); (P.C.-H.); (M.Á.-C.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (A.L.-C.)
| |
Collapse
|
4
|
Sun X, Yang X, Gui W, Liu S, Gui Q. Sirtuins and autophagy in lipid metabolism. Cell Biochem Funct 2023; 41:978-987. [PMID: 37755711 DOI: 10.1002/cbf.3860] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/28/2023]
Abstract
Sirtuins are a family of NAD+ -dependent deacetylases that regulate some important biological processes, including lipid metabolism and autophagy, through their deacetylase function. Autophagy is a new discovery in the field of lipid metabolism, which may provide a new idea for the regulation of lipid metabolism. There are many tandem parts in the regulation process of lipid metabolism and autophagy of sirtuins protein family. This paper summarized these tandem parts and proposed the possibility of sirtuins regulating lipid autophagy, as well as the interaction and synergy between sirtuins protein family. Currently, some natural drugs have been reported to affect metabolism by regulating sirtuins, some of which regulate autophagy by targeting sirtuins.
Collapse
Affiliation(s)
- Xuan Sun
- Department of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiaoting Yang
- Department of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Wanfei Gui
- Department of Medicine, Chuanshan College, University of South China, Hengyang, China
| | - Songling Liu
- Department of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Qingjun Gui
- Department of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
5
|
Lapenna D. Glutathione and glutathione-dependent enzymes: From biochemistry to gerontology and successful aging. Ageing Res Rev 2023; 92:102066. [PMID: 37683986 DOI: 10.1016/j.arr.2023.102066] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 08/24/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
The tripeptide glutathione (GSH), namely γ-L-glutamyl-L-cysteinyl-glycine, is an ubiquitous low-molecular weight thiol nucleophile and reductant of utmost importance, representing the central redox agent of most aerobic organisms. GSH has vital functions involving also antioxidant protection, detoxification, redox homeostasis, cell signaling, iron metabolism/homeostasis, DNA synthesis, gene expression, cysteine/protein metabolism, and cell proliferation/differentiation or death including apoptosis and ferroptosis. Various functions of GSH are exerted in concert with GSH-dependent enzymes. Indeed, although GSH has direct scavenging antioxidant effects, its antioxidant function is substantially accomplished by glutathione peroxidase-catalyzed reactions with reductive removal of H2O2, organic peroxides such as lipid hydroperoxides, and peroxynitrite; to this antioxidant activity also contribute peroxiredoxins, enzymes further involved in redox signaling and chaperone activity. Moreover, the detoxifying function of GSH is basically exerted in conjunction with glutathione transferases, which have also antioxidant properties. GSH is synthesized in the cytosol by the ATP-dependent enzymes glutamate cysteine ligase (GCL), which catalyzes ligation of cysteine and glutamate forming γ-glutamylcysteine (γ-GC), and glutathione synthase, which adds glycine to γ-GC resulting in GSH formation; GCL is rate-limiting for GSH synthesis, as is the precursor amino acid cysteine, which may be supplemented as N-acetylcysteine (NAC), a therapeutically available compound. After its cell export, GSH is degraded extracellularly by the membrane-anchored ectoenzyme γ-glutamyl transferase, a process occurring, as GSH synthesis and export, in the γ-glutamyl cycle. GSH degradation occurs also intracellularly by the cytoplasmic enzymatic ChaC family of γ-glutamyl cyclotransferase. Synthesis and degradation of GSH, together with its export, translocation to cell organelles, utilization for multiple essential functions, and regeneration from glutathione disulfide by glutathione reductase, are relevant to GSH homeostasis and metabolism. Notably, GSH levels decline during aging, an alteration generally related to impaired GSH biosynthesis and leading to cell dysfunction. However, there is evidence of enhanced GSH levels in elderly subjects with excellent physical and mental health status, suggesting that heightened GSH may be a marker and even a causative factor of increased healthspan and lifespan. Such aspects, and much more including GSH-boosting substances administrable to humans, are considered in this state-of-the-art review, which deals with GSH and GSH-dependent enzymes from biochemistry to gerontology, focusing attention also on lifespan/healthspan extension and successful aging; the significance of GSH levels in aging is considered also in relation to therapeutic possibilities and supplementation strategies, based on the use of various compounds including NAC-glycine, aimed at increasing GSH and related defenses to improve health status and counteract aging processes in humans.
Collapse
Affiliation(s)
- Domenico Lapenna
- Dipartimento di Medicina e Scienze dell'Invecchiamento, and Laboratorio di Fisiopatologia dello Stress Ossidativo, Center for Advanced Studies and Technology (CAST, former CeSI-MeT, Center of Excellence on Aging), Università degli Studi "G. d'Annunzio" Chieti Pescara, U.O.C. Medicina Generale 2, Ospedale Clinicizzato "Santissima Annunziata", Via dei Vestini, 66100 Chieti, Italy.
| |
Collapse
|
6
|
Buitrago Ramírez JR, Marreiro Gomes RM, de Sousa Araujo AC, Muñoz Buitrago SA, Piraine Souza J, Monserrat JM. The Effects of Lipoic Acid on Yolk Nutrient Utilization, Energy Metabolism, and Redox Balance over Time in Artemia sp. Antioxidants (Basel) 2023; 12:1439. [PMID: 37507976 PMCID: PMC10376159 DOI: 10.3390/antiox12071439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/30/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Lipoic acid (LA) is a mitochondrial coenzyme that, depending on the concentration and exposure time, can behave as an antioxidant or pro-oxidant agent and has a proven ability to modulate metabolism by promoting lipid and glucose oxidation for energy production. To assess the effects of LA on energy metabolism and redox balance over time, Artemia sp. nauplii was used as an animal model. The administered concentrations of the antioxidant were 0.05, 0.1, 0.5, 1.0, 5.0, and 10.0 µM. Therefore, possible differences in protein, triglyceride, glucose, and lactate concentrations in the artemia samples and total ammoniacal nitrogen (TAN) in the culture water were evaluated. We also measured the effects of LA on in vivo activity of the electron transport system (ETS), antioxidant capacity, and production of reactive oxygen species (ROS) at 6, 12, 18, and 24 h post-hatching. There was a decrease in glucose concentration in the LA-treated animals, and a decrease in ammonia production was observed in the 0.5 µM LA treatment. ETS activity was positively regulated by the addition of LA, with the most significant effects at concentrations of 5.0 and 10.0 µM at 12 and 24 h. For ETS activity, treatments with LA presented the highest values at 24 h, a period when ROS production decreased significantly, for the treatment with 10.0 µM. LA showed positive regulation of energy metabolism together with a decrease in ROS and TAN excretion.
Collapse
Affiliation(s)
- Juan Rafael Buitrago Ramírez
- Programa de Pós Graduação em Aquicultura, Instituto de Oceanografia (IO), Universidade Federal do Rio Grande-FURG, Rua do Hotel, n° 2, Cassino, Rio Grande 96210-030, RS, Brazil
- Laboratório de Bioquímica Funcional de Organismos Aquáticos (BIFOA), Instituto de Oceanografia (IO), Universidade Federal do Rio Grande-FURG, Rua do Hotel, n° 2, Cassino, Rio Grande 96210-030, RS, Brazil
| | - Robson Matheus Marreiro Gomes
- Programa de Pós Graduação em Aquicultura, Instituto de Oceanografia (IO), Universidade Federal do Rio Grande-FURG, Rua do Hotel, n° 2, Cassino, Rio Grande 96210-030, RS, Brazil
- Laboratório de Bioquímica Funcional de Organismos Aquáticos (BIFOA), Instituto de Oceanografia (IO), Universidade Federal do Rio Grande-FURG, Rua do Hotel, n° 2, Cassino, Rio Grande 96210-030, RS, Brazil
| | - Alan Carvalho de Sousa Araujo
- Programa de Pós Graduação em Aquicultura, Instituto de Oceanografia (IO), Universidade Federal do Rio Grande-FURG, Rua do Hotel, n° 2, Cassino, Rio Grande 96210-030, RS, Brazil
- Laboratório de Bioquímica Funcional de Organismos Aquáticos (BIFOA), Instituto de Oceanografia (IO), Universidade Federal do Rio Grande-FURG, Rua do Hotel, n° 2, Cassino, Rio Grande 96210-030, RS, Brazil
| | - Sonia Astrid Muñoz Buitrago
- Programa de Pós Graduação em Aquicultura, Instituto de Oceanografia (IO), Universidade Federal do Rio Grande-FURG, Rua do Hotel, n° 2, Cassino, Rio Grande 96210-030, RS, Brazil
- Laboratório de Bioquímica Funcional de Organismos Aquáticos (BIFOA), Instituto de Oceanografia (IO), Universidade Federal do Rio Grande-FURG, Rua do Hotel, n° 2, Cassino, Rio Grande 96210-030, RS, Brazil
| | - Jean Piraine Souza
- Programa de Pós Graduação em Aquicultura, Instituto de Oceanografia (IO), Universidade Federal do Rio Grande-FURG, Rua do Hotel, n° 2, Cassino, Rio Grande 96210-030, RS, Brazil
- Laboratório de Bioquímica Funcional de Organismos Aquáticos (BIFOA), Instituto de Oceanografia (IO), Universidade Federal do Rio Grande-FURG, Rua do Hotel, n° 2, Cassino, Rio Grande 96210-030, RS, Brazil
| | - José María Monserrat
- Programa de Pós Graduação em Aquicultura, Instituto de Oceanografia (IO), Universidade Federal do Rio Grande-FURG, Rua do Hotel, n° 2, Cassino, Rio Grande 96210-030, RS, Brazil
- Laboratório de Bioquímica Funcional de Organismos Aquáticos (BIFOA), Instituto de Oceanografia (IO), Universidade Federal do Rio Grande-FURG, Rua do Hotel, n° 2, Cassino, Rio Grande 96210-030, RS, Brazil
- Instituto of Ciências Biológicas (ICB), Universidade Federal do Rio Grande-FURG, Av. Itália, Km 08, Rio Grande 96201-900, RS, Brazil
| |
Collapse
|
7
|
Fiore G, Pascuzzi MC, Di Profio E, Corsello A, Agostinelli M, La Mendola A, Milanta C, Campoy C, Calcaterra V, Zuccotti G, Verduci E. Bioactive compounds in childhood obesity and associated metabolic complications: Current evidence, controversies and perspectives. Pharmacol Res 2023; 187:106599. [PMID: 36503001 DOI: 10.1016/j.phrs.2022.106599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022]
Abstract
Obesity represents the most frequent chronic disease among children worldwide, with a significant global burden on society. Metabolically unhealthy obesity (MUO) can affect children since their first years of life, and novel therapeutic strategies to tackle metabolic complications are under investigation. This review focuses on bioactive compounds and their possible beneficial effects on obesity, particularly omega-3, docosahexaenoic acid, vitamin D, biotics, polysaccharide macromolecules, polyphenols, inositols, alpha lipoic acid, and bromelaine. Our aim is to summarize current evidence about bioactive compounds in the treatment of obesity, highlighting recent findings on their use in children and adolescents. Most studied molecules are omega-3 and vitamin D, despite the heterogeneity between the studies. Moreover, given the emerging interest in the gut-brain axis in the link between metabolic health and microbiota, various studies on prebiotics, probiotics, synbiotics, postbiotics and polysaccharide macromolecules have been considered. Some preclinical studies seem to highlight a possible role of the polyphenols, even if their clinical evidence is still discussed. Lastly, we describe possible effects of inositols and alpha-lipoic acid. Despite some dietary supplements seem to be promising in overweight subjects, only in a few of them a dose/response efficacy has been found in the pediatric age. Innovative, well-designed and targeted clinical trials are then needed to prove the beneficial effects of these compounds that could support the standard behavioral therapy for obesity.
Collapse
Affiliation(s)
- Giulia Fiore
- Department of Paediatrics, Vittore Buzzi Children's Hospital, University of Milan, Italy.
| | | | - Elisabetta Di Profio
- Department of Paediatrics, Vittore Buzzi Children's Hospital, University of Milan, Italy.
| | - Antonio Corsello
- Department of Paediatrics, Vittore Buzzi Children's Hospital, University of Milan, Italy.
| | - Marta Agostinelli
- Department of Paediatrics, Vittore Buzzi Children's Hospital, University of Milan, Italy.
| | - Alice La Mendola
- Department of Paediatrics, Vittore Buzzi Children's Hospital, University of Milan, Italy.
| | - Chiara Milanta
- Department of Paediatrics, Vittore Buzzi Children's Hospital, University of Milan, Italy.
| | - Cristina Campoy
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain; EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain; Spanish Network of Biomedical Research in Epidemiology and Public Health (CIBERESP), Granada's node, Institute of Health Carlos III, 28029 Madrid, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), San Cecilio University Hospital. Health Sciences Technological Park, 18016 Granada, Spain.
| | - Valeria Calcaterra
- Department of Paediatrics, Vittore Buzzi Children's Hospital, University of Milan, Italy; Pediatric and Adolescent Unit, Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy.
| | - Gianvincenzo Zuccotti
- Department of Paediatrics, Vittore Buzzi Children's Hospital, University of Milan, Italy; Department of Biomedical and Clinical Sciences L. Sacco, University of Milan, 20144 Milan, Italy; Pediatric Clinical Research Center, Fondazione Romeo ed Enrica Invernizzi, University of Milan, Milan, Italy.
| | - Elvira Verduci
- Department of Paediatrics, Vittore Buzzi Children's Hospital, University of Milan, Italy; Department of Health Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
8
|
Wu QJ, Zhang TN, Chen HH, Yu XF, Lv JL, Liu YY, Liu YS, Zheng G, Zhao JQ, Wei YF, Guo JY, Liu FH, Chang Q, Zhang YX, Liu CG, Zhao YH. The sirtuin family in health and disease. Signal Transduct Target Ther 2022; 7:402. [PMID: 36581622 PMCID: PMC9797940 DOI: 10.1038/s41392-022-01257-8] [Citation(s) in RCA: 212] [Impact Index Per Article: 106.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/10/2022] [Accepted: 11/18/2022] [Indexed: 12/30/2022] Open
Abstract
Sirtuins (SIRTs) are nicotine adenine dinucleotide(+)-dependent histone deacetylases regulating critical signaling pathways in prokaryotes and eukaryotes, and are involved in numerous biological processes. Currently, seven mammalian homologs of yeast Sir2 named SIRT1 to SIRT7 have been identified. Increasing evidence has suggested the vital roles of seven members of the SIRT family in health and disease conditions. Notably, this protein family plays a variety of important roles in cellular biology such as inflammation, metabolism, oxidative stress, and apoptosis, etc., thus, it is considered a potential therapeutic target for different kinds of pathologies including cancer, cardiovascular disease, respiratory disease, and other conditions. Moreover, identification of SIRT modulators and exploring the functions of these different modulators have prompted increased efforts to discover new small molecules, which can modify SIRT activity. Furthermore, several randomized controlled trials have indicated that different interventions might affect the expression of SIRT protein in human samples, and supplementation of SIRT modulators might have diverse impact on physiological function in different participants. In this review, we introduce the history and structure of the SIRT protein family, discuss the molecular mechanisms and biological functions of seven members of the SIRT protein family, elaborate on the regulatory roles of SIRTs in human disease, summarize SIRT inhibitors and activators, and review related clinical studies.
Collapse
Affiliation(s)
- Qi-Jun Wu
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tie-Ning Zhang
- grid.412467.20000 0004 1806 3501Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Huan-Huan Chen
- grid.412467.20000 0004 1806 3501Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xue-Fei Yu
- grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jia-Le Lv
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu-Yang Liu
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ya-Shu Liu
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Gang Zheng
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun-Qi Zhao
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi-Fan Wei
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jing-Yi Guo
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fang-Hua Liu
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qing Chang
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi-Xiao Zhang
- grid.412467.20000 0004 1806 3501Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cai-Gang Liu
- grid.412467.20000 0004 1806 3501Department of Cancer, Breast Cancer Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu-Hong Zhao
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
9
|
Arisha SM. Alpha-lipoic acid-role in improving both reserpine toxicity and paroxetine treatment in the cerebral cortex of albino rats; histological, ultrastructural, immunohistohemical and biochemical studies. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2022. [DOI: 10.1186/s43088-022-00265-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Reserpine is a monoamine depletory drug cause oxidative damage and used to induce depression-like features in rodent model. Paroxetine is an antidepressant drug that exerts its effects by inhibiting dopaminergic neurons although it may exert much pathological damage. Alpha-lipoic acid (ALA) is an endogenous antioxidant co-factor of important enzymatic complexes. The present study was aimed to elucidate the possible protective effect of ALA in the improvement of the deleterious cerebral cortex injury after reserpine and paroxetine treatment. Forty adult male albino rats were equally divided into 5 groups. Group I served as control group orally treated with saline solution all the experiment period. Group II animals orally treated with ALA (200 mg/kg/day) for six weeks. The induction of depression-like features occurred when the rest of animals were intraperitoneally treated with 25 mg/kg of reserpine once daily for consecutive 14 day. Then these animals were divided into; Group III (reserpine group) animals in this group were sacrificed on 15th day. Group IV; reserpine-treated animals were treated with paroxetine (20 mg/kg) daily for 6 weeks. Group V, animals in this group were received paroxetine and ALA daily for 6 weeks.
Results
Reserpine-treated rats showed disorganized layers of cerebral cortex with degenerative, apoptotic and necrotic changes. Ultrastructure changes include both pyramidal and granule cells with severe degenerative, necrotic and apoptotic features. The nuclei appeared pyknotic; irregular with chromatin condensation as well as the cytoplasm of these cells contained many degenerated organelles. In addition, a significant increase in total oxidative stress and decrease in total antioxidant capacity, norepinephrine, dopamine and serotonin levels were recorded. The same treatment showed significant decrease in proliferating cell nuclear antigen (PCNA) expression and significant increase in caspase-3 expression in the granule and pyramidal cells. After paroxetine-treatment these parameters were more or less similar to those observed in reserpine-treated ones. While an obvious improvement was appeared when animal treated with both paroxetine and ALA and; all parameters restored its normal features.
Conclusions
This study concluded that; ALA treatment attenuated the cerebral injury induced by reserpine and improved the effects of paroxetine in rats due to its anti-inflammatory, anti-apoptotic and antioxidant activities.
Collapse
|
10
|
Mohammadshahi M, Zakizadeh E, Ahmadi-Angali K, Ravanbakhsh M, Helli B. The synergic effects of alpha-lipoic acid supplementation and electrical isotonic contraction on anthropometric measurements and the serum levels of VEGF, NO, sirtuin-1, and PGC1-α in obese people undergoing a weight loss diet. Arch Physiol Biochem 2022; 128:1195-1201. [PMID: 32407179 DOI: 10.1080/13813455.2020.1762660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Background: The anti-obesity effects of Alpha-lipoic acid (α-LA) and isotonic contraction has been reported. However, the underlying mechanism is not fully understood. This study aimed to investigate the effect of 1200 mg/day α-LA supplementation and 3 sessions per week of Faradic (an electrical stimulating system) on anthropometric parameters, body composition, VEGF, Sirtuin-1, nitric oxide (NO), and PGC1-α in obese people undergoing a weight loss regime.Methods: This randomised clinical trial was carried out on 100 obese adults. The subjects were randomly assigned to four groups of 25 subjects including Faradic, α-LA, α-LA + Faradic, and control. A Bio Impedance Analyser (BIA) was used to estimate anthropometric measurements including weight, body mass index (BMI), fat mass, and fat free mass. The serum levels of Sirtuin-1, PGC1-α, VEGF, and NO levels were measured. All measurements were done at baseline and after 8 weeks of the intervention.Results: A significant weight reduction was observed in all four groups compared to baseline (p<.01). The placebo group had significantly higher weight, BMI, weight circumstance (WC), and body fat (BF) compared with the other groups. The α-LA + Faradic group had significantly lower weight, BMI, BF, WC than control, faradic, and α-LA groups and higher, Sirtuin and PGC than the control group (all p < .05).Conclusions: The findings indicated that the α-LA and Faradic interventions may have a synergistic effect on weight, BMI, BF, WC, and SLM, possibly through changes in serum level of VEGF, NO, and PGC. Further studies are warranted to clarify the mutual effects of -α-LA and Faradic on obesity and its molecular mechanisms. Name of the registry: Iranian Registry of Clinical TrialsTrial registration number: IRCT20131117015424N2Date of registration: 04/04/2018URL of trial registry record: https://www.irct.ir/search/result?query=IRCT20131117015424N2.
Collapse
Affiliation(s)
- Majid Mohammadshahi
- Department of Nutrition, Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical sciences, Ahvaz, Iran
| | - Elahe Zakizadeh
- Department of Nutrition, Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical sciences, Ahvaz, Iran
| | - Kambiz Ahmadi-Angali
- Faculty of Public Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Majid Ravanbakhsh
- Musculoskeletal Rehabilitation Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bijan Helli
- Department of Nutrition, Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical sciences, Ahvaz, Iran
| |
Collapse
|
11
|
Liu Y, Li Y, Wang J, Yang L, Yu X, Huang P, Song H, Zheng P. Salvia-Nelumbinis naturalis improves lipid metabolism of NAFLD by regulating the SIRT1/AMPK signaling pathway. BMC Complement Med Ther 2022; 22:213. [PMID: 35945571 PMCID: PMC9361555 DOI: 10.1186/s12906-022-03697-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 08/04/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Salvia-Nelumbinis naturalis (SNN), the extract of Chinese herbal medicine, has shown effects on NAFLD. This study aims to explore the underlying mechanism of SNN for regulating the lipid metabolism disorder in NAFLD based on the SIRT1/AMPK signaling pathway.
Methods
Male C57BL/6J mice fed with a high-fat diet (HFD) were used to establish the NAFLD model. Dynamic changes of mice including body weight, liver weight, serological biochemical indexes, liver histopathological changes, and protein level of AMPK and SIRT1 were monitored. After18 weeks, SNN treatment was administrated to the NAFLD mice for another 4 weeks. Besides the aforementioned indices, TC and TG of liver tissues were also measured. Western blot and quantitative RT-PCR were used to detect the expression and/or activation of SIRT1 and AMPK, as well as the molecules associated with lipid synthesis and β-oxidation. Furthermore, AML12 cells with lipid accumulation induced by fatty acids were treated with LZG and EX527 (SIRT1 inhibitor) or Compound C (AMPK inhibitor ) to confirm the potential pharmacological mechanism.
Results
Dynamic observation found the mice induced by HFD with gradually increased body and liver weight, elevated serum cholesterol, hepatic lipid accumulation, and liver injury. After 16 weeks, these indicators have shown obvious changes. Additionally, the hepatic level of SIRT1 and AMPK activation was identified gradually decreased with NAFLD progress. The mice with SNN administration had lower body weight, liver weight, and serum level of LDL-c and ALT than those of the NAFLD model. Hepatosteatosis and hepatic TG content in the liver tissues of the SNN group were significantly reduced. When compared with control mice, the NAFLD mice had significantly decreased hepatic expression of SIRT1, p-AMPK, p-ACC, ACOX1, and increased total Acetylated-lysine, SUV39H2, and SREBP-1c. The administration of SNN reversed the expression of these molecules. In vitro experiments showed the effect of SNN in ameliorating hepatosteatosis and regulating the expression of lipid metabolism-related genes in AML12 cells, which were diminished by EX527 or Compound C co-incubation.
Conclusions
Taken together, the SIRT1/AMPK signaling pathway, involved in hepatic lipid synthesis and degradation, plays a pivotal role in the pathogenesis of NAFLD development. The regulation of SIRT1/AMPK signaling greatly contributes to the underlying therapeutic mechanism of SNN for NAFLD.
Collapse
|
12
|
Ashkar F, Bhullar KS, Wu J. The Effect of Polyphenols on Kidney Disease: Targeting Mitochondria. Nutrients 2022; 14:nu14153115. [PMID: 35956292 PMCID: PMC9370485 DOI: 10.3390/nu14153115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 02/01/2023] Open
Abstract
Mitochondrial function, including oxidative phosphorylation (OXPHOS), mitochondrial biogenesis, and mitochondria dynamics, are essential for the maintenance of renal health. Through modulation of mitochondrial function, the kidneys are able to sustain or recover acute kidney injury (AKI), chronic kidney disease (CKD), nephrotoxicity, nephropathy, and ischemia perfusion. Therapeutic improvement in mitochondrial function in the kidneys is related to the regulation of adenosine triphosphate (ATP) production, free radicals scavenging, decline in apoptosis, and inflammation. Dietary antioxidants, notably polyphenols present in fruits, vegetables, and plants, have attracted attention as effective dietary and pharmacological interventions. Considerable evidence shows that polyphenols protect against mitochondrial damage in different experimental models of kidney disease. Mechanistically, polyphenols regulate the mitochondrial redox status, apoptosis, and multiple intercellular signaling pathways. Therefore, this review attempts to focus on the role of polyphenols in the prevention or treatment of kidney disease and explore the molecular mechanisms associated with their pharmacological activity.
Collapse
Affiliation(s)
| | | | - Jianping Wu
- Correspondence: ; Tel.: +1-780-492-6885; Fax: +1-780-492-8524
| |
Collapse
|
13
|
He B, Wang Z, Moreau R. Chylomicron production is repressed by RPTOR knockdown, R-α-lipoic acid and 4-phenylbutyric acid in human enterocyte-like Caco-2 cells. J Nutr Biochem 2022; 108:109087. [PMID: 35691593 DOI: 10.1016/j.jnutbio.2022.109087] [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: 08/17/2021] [Revised: 03/28/2022] [Accepted: 05/14/2022] [Indexed: 10/18/2022]
Abstract
Although the role of mechanistic target of rapamycin complex 1 (mTORC1) in lipid metabolism has been the subject of previous research, its function in chylomicron production is not known. In this study, we created three stable human colorectal adenocarcinoma Caco-2 cell lines exhibiting normal, low or high mTORC1 kinase activity, and used these cells to investigate the consequences of manipulating mTORC1 activity on enterocyte differentiation and chylomicron-like particle production. Constitutively active mTORC1 induced Caco-2 cell proliferation and differentiation (as judged by alkaline phosphatase activity) but weakened transepithelial electrical resistance (TEER). Repressed mTORC1 activity due to the knockdown of RPTOR significantly decreased the expression of lipogenic genes FASN, DGAT1 and DGAT2, lipoprotein assembly genes APOB and MTTP, reduced protein expression of APOB, MTTP and FASN, downregulated the gene expression of very long-chain fatty acyl-CoA ligase (FATP2), acyl-CoA binding protein (DBI), and prechylomicron transport vesicle-associated proteins VAMP7 (vesicle-associated membrane protein 7) and SAR1B (secretion associated Ras related GTPase 1B) resulting in the repression of apoB-containing triacylglycerol-rich lipoprotein secretion. Exposure of Caco-2 cells harboring a constitutively active mTORC1 to short-chain fatty acid derivatives, R-α-lipoic acid and 4-phenylbutyric acid, downregulated chylomicron-like particle secretion by interfering with the lipidation and assembly of the particles, and concomitantly repressed mTORC1 activity with no change to Raptor abundance or PRAS40 (Thr246) phosphorylation. R-α-lipoic acid and 4-phenylbutyric acid may be useful to mitigate intestinal lipoprotein overproduction and associated postprandial inflammation.
Collapse
Affiliation(s)
- Bo He
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Zhigang Wang
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Régis Moreau
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| |
Collapse
|
14
|
Chakraborty A, Minor KE, Nizami HL, Chiao YA, Lee CF. Harnessing NAD + Metabolism as Therapy for Cardiometabolic Diseases. Curr Heart Fail Rep 2022; 19:157-169. [PMID: 35556214 PMCID: PMC9339518 DOI: 10.1007/s11897-022-00550-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2022] [Indexed: 01/10/2023]
Abstract
PURPOSE OF THE REVIEW This review summarizes current understanding on the roles of nicotinamide adenine dinucleotide (NAD+) metabolism in the pathogeneses and treatment development of metabolic and cardiac diseases. RECENT FINDINGS NAD+ was identified as a redox cofactor in metabolism and a co-substrate for a wide range of NAD+-dependent enzymes. NAD+ redox imbalance and depletion are associated with many pathologies where metabolism plays a key role, for example cardiometabolic diseases. This review is to delineate the current knowledge about harnessing NAD+ metabolism as potential therapy for cardiometabolic diseases. The review has summarized how NAD+ redox imbalance and depletion contribute to the pathogeneses of cardiometabolic diseases. Therapeutic evidence involving activation of NAD+ synthesis in pre-clinical and clinical studies was discussed. While activation of NAD+ synthesis shows great promise for therapy, the field of NAD+ metabolism is rapidly evolving. Therefore, it is expected that new mechanisms will be discovered as therapeutic targets for cardiometabolic diseases.
Collapse
Affiliation(s)
- Akash Chakraborty
- Cardiovascular Biology Research Program, MS 45, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Keaton E Minor
- Cardiovascular Biology Research Program, MS 45, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Hina Lateef Nizami
- Cardiovascular Biology Research Program, MS 45, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Ying Ann Chiao
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Chi Fung Lee
- Cardiovascular Biology Research Program, MS 45, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA.
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
15
|
Lu Y, Shao M, Xiang H, Wang J, Ji G, Wu T. Qinggan Huoxue Recipe Alleviates Alcoholic Liver Injury by Suppressing Endoplasmic Reticulum Stress Through LXR-LPCAT3. Front Pharmacol 2022; 13:824185. [PMID: 35431945 PMCID: PMC9009225 DOI: 10.3389/fphar.2022.824185] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/07/2022] [Indexed: 11/25/2022] Open
Abstract
Endoplasmic reticulum stress (ERS) plays a key role in alcohol liver injury (ALI). Lysophosphatidylcholine acyltransferase 3 (LPCAT3) is a potential modifier of ERS. It was examined whether the protective effect of Qinggan Huoxue Recipe (QGHXR) against ALI was associated with LPCAT3 by suppressing ERS from in vivo and in vitro experiment. Male C57BL/6 mice were randomly divided into five groups (n = 10, each) and treated for 8 weeks as follows: the control diet-fed group (pair-fed), ethanol diet-fed group (EtOH-fed), QGHXR group (EtOH-fed + QGHXR), Qinggan recipe group (EtOH-fed + QGR), and Huoxue recipe group (EtOH-fed + HXR). QGHXR, QGR, and HXR groups attenuated liver injury mainly manifested in reducing serum ALT, AST, and liver TG and reducing the severity of liver cell necrosis and steatosis in ALI mouse models. QGHXR mainly inhibited the mRNA levels of Lxrα, Perk, Eif2α, and Atf4 and activated the mRNA levels of Lpcat3 and Ire1α, while inhibiting the protein levels of LPCAT3, eIF2α, IRE1α, and XBP1u and activating the protein levels of GRP78 to improve ALI. QGR was more inclined to improve ALI by inhibiting the mRNA levels of Lxrα, Perk, Eif2α, Atif4, and Chop and activating the mRNA levels of Lpcat3 and Ire1α while inhibiting the protein levels of LPCAT3, PERK, eIF2α, IRE1α, and XBP1u. HXR was more inclined to improve ALI by inhibiting the mRNA levels of Perk, Eif2α, Atf4, and Chop mRNA while inhibiting the protein levels of LPCAT3, PERK, eIF2α, IRE1α, and XBP1u and activating the protein levels of GRP78. Ethanol (100 mM) was used to intervene HepG2 and AML12 to establish an ALI cell model and treated by QGHXR-, QGR-, and HXR-medicated serum (100 mg/L). QGHXR, QGR, and HXR groups mainly reduced the serum TG level and the expression of inflammatory factors such as IL-6 and TNF-α in the liver induced by ethanol. In AML12 cells, QGHXR and its disassembly mainly activated Grp78 mRNA expression together with inhibiting Lxrα, Lpcat3, Eif2α, Atf4, and Xbp1 mRNA expression. The protein expression of eIF2α and XBP1u was inhibited, and the expression of PERK and GRP78 was activated to alleviate ALI. In HepG2 cells, QGHXR mainly alleviated ALI by inhibiting the mRNA expression of LPCAT3, CHOP, IRE1α, XBP1, eIF2α, CHOP, and IRE1α protein. QGR was more inclined to inhibit the protein expression of PERK, and HXR was more likely to inhibit the protein expression of ATF4.
Collapse
Affiliation(s)
- Yifei Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingmei Shao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Teaching Department, Baoshan District Hospital of Intergrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Hongjiao Xiang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Junmin Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Guang Ji, ; Tao Wu,
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Guang Ji, ; Tao Wu,
| |
Collapse
|
16
|
Najafi N, Mehri S, Ghasemzadeh Rahbardar M, Hosseinzadeh H. Effects of alpha lipoic acid on metabolic syndrome: A comprehensive review. Phytother Res 2022; 36:2300-2323. [PMID: 35234312 DOI: 10.1002/ptr.7406] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 01/22/2022] [Accepted: 01/26/2022] [Indexed: 12/13/2022]
Abstract
Metabolic syndrome (MetS) is a multifactorial disease with medical conditions such as hypertension, diabetes, obesity, dyslipidemia, and insulin resistance. Alpha-lipoic acid (α-LA) possesses various pharmacological effects, including antidiabetic, antiobesity, hypotensive, and hypolipidemia actions. It exhibits reactive oxygen species scavenger properties against oxidation and age-related inflammation and refines MetS components. Also, α-LA activates the 5' adenosine monophosphate-activated protein kinase and inhibits the NFκb. It can decrease cholesterol biosynthesis, fatty acid β-oxidation, and vascular stiffness. α-LA decreases lipogenesis, cholesterol biosynthesis, low-density lipoprotein and very low-density lipoprotein levels, and atherosclerosis. Moreover, α-LA increases insulin secretion, glucose transport, and insulin sensitivity. These changes occur via PI3K/Akt activation. On the other hand, α-LA treats central obesity by increasing adiponectin levels and mitochondrial biogenesis and can reduce food intake mainly by SIRT1 stimulation. In this review, the most relevant articles have been discussed to determine the effects of α-LA on different components of MetS with a special focus on different molecular mechanisms behind these effects. This review exhibits the potential properties of α-LA in managing MetS; however, high-quality studies are needed to confirm the clinical efficacy of α-LA.
Collapse
Affiliation(s)
- Nahid Najafi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmacodynamics and Toxicology, School Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soghra Mehri
- Department of Pharmacodynamics and Toxicology, School Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
17
|
Ramos CSCB, Silva VAPD, Corrêa LBNS, Abboud RDS, Boaventura GT, Chagas MA. Long-Term Alpha-Lipoic Acid (ALA) Antioxidant Therapy Reduces Damage in the Cardiovascular System of Streptozotocin-Induced Diabetic Rats. J Diet Suppl 2021; 20:444-458. [PMID: 34806528 DOI: 10.1080/19390211.2021.2004290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Cellular damage, lipid oxidation and the action of inflammatory cytokines are implicated in the evolution of vascular complications associated with diabetes mellitus (DM) hyperglycemia. In contrast, alpha-lipoic acid (ALA) is a supplement with antioxidant and anti-inflammatory effects. This study aims to evaluate the overall effects of ALA supplementation by assessing its long-term systemic action on the vascular morphology of rats with induced diabetes. A total of 28 male rats were divided into 4 groups with seven animals each. For diabetes induction, two groups received streptozotocin. The animals in the lipoic and diabetic lipoic groups received ALA supplement. After 8 weeks the animals were anesthetized and blood collected was for hematological, biochemical and serological analyses. The thoracic aorta was removed, processed for paraffin and histological sections were stained for morphometric analysis. In diabetic groups, an improvement in hematological profile was observed, with platelet reduction in the diabetic lipoic group. ALA addition to the diet attenuated the negative effects in lipid profile; moreover, renal, hepatic and inflammatory parameters reduced or displayed values close to the values of the normal control. The anti-inflammatory effect of ALA was observed in diabetic animals, with a reduction of inflammatory citokines, accompanied by the improvement of morphological parameters in the aorta. In conclusion, long-term supplementation with ALA promoted systemic improvement, thus reducing the risk of vascular diseases. The changes in the renal and hepatic parameters without any negative impact in the hematological profile also show that ALA can be indicated as a low-risk prophylaxis or complementary therapy.
Collapse
Affiliation(s)
- Cristiane Simões Coelho Britto Ramos
- Department of Morphology, Laboratory of Cellular and Extracellular Biomorphology. Biomedical Institute, Federal Fluminense University, Niteroi, Brazil
| | - Vivian Alves Pereira da Silva
- Department of Morphology, Laboratory of Cellular and Extracellular Biomorphology. Biomedical Institute, Federal Fluminense University, Niteroi, Brazil
| | - Lanna Beatriz Neves Silva Corrêa
- Department of Morphology, Laboratory of Cellular and Extracellular Biomorphology. Biomedical Institute, Federal Fluminense University, Niteroi, Brazil
| | - Renato de Souza Abboud
- Department of Morphology, Laboratory of Cellular and Extracellular Biomorphology. Biomedical Institute, Federal Fluminense University, Niteroi, Brazil
| | - Gilson Teles Boaventura
- Laboratory of Experimental Nutrition. Department of Nutrition and Dietetics, Federal Fluminense University, Rio de Janeiro, Brazil
| | - Mauricio Alves Chagas
- Department of Morphology, Laboratory of Cellular and Extracellular Biomorphology. Biomedical Institute, Federal Fluminense University, Niteroi, Brazil
| |
Collapse
|
18
|
Mahmoudinezhad M, Farhangi MA. Alpha lipoic acid supplementation affects serum lipids in a dose and duration-dependent manner in different health status. INT J VITAM NUTR RES 2021. [PMID: 34605276 DOI: 10.1024/0300-9831/a000732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background: Many studies have investigated the effect of ALA supplementation on lipid profile, and different results have been obtained from these studies. The current systematic review and dose-response meta-analysis was conducted to achive a strong conclusion about the effect of ALA supplementation on lipid profile including total cholesterol (TC), low- and high-density lipoprotein cholesterol (LDL, HDL) and triglyceride (TG). Methods: A systematic search was performed in PubMed, SCOPUS, ProQuest and Embase for randomized placebo-controlled human trials that examined the effect of ALA supplementation on lipid profile up to November 2020. The dose and duration of ALA supplementation for included studies were ranged between 300-1200 mg/d and 2-16 weeks respectively. Weighted mean differences (WMD) and 95% confidence intervals (CIs) were used to evaluate the effect size. Cochran's Q and I2 tests were also used to assess between-study's heterogeneity. In addition, subgroup analysis was performed to investigate potential sources of heterogeneity. Dose-response relationship was done using fractional polynomial modeling. Results: Among all eligible studies, 12 studies with a total number of 548 participants were selected. ALA caused a significant reduction on TC (WMD): -10.78 mg/dl, 95% CI: -20.81, -0.74, P=0.002), LDL (WMD: -10.88 mg/dl, 95% CI: -19.52, -2.24, P=0.014) and TG (WMD: -31.02 mg/dl, 95% CI: -49.63, -12.42, P<0.001). There was also a non-significant increaes in HDL concentrations. In addition, dose-response analysis showed a positive association between LDL (Pnon-linearity=0.026), TG (Pnon-linearity<0.001) and duration of intervention in a non-linear model. Conclusion: The present meta-analysis revealed the beneficial effects of ALA supplementation on TC, LDL and TG levels. Moreover, the beneficial effects of ALA supplementation on LDL and TG levels was duration-dependent.
Collapse
|
19
|
Antioxidants Supplementation Reduces Ceramide Synthesis Improving the Cardiac Insulin Transduction Pathway in a Rodent Model of Obesity. Nutrients 2021; 13:nu13103413. [PMID: 34684414 PMCID: PMC8541644 DOI: 10.3390/nu13103413] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 01/10/2023] Open
Abstract
Obesity-related disruption in lipid metabolism contributes to cardiovascular dysfunction. Despite numerous studies on lipid metabolism in the left ventricle, there is no data describing the influence of n-acetylcysteine (NAC) and α-lipoic acid (ALA), as glutathione precursors, on sphingolipid metabolism, and insulin resistance (IR) occurrence. The aim of our experiment was to evaluate the influence of chronic antioxidants administration on myocardial sphingolipid state and intracellular insulin signaling as a potential therapeutic strategy for obesity-related cardiovascular IR. The experiment was conducted on male Wistar rats fed a standard rodent chow or a high-fat diet with intragastric administration of NAC or ALA for eight weeks. Cardiac and plasma sphingolipid species were assessed by high-performance liquid chromatography (HPLC). The proteins expressed from sphingolipid and insulin signaling pathways were determined by Western blot. Antioxidant supplementation markedly reduced ceramide accumulation by lowering the expression of selected proteins from the sphingolipid pathway and simultaneously increased the myocardial sphingosine-1-phosphate level. Moreover, NAC and ALA augmented the expression of GLUT4 and the phosphorylation state of Akt (Ser473) and GSK3β (Ser9), which improved the intracellular insulin transduction pathway. Based on our results, we may postulate that NAC and ALA have a beneficial influence on the cardiac ceramidose under IR conditions.
Collapse
|
20
|
Onder NT, Alcay S, Nur Z. Effects of alpha-lipoic acid on ram semen cryopreservation and post-thaw life span. Andrologia 2021; 54:e14249. [PMID: 34553400 DOI: 10.1111/and.14249] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/04/2021] [Accepted: 09/10/2021] [Indexed: 11/30/2022] Open
Abstract
The aim of the present study was to evaluate the effects of an alpha-lipoic acid-supplemented extender on ram semen at a post-thaw stage and after incubation (6 hr). Semen samples were collected from five Kivircik Rams. Pooled semen was diluted with an egg yolk-based extender with different concentrations of alpha-lipoic acid (0.25 mmol L-1 , 0.5 mmol L-1 and 1 mmol L-1 ) and without alpha-lipoic acid. Motility, plasma membrane functional integrity (HOST), acrosome integrity (FITC-Pisum sativum agglutinin) and DNA integrity (TUNEL) were assessed at post-thaw and 6 hr after incubation of the frozen-thawed semen. At the post-thaw stage, 0.25 mmol L-1 alpha-lipoic acid had a positive effect on sperm motility and plasma membrane functional integrity compared to the control group (p < .05). At the post-incubation stage (6 hr), it was determined that the motility and plasma membrane functional integrity of the antioxidant groups were adversely affected compared to the control group (p < .05) and 1 mmol L-1 dose of alpha-lipoic acid had a harmful effect on DNA integrity compared to the control group (p < .05). The results of the study demonstrated that alpha-lipoic acid has positive effects at post-thaw but have harmful effects on long term to ram semen.
Collapse
Affiliation(s)
- Nail Tekin Onder
- Department of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine, Kafkas University, Kars, Turkey.,Department of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Selim Alcay
- Department of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine, Kafkas University, Kars, Turkey
| | - Zekariya Nur
- Department of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine, Kafkas University, Kars, Turkey
| |
Collapse
|
21
|
Atractylodes chinensis Water Extract Ameliorates Obesity via Promotion of the SIRT1/AMPK Expression in High-Fat Diet-Induced Obese Mice. Nutrients 2021; 13:nu13092992. [PMID: 34578872 PMCID: PMC8470677 DOI: 10.3390/nu13092992] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity remains a continuing global health concern, as it is associated with an increased risk of many chronic diseases. Atractylodes chinensis Koidz. (Ac) is traditionally used in the treatment of inflammatory diseases, such as arthritis, hepatitis, and gastric ulcers. Despite the diverse pharmacological activities of Ac, scientific evidence for the use of Ac in obesity is still limited. Therefore, the present study aimed to determine the anti-obesity effects of Ac. C57BL/6N mice were divided into five groups as follows: chow diet group (CON), 45% HFD group, HFD + oral administration of orlistat group, and HFD + oral administration of Ac groups. RT-PCR and western blotting were used to examine the expression of molecules relating to obesity progression. Ac-administered mice showed dramatically decreased body weight and weight gain compared to the high-fat diet (HFD)-fed mice. In addition, Ac administration attenuated the protein expression levels of adipogenic transcription factors in the white adipose tissue (WAT) and livers of HFD-fed mice. Furthermore, Ac administration declined the expression levels of lipogenic genes, while enhancing those of the fatty acid oxidation genes in the WAT of HFD-fed mice. Importantly, Ac administration highly upregulated the AMP-activated kinase (AMPK) and sirtuin 1 (SIRT1) expression levels in WAT of the HFD-induced obese mouse model. Our results provide evidence that Ac can effectively ameliorate weight gain and adipose tissue expansion.
Collapse
|
22
|
Zhang P, Wang W, Mao M, Gao R, Shi W, Li D, Calderone R, Sui B, Tian X, Meng X. Similarities and Differences: A Comparative Review of the Molecular Mechanisms and Effectors of NAFLD and AFLD. Front Physiol 2021; 12:710285. [PMID: 34393826 PMCID: PMC8362097 DOI: 10.3389/fphys.2021.710285] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and alcoholic fatty liver disease (AFLD) are the most prevalent metabolic liver diseases globally. Due to the complex pathogenic mechanisms of NAFLD and AFLD, no specific drugs were approved at present. Lipid accumulation, oxidative stress, insulin resistance, inflammation, and dietary habits are all closely related to the pathogenesis of NAFLD and AFLD. However, the mechanism that promotes disease progression has not been fully elucidated. Meanwhile, the gut microbiota and their metabolites also play an important role in the pathogenesis and development of NAFLD and AFLD. This article comparatively reviewed the shared and specific signaling pathways, clinical trials, and potential intervention effectors of NAFLD and AFLD, revealing their similarities and differences. By comparing the shared and specific molecular regulatory mechanisms, this paper provides mutual reference strategies for preventing and treating NAFLD, AFLD, and related metabolic diseases. Furthermore, it provides enlightenment for discovering novel therapies of safe and effective drugs targeting the metabolic liver disease.
Collapse
Affiliation(s)
- Pengyi Zhang
- School of Sports and Health, Shandong Sport University, Jinan, China
| | - Weiya Wang
- School of Sports and Health, Shandong Sport University, Jinan, China.,Shandong Academy of Pharmaceutical Science, Jinan, China
| | - Min Mao
- Department of Allied Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ruolin Gao
- School of Sports and Health, Shandong Sport University, Jinan, China
| | - Wenting Shi
- School of Sports and Health, Shandong Sport University, Jinan, China
| | - Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Richard Calderone
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Bo Sui
- School of Sports and Health, Shandong Sport University, Jinan, China
| | - Xuewen Tian
- School of Sports and Health, Shandong Sport University, Jinan, China
| | - Xiangjing Meng
- Shandong Academy of Pharmaceutical Science, Jinan, China
| |
Collapse
|
23
|
Mohammed MA, Gharib DM, Reyad HR, Mohamed AA, Elroby FA, Mahmoud HS. Antioxidant and anti-inflammatory properties of alpha-lipoic acid protect against valproic acid-induced liver injury. Can J Physiol Pharmacol 2021; 99:499-505. [PMID: 33275538 DOI: 10.1139/cjpp-2019-0456] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Valproic acid (VPA) is one of the most used antiepileptic drugs despite of its many adverse effects such as anemia, leucopenia, thrombocytopenia, and liver toxicity. The hepatoprotective effect of alpha-lipoic acid (ALA) was confirmed. The aim of this study was to detect the protective effect of ALA against the adverse effects of VPA. To study this, 30 white albino Wistar male rats were divided into four groups. Group I was the control group; Group II included rats that received ALA (100 mg·kg-1·day-1) orally for 14 days; Group III and Group IV included rats that received VPA (500 mg·kg-1·day-1) for 15 days intraperitoneally, but Group IV rats received ALA (100 mg·kg-1·day-1) orally for 14 days prior to VPA. Blood samples were collected and livers were excised from rats for colorimetric analysis and quantitative real-time PCR. The rats that received VPA showed leucopenia, thrombocytopenia, a significant decrease of superoxide dismutase, glutathione, nuclear factor erythroid 2-related factor 2, and sirtuin 1, besides a significant increase of malondialdehyde and tumor necrosis factor α. Prior treatment with ALA prevented all these results; ALA protected against VPA-induced liver damage and hematological disturbance via antioxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
| | | | - Hoda Ramadan Reyad
- Department of Biochemistry, Faculty of Medicine, Beni-Suef University, Egypt
| | - Alaa Aboud Mohamed
- Department of Biochemistry, Faculty of Medicine, Beni-Suef University, Egypt
| | - Fadwa A Elroby
- Department of Forensic Medicine and Toxicology, Faculty of Medicine, Beni-Suef University, Egypt
| | - Hoda Sayed Mahmoud
- Department of Forensic Medicine and Toxicology, Faculty of Medicine, Beni-Suef University, Egypt
| |
Collapse
|
24
|
Martínez-Gutiérrez A, Fernández-Duran I, Marazuela-Duque A, Simonet NG, Yousef I, Martínez-Rovira I, Martínez-Hoyos J, Vaquero A. Shikimic acid protects skin cells from UV-induced senescence through activation of the NAD+-dependent deacetylase SIRT1. Aging (Albany NY) 2021; 13:12308-12333. [PMID: 33901008 PMCID: PMC8148468 DOI: 10.18632/aging.203010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/31/2021] [Indexed: 01/10/2023]
Abstract
UV radiation is one of the main contributors to skin photoaging by promoting the accumulation of cellular senescence, which in turn induces a proinflammatory and tissue-degrading state that favors skin aging. The members of the sirtuin family of NAD+-dependent enzymes play an anti-senescence role and their activation suggests a promising approach for preventing UV-induced senescence in the treatment of skin aging. A two-step screening designed to identify compounds able to protect cells from UV-induced senescence through sirtuin activation identified shikimic acid (SA), a metabolic intermediate in many organisms, as a bona-fide candidate. The protective effects of SA against senescence were dependent on specific activation of SIRT1 as the effect was abrogated by the SIRT1 inhibitor EX-527. Upon UV irradiation SA induced S-phase accumulation and a decrease in p16INK4A expression but did not protect against DNA damage or increased polyploidies. In contrast, SA reverted misfolded protein accumulation upon senescence, an effect that was abrogated by EX-527. Consistently, SA induced an increase in the levels of the chaperone BiP, resulting in a downregulation of unfolded protein response (UPR) signaling and UPR-dependent autophagy, avoiding their abnormal hyperactivation during senescence. SA did not directly activate SIRT1 in vitro, suggesting that SIRT1 is a downstream effector of SA signaling specifically in the response to cellular senescence. Our study not only uncovers a shikimic acid/SIRT1 signaling pathway that prevents cellular senescence, but also reinforces the role of sirtuins as key regulators of cell proteostasis.
Collapse
Affiliation(s)
- Alfredo Martínez-Gutiérrez
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Barcelona 08916, Badalona, Spain.,Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona 08908, Spain.,Mesostetic Pharma Group, Barcelona 08840, Viladecans, Spain
| | - Irene Fernández-Duran
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Barcelona 08916, Badalona, Spain
| | - Anna Marazuela-Duque
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Barcelona 08916, Badalona, Spain.,Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona 08908, Spain
| | - Nicolás G Simonet
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Barcelona 08916, Badalona, Spain.,Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona 08908, Spain
| | - Ibraheem Yousef
- MIRAS Beamline, ALBA-CELLS Synchrotron, Barcelona 08290, Cerdanyola del Vallès, Spain
| | - Immaculada Martínez-Rovira
- MIRAS Beamline, ALBA-CELLS Synchrotron, Barcelona 08290, Cerdanyola del Vallès, Spain.,Ionizing Radiation Research Group, Physics Department, Universitat Autònoma de Barcelona (UAB), Barcelona 08193, Cerdanyola del Vallès, Spain
| | | | - Alejandro Vaquero
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Barcelona 08916, Badalona, Spain.,Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona 08908, Spain
| |
Collapse
|
25
|
Elbadawy AM, Abd Elmoniem RO, Elsayed AM. Alpha lipoic acid and diabetes mellitus: potential effects on peripheral neuropathy and different metabolic parameters. ALEXANDRIA JOURNAL OF MEDICINE 2021. [DOI: 10.1080/20905068.2021.1907961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
26
|
Roberts JD, Willmott AGB, Beasley L, Boal M, Davies R, Martin L, Chichger H, Gautam L, Del Coso J. The Impact of Decaffeinated Green Tea Extract on Fat Oxidation, Body Composition and Cardio-Metabolic Health in Overweight, Recreationally Active Individuals. Nutrients 2021; 13:nu13030764. [PMID: 33652910 PMCID: PMC7996723 DOI: 10.3390/nu13030764] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/14/2021] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
This study investigated the effect of decaffeinated green tea extract (dGTE), with or without antioxidant nutrients, on fat oxidation, body composition and cardio-metabolic health measures in overweight individuals engaged in regular exercise. Twenty-seven participants (20 females, 7 males; body mass: 77.5 ± 10.5 kg; body mass index: 27.4 ± 3.0 kg·m2; peak oxygen uptake (V.O2peak): 30.2 ± 5.8 mL·kg−1·min−1) were randomly assigned, in a double-blinded manner, either: dGTE (400 mg·d−1 (−)-epigallocatechin−3-gallate (EGCG), n = 9); a novel dGTE+ (400 mg·d−1 EGCG, quercetin (50 mg·d−1) and α-lipoic acid (LA, 150 mg·d−1), n = 9); or placebo (PL, n = 9) for 8 weeks, whilst maintaining standardised, aerobic exercise. Fat oxidation (‘FATMAX’ and steady state exercise protocols), body composition, cardio-metabolic and blood measures (serum glucose, insulin, leptin, adiponectin, glycerol, free fatty acids, total cholesterol, high [HDL-c] and low-density lipoprotein cholesterol [LDL-c], triglycerides, liver enzymes and bilirubin) were assessed at baseline, week 4 and 8. Following 8 weeks of dGTE+, maximal fat oxidation (MFO) significantly improved from 154.4 ± 20.6 to 224.6 ± 23.2 mg·min−1 (p = 0.009), along with a 22.5% increase in the exercise intensity at which fat oxidation was deemed negligible (FATMIN; 67.6 ± 3.6% V.O2peak, p = 0.003). Steady state exercise substrate utilisation also improved for dGTE+ only, with respiratory exchange ratio reducing from 0.94 ± 0.01 at week 4, to 0.89 ± 0.01 at week 8 (p = 0.004). This corresponded with a significant increase in the contribution of fat to energy expenditure for dGTE+ from 21.0 ± 4.1% at week 4, to 34.6 ± 4.7% at week 8 (p = 0.006). LDL-c was also lower (normalised fold change of −0.09 ± 0.06) for dGTE+ by week 8 (p = 0.038). No other significant effects were found in any group. Eight weeks of dGTE+ improved MFO and substrate utilisation during exercise, and lowered LDL-c. However, body composition and cardio-metabolic markers in healthy, overweight individuals who maintained regular physical activity were largely unaffected by dGTE.
Collapse
Affiliation(s)
- Justin D. Roberts
- Cambridge Centre for Sport and Exercise Sciences, School of Psychology and Sport Science, Anglia Ruskin University, Cambridge CB1 1PT, UK; (A.G.B.W.); (M.B.); (R.D.); (L.M.)
- Correspondence: ; Tel.: +44-845-196-5154
| | - Ashley G. B. Willmott
- Cambridge Centre for Sport and Exercise Sciences, School of Psychology and Sport Science, Anglia Ruskin University, Cambridge CB1 1PT, UK; (A.G.B.W.); (M.B.); (R.D.); (L.M.)
| | - Liam Beasley
- Department for Health, University of Bath, Bath BA2 7AY, UK;
| | - Mariette Boal
- Cambridge Centre for Sport and Exercise Sciences, School of Psychology and Sport Science, Anglia Ruskin University, Cambridge CB1 1PT, UK; (A.G.B.W.); (M.B.); (R.D.); (L.M.)
| | - Rory Davies
- Cambridge Centre for Sport and Exercise Sciences, School of Psychology and Sport Science, Anglia Ruskin University, Cambridge CB1 1PT, UK; (A.G.B.W.); (M.B.); (R.D.); (L.M.)
| | - Laurence Martin
- Cambridge Centre for Sport and Exercise Sciences, School of Psychology and Sport Science, Anglia Ruskin University, Cambridge CB1 1PT, UK; (A.G.B.W.); (M.B.); (R.D.); (L.M.)
| | - Havovi Chichger
- School of Life Sciences, Anglia Ruskin University, Cambridge CB1 1PT, UK; (H.C.); (L.G.)
| | - Lata Gautam
- School of Life Sciences, Anglia Ruskin University, Cambridge CB1 1PT, UK; (H.C.); (L.G.)
| | - Juan Del Coso
- Centre for Sport Studies, Rey Juan Carlos University, 28943, Fuenlabrada, Spain;
| |
Collapse
|
27
|
Ben Y, Hao J, Zhang Z, Xiong Y, Zhang C, Chang Y, Yang F, Li H, Zhang T, Wang X, Xu Q. Astragaloside IV Inhibits Mitochondrial-Dependent Apoptosis of the Dorsal Root Ganglion in Diabetic Peripheral Neuropathy Rats Through Modulation of the SIRT1/p53 Signaling Pathway. Diabetes Metab Syndr Obes 2021; 14:1647-1661. [PMID: 33883914 PMCID: PMC8055373 DOI: 10.2147/dmso.s301068] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/12/2021] [Indexed: 12/21/2022] Open
Abstract
PURPOSE To investigate the effect of astragaloside IV (AS-IV) on mitochondrial-dependent apoptosis in the dorsal root ganglion of diabetic peripheral neuropathy (DPN) rats through the SIRT1/p53 pathway. METHODS Diabetic rat model was induced by high-carbohydrate/high-fat diet and intraperitoneal injection of STZ. Diabetic rats were divided into three groups (n =16 per group): DPN group, AS-IV group (60mg/kg/d) and α-lipoic acid (ALA) group (60mg/kg/d). Weight and blood glucose levels were monitored every 4 weeks for 12 weeks. DPN was evaluated using the Von Frey Filaments Test and nerve conduction velocity. The dorsal root ganglia of rats were isolated and the pathological changes of mitochondria were observed by electron microscopy. The activity of mitochondrial electron transport chain complex, mitochondrial membrane potential, malonaldehyde (MDA) and glutathione (GSH) levels were measured. Neural apoptosis was detected using the Terminal Deoxynucleotidyl Nick-End Labeling (TUNEL) assay kit. The cleaved caspase-3, major proteins in the SIRT1/p53 pathway, including SIRT1, acetyl p53, Drp1, BAX, and BCL-2, were detected using immunohistochemistry and Western blot. Gene expression of major proteins in the SIRT1/p53 pathway was also detected. RESULTS After 12 weeks of treatment, AS-IV and ALA did not significantly affect body weight or fasting glucose levels, but reduced mechanical abnormal pain in DPN and improved nerve conduction velocity. AS-IV and ALA increased the level of GSH and decreased the level of MDA. Both AS-IV and ALA can reduce mitochondrial damage, improve mitochondrial electron transport chain complex activity and mitochondrial membrane potential, and reduce the percentages of positive cells with DNA fragmentation and the expression of cleaved caspase-3 protein. AS-IV and ALA up-regulated the expression of SIRT1 and down-regulated the expression of acetyl-p53, Drp1 and the ratio of BAX to BCL-2. Changes in gene expression were similar. CONCLUSION AS-IV can reduce the occurrence of mitochondrial-dependent apoptosis by regulating the SIRT1/p53 pathway. It has a similar therapeutic effect as ALA and is therefore a promising drug for the potential treatment of DPN.
Collapse
Affiliation(s)
- Ying Ben
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
| | - Juan Hao
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
| | - Zhihong Zhang
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
| | - Yunzhao Xiong
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
| | - Cuijuan Zhang
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
| | - Yi Chang
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
| | - Fan Yang
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
| | - Hui Li
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
| | - Tianya Zhang
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
| | - Xiangting Wang
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
| | - Qingyou Xu
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
- Correspondence: Qingyou Xu Hebei University of Chinese Medicine, No. 326 Xinshinan Road, Qiaoxi District, Shijiazhuang, Hebei Province, 050090, People’s Republic of ChinaTel +86 13832368865Fax +86 311 89926000 Email
| |
Collapse
|
28
|
Diane A, Mahmoud N, Bensmail I, Khattab N, Abunada HA, Dehbi M. Alpha lipoic acid attenuates ER stress and improves glucose uptake through DNAJB3 cochaperone. Sci Rep 2020; 10:20482. [PMID: 33235302 PMCID: PMC7687893 DOI: 10.1038/s41598-020-77621-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 10/26/2020] [Indexed: 12/21/2022] Open
Abstract
Persistent ER stress, mitochondrial dysfunction and failure of the heat shock response (HSR) are fundamental hallmarks of insulin resistance (IR); one of the early core metabolic aberrations that leads to type 2 diabetes (T2D). The antioxidant α-lipoic acid (ALA) has been shown to attenuate metabolic stress and improve insulin sensitivity in part through activation of the heat shock response (HSR). However, these studies have been focused on a subset of heat shock proteins (HSPs). In the current investigation, we assessed whether ALA has an effect on modulating the expression of DNAJB3/HSP40 cochaperone; a potential therapeutic target with a novel role in mitigating metabolic stress and promoting insulin signaling. Treatment of C2C12 cells with 0.3 mM of ALA triggers a significant increase in the expression of DNAJB3 mRNA and protein. A similar increase in DNAJB3 mRNA was also observed in HepG2 cells. We next investigated the significance of such activation on endoplasmic reticulum (ER) stress and glucose uptake. ALA pre-treatment significantly reduced the expression of ER stress markers namely, GRP78, XBP1, sXBP1 and ATF4 in response to tunicamycin. In functional assays, ALA treatment abrogated significantly the tunicamycin-mediated transcriptional activation of ATF6 while it enhanced the insulin-stimulated glucose uptake and Glut4 translocation. Silencing the expression of DNAJB3 but not HSP72 abolished the protective effect of ALA on tunicamycin-induced ER stress, suggesting thus that DNAJB3 is a key mediator of ALA-alleviated tunicamycin-induced ER stress. Furthermore, the effect of ALA on insulin-stimulated glucose uptake is significantly reduced in C2C12 and HepG2 cells transfected with DNAJB3 siRNA. In summary, our results are supportive of an essential role of DNAJB3 as a molecular target through which ALA alleviates ER stress and improves glucose uptake.
Collapse
Affiliation(s)
- Abdoulaye Diane
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Naela Mahmoud
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.,College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Ilham Bensmail
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Namat Khattab
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Hanan A Abunada
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Mohammed Dehbi
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar. .,College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.
| |
Collapse
|
29
|
The Alpha-Lipoic Acid Improves Survival and Prevents Irinotecan-Induced Inflammation and Intestinal Dysmotility in Mice. Pharmaceuticals (Basel) 2020; 13:ph13110361. [PMID: 33152996 PMCID: PMC7692584 DOI: 10.3390/ph13110361] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 01/07/2023] Open
Abstract
Irinotecan, an anticancer drug, induces diarrhea and intestinal inflammation, resulting in an increase in the cost of care and in treatment delays. In this study, we investigated whether alpha-lipoic acid (α-LA) could improve irinotecan-mediated intestinal inflammation, diarrhea and dysmotility. Intestinal mucositis was induced by irinotecan injection (75 mg/kg, i.p., for 4 days) in Swiss mice. α-LA (50, 100 or 200 mg/kg, gavage) was administered daily 1 h before the injection of irinotecan. Duodenum tissues were obtained for inflammation and proliferation analysis. The outcomes: diarrhea, intestinal dysmotility, weight body loss and survival were evaluated. Compared with the control condition, irinotecan diminished (p < 0.05) intestinal villus height, caused a loss of crypt integrity and intense inflammatory cell infiltration, increased myeloperoxidase (MPO), IL-6 and IL-1β levels and decreased reduced glutathione (GSH) levels in duodenum segments and increased gastric retention and decreased liquid retention in the medial intestinal segment, resulting in increased intestinal transit, severe diarrhea and reduced survival (approximately 72%). Furthermore, α-LA (200 mg/kg) pretreatment ameliorated (p < 0.05) these irinotecan-induced effects. Our findings show that α-LA reduced irinotecan-induced inflammation, intestinal dysmotility and diarrhea, resulting in improved survival. α-LA may be a useful therapeutic agent for the treatment of gut dysmotility in patients with intestinal mucositis associated with irinotecan treatment.
Collapse
|
30
|
Cai J, Chen J, Zeng Q, Liu J, Zhang Y, Cheng H, Yao S, Chen Q. Assessment of the efficacy of α-lipoic acid in treatment of diabetes mellitus patients with erectile dysfunction: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e22161. [PMID: 32899103 PMCID: PMC7478782 DOI: 10.1097/md.0000000000022161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Diabetes mellitus with erectile dysfunction (DMED) is one of the most common causes of disability in diabetic population, and its pathogenesis is related to a variety of factors. Because its pathogenesis is complex and the existing treatment methods have limitations, DMED is difficult to treat in clinical. Recently, some studies have shown that α-lipoic acid (ALA) is associated with DMED, but there is no systematic review and meta-analysis on the relationship between ALA and DMED. METHODS We will search each database from the built-in until July 2020. The English literature mainly searches Cochrane Library, PubMed, EMBASE, and Web of Science, while the Chinese literature comes from CNKI, CBM, VIP, and Wangfang database. Simultaneously we will retrieve clinical registration tests and grey literatures. This study only screen the clinical randomized controlled trials (RCTs) about ALA for DMED to assess its efficacy. The 2 researchers worked independently on literature selection, data extraction, and quality assessment. The dichotomous data is represented by relative risk (RR), and the continuous is expressed by mean difference (MD) or standard mean difference (SMD), eventually the data is synthesized using a fixed effect model (FEM) or a random effect model (REM) depending on whether or not heterogeneity exists. Erectile dysfunction (ED) will be diagnosed by the International Index of Erectile Function 5 (IIEF-5) score. Finally, meta-analysis was conducted by RevMan software version 5.3. RESULTS This study will synthesize and provide high quality to evaluate the effectiveness of ALA supplementation for the treatment of DMED. CONCLUSION This systematic review aims to provide new options for ALA supplementation treatment of DMED in terms of its efficacy and safety. PROSPERO REGISTRATION NUMBER INPLASY202070130.
Collapse
|
31
|
He B, Moreau R. R-α-Lipoic Acid and 4-Phenylbutyric Acid Have Distinct Hypolipidemic Mechanisms in Hepatic Cells. Biomedicines 2020; 8:biomedicines8080289. [PMID: 32824248 PMCID: PMC7460023 DOI: 10.3390/biomedicines8080289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 11/16/2022] Open
Abstract
The constitutive activation of the mechanistic target of rapamycin complex 1 (mTORC1) leads to the overproduction of apoB-containing triacylglycerol-rich lipoproteins in HepG2 cells. R-α-lipoic acid (LA) and 4-phenylbutyric acid (PBA) have hypolipidemic function but their mechanisms of action are not well understood. Here, we reported that LA and PBA regulate hepatocellular lipid metabolism via distinct mechanisms. The use of SQ22536, an inhibitor of adenylyl cyclase, revealed cAMP’s involvement in the upregulation of CPT1A expression by LA but not by PBA. LA decreased the secretion of proprotein convertase subtilisin/kexin type 9 (PCSK9) in the culture media of hepatic cells and increased the abundance of LDL receptor (LDLR) in cellular extracts in part through transcriptional upregulation. Although PBA induced LDLR gene expression, it did not translate into more LDLR proteins. PBA regulated cellular lipid homeostasis through the induction of CPT1A and INSIG2 expression via an epigenetic mechanism involving the acetylation of histone H3, histone H4, and CBP-p300 at the CPT1A and INSIG2 promoters.
Collapse
|
32
|
Tai S, Zheng Q, Zhai S, Cai T, Xu L, Yang L, Jiao L, Zhang C. Alpha-Lipoic Acid Mediates Clearance of Iron Accumulation by Regulating Iron Metabolism in a Parkinson's Disease Model Induced by 6-OHDA. Front Neurosci 2020; 14:612. [PMID: 32670009 PMCID: PMC7330090 DOI: 10.3389/fnins.2020.00612] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 05/18/2020] [Indexed: 01/18/2023] Open
Abstract
The disruption of neuronal iron homeostasis and oxidative stress are related to the pathogenesis of Parkinson's disease (PD). Alpha-lipoic acid (ALA) is a naturally occurring enzyme cofactor with antioxidant and iron chelator properties and has many known effects. ALA has neuroprotective effects on PD. However, its underlying mechanism remains unclear. In the present study, we established PD models induced by 6-hydroxydopamine (6-OHDA) to explore the neuroprotective ability of ALA and its underlying mechanism in vivo and in vitro. Our results showed that ALA could provide significant protection from 6-OHDA-induced cell damage in vitro by decreasing the levels of intracellular reactive oxygen species and iron. ALA significantly promoted the survival of the dopaminergic neuron in the 6-OHDA-induced PD rat model and remarkably ameliorated motor deficits by dramatically inhibiting the decrease in tyrosine hydroxylase expression and superoxide dismutase activity in the substantia nigra. Interestingly, ALA attenuated 6-OHDA-induced iron accumulation both in vivo and in vitro by antagonizing the 6-OHDA-induced upregulation of iron regulatory protein 2 and divalent metal transporter 1. These results indicated that the neuroprotective mechanism of ALA against neurological injury induced by 6-OHDA may be related to the regulation of iron homeostasis and reduced oxidative stress levels. Therefore, ALA may provide neuroprotective therapy for PD and other diseases related to iron metabolism disorder.
Collapse
Affiliation(s)
- Shengyan Tai
- Department of Biology, College of Basic Medical, Guizhou Medical University, Guiyang, China
| | - Qian Zheng
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Suzhen Zhai
- Department of Biology, College of Basic Medical, Guizhou Medical University, Guiyang, China
| | - Ting Cai
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Li Xu
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lizhu Yang
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ling Jiao
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chunlin Zhang
- Department of Biology, College of Basic Medical, Guizhou Medical University, Guiyang, China
| |
Collapse
|
33
|
Huang H, Wang ZJ, Zhang HB, Liang JX, Cao WD, Wu Q, He CP, Chen C. The Function of PPARγ/AMPK/SIRT-1 Pathway in Inflammatory Response of Human Articular Chondrocytes Stimulated by Advanced Glycation End Products. Biol Pharm Bull 2020; 42:1303-1309. [PMID: 31366866 DOI: 10.1248/bpb.b19-00036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Accumulation of advanced glycation end products (AGEs) in the articular cartilage is a major risk factor for osteoarthritis (OA). To determine the mechanistic basis of AGE action in OA, we treated human articular chondrocytes with AGEs, and found that they not only up-regulated the pro-inflammatory cytokines interleukin (IL)-1β and tumor necrosis factor (TNF)-α, but also inhibited AMP-activated protein kinase (AMPK) phosphorylation and decreased sirtuin 1 (SIRT-1) levels in a concentration- and time-dependent manner. Pioglitazone, a peroxisome proliferator-activated receptor-γ (PPARγ) agonist restored the inhibited AMPK and SIRT-1 by AGEs. Pre-treatment of the cells with the agonists or antagonists of AMPK and SIRT-1 respectively abolished and augmented the inflammatory state induced by AGEs. Furthermore, AMPK agonist also restored the levels of SIRT-1 in the AGE-stimulated chondrocytes. Our findings indicate AGEs induce an inflammatory response in human articular chondrocytes via the PPARγ/AMPK/SIRT-1 pathway, which is therefore a potential target in OA therapy.
Collapse
Affiliation(s)
- Hao Huang
- Department of Orthopedics, The 921st Hospital of the People's Liberation Army, The Second Affiliated Hospital of Hunan Normal University
| | - Zhao-Jun Wang
- Department of Orthopedics, The 921st Hospital of the People's Liberation Army, The Second Affiliated Hospital of Hunan Normal University
| | - Hai-Bin Zhang
- Department of Orthopedics, The 921st Hospital of the People's Liberation Army, The Second Affiliated Hospital of Hunan Normal University
| | - Jian-Xia Liang
- Department of Orthopedics, The 921st Hospital of the People's Liberation Army, The Second Affiliated Hospital of Hunan Normal University
| | - Wen-Dong Cao
- Department of Orthopedics, The 921st Hospital of the People's Liberation Army, The Second Affiliated Hospital of Hunan Normal University
| | - Qi Wu
- Department of Orthopedics, The 921st Hospital of the People's Liberation Army, The Second Affiliated Hospital of Hunan Normal University
| | - Chao-Peng He
- Department of Orthopedics, The 921st Hospital of the People's Liberation Army, The Second Affiliated Hospital of Hunan Normal University
| | - Cheng Chen
- Department of Orthopedics, The 921st Hospital of the People's Liberation Army, The Second Affiliated Hospital of Hunan Normal University
| |
Collapse
|
34
|
Johnson AA, Stolzing A. The role of lipid metabolism in aging, lifespan regulation, and age-related disease. Aging Cell 2019; 18:e13048. [PMID: 31560163 PMCID: PMC6826135 DOI: 10.1111/acel.13048] [Citation(s) in RCA: 237] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/11/2019] [Accepted: 09/04/2019] [Indexed: 12/18/2022] Open
Abstract
An emerging body of data suggests that lipid metabolism has an important role to play in the aging process. Indeed, a plethora of dietary, pharmacological, genetic, and surgical lipid‐related interventions extend lifespan in nematodes, fruit flies, mice, and rats. For example, the impairment of genes involved in ceramide and sphingolipid synthesis extends lifespan in both worms and flies. The overexpression of fatty acid amide hydrolase or lysosomal lipase prolongs life in Caenorhabditis elegans, while the overexpression of diacylglycerol lipase enhances longevity in both C. elegans and Drosophila melanogaster. The surgical removal of adipose tissue extends lifespan in rats, and increased expression of apolipoprotein D enhances survival in both flies and mice. Mouse lifespan can be additionally extended by the genetic deletion of diacylglycerol acyltransferase 1, treatment with the steroid 17‐α‐estradiol, or a ketogenic diet. Moreover, deletion of the phospholipase A2 receptor improves various healthspan parameters in a progeria mouse model. Genome‐wide association studies have found several lipid‐related variants to be associated with human aging. For example, the epsilon 2 and epsilon 4 alleles of apolipoprotein E are associated with extreme longevity and late‐onset neurodegenerative disease, respectively. In humans, blood triglyceride levels tend to increase, while blood lysophosphatidylcholine levels tend to decrease with age. Specific sphingolipid and phospholipid blood profiles have also been shown to change with age and are associated with exceptional human longevity. These data suggest that lipid‐related interventions may improve human healthspan and that blood lipids likely represent a rich source of human aging biomarkers.
Collapse
|
35
|
Salehi B, Berkay Yılmaz Y, Antika G, Boyunegmez Tumer T, Fawzi Mahomoodally M, Lobine D, Akram M, Riaz M, Capanoglu E, Sharopov F, Martins N, Cho WC, Sharifi-Rad J. Insights on the Use of α-Lipoic Acid for Therapeutic Purposes. Biomolecules 2019; 9:biom9080356. [PMID: 31405030 PMCID: PMC6723188 DOI: 10.3390/biom9080356] [Citation(s) in RCA: 192] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/26/2019] [Accepted: 07/26/2019] [Indexed: 12/22/2022] Open
Abstract
α-lipoic acid (ALA, thioctic acid) is an organosulfur component produced from plants, animals, and humans. It has various properties, among them great antioxidant potential and is widely used as a racemic drug for diabetic polyneuropathy-associated pain and paresthesia. Naturally, ALA is located in mitochondria, where it is used as a cofactor for pyruvate dehydrogenase (PDH) and α-ketoglutarate dehydrogenase complexes. Despite its various potentials, ALA therapeutic efficacy is relatively low due to its pharmacokinetic profile. Data suggests that ALA has a short half-life and bioavailability (about 30%) triggered by its hepatic degradation, reduced solubility as well as instability in the stomach. However, the use of various innovative formulations has greatly improved ALA bioavailability. The R enantiomer of ALA shows better pharmacokinetic parameters, including increased bioavailability as compared to its S enantiomer. Indeed, the use of amphiphilic matrices has capability to improve ALA bioavailability and intestinal absorption. Also, ALA's liquid formulations are associated with greater plasma concentration and bioavailability as compared to its solidified dosage form. Thus, improved formulations can increase both ALA absorption and bioavailability, leading to a raise in therapeutic efficacy. Interestingly, ALA bioavailability will be dependent on age, while no difference has been found for gender. The present review aims to provide an updated on studies from preclinical to clinical trials assessing ALA's usages in diabetic patients with neuropathy, obesity, central nervous system-related diseases and abnormalities in pregnancy.
Collapse
Affiliation(s)
- Bahare Salehi
- Student Research Committee, School of Medicine, Bam University of Medical Sciences, Bam 44340847, Iran
| | - Yakup Berkay Yılmaz
- Graduate Program of Biomolecular Sciences, Institute of Natural and Applied Sciences, Canakkale Onsekiz Mart University, Canakkale 17020, Turkey
| | - Gizem Antika
- Graduate Program of Biomolecular Sciences, Institute of Natural and Applied Sciences, Canakkale Onsekiz Mart University, Canakkale 17020, Turkey
| | - Tugba Boyunegmez Tumer
- Department of Molecular Biology and Genetics, Faculty of Arts and Science, Canakkale Onsekiz Mart University, Canakkale 17020, Turkey
| | | | - Devina Lobine
- Department of Health Sciences; Faculty of Science, University of Mauritius, Réduit 80837, Mauritius
| | - Muhammad Akram
- Department of Eastern Medicine, Government College University Faisalabad; Faisalabad 38000, Pakistan
| | - Muhammad Riaz
- Department of Allied Health Sciences, Sargodha Medical College, University of Sargodha, Sargodha 40100, Pakistan
| | - Esra Capanoglu
- Faculty of Chemical & Metallurgical Engineering, Food Engineering Department, Istanbul Technical University, Maslak 34469, Turkey
| | - Farukh Sharopov
- Department of Pharmaceutical Technology, Avicenna Tajik State Medical University, Rudaki 139, Dushanbe 734003, Tajikistan.
| | - Natália Martins
- Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Hong Kong.
| | - Javad Sharifi-Rad
- Zabol Medicinal Plants Research Center, Zabol University of Medical Sciences, Zabol 61615-585, Iran.
| |
Collapse
|
36
|
Dziewulska A, Dobosz AM, Dobrzyn A, Smolinska A, Kolczynska K, Ntambi JM, Dobrzyn P. SCD1 regulates the AMPK/SIRT1 pathway and histone acetylation through changes in adenine nucleotide metabolism in skeletal muscle. J Cell Physiol 2019; 235:1129-1140. [PMID: 31241768 DOI: 10.1002/jcp.29026] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/11/2019] [Indexed: 01/06/2023]
Abstract
Stearoyl-CoA desaturase (SCD) is a rate-limiting enzyme that catalyzes the synthesis of monounsaturated fatty acids. It plays an important role in regulating skeletal muscle metabolism. Lack of the SCD1 gene increases the rate of fatty acid β-oxidation through activation of the AMP-activated protein kinase (AMPK) pathway and the upregulation of genes that are related to fatty acid oxidation. The mechanism of AMPK activation under conditions of SCD1 deficiency has been unclear. In the present study, we found that the ablation/inhibition of SCD1 led to AMPK activation in skeletal muscle through an increase in AMP levels whereas muscle-specific SCD1 overexpression decreased both AMPK phosphorylation and the adenosine monophosphate/adenosine triphosphate (AMP/ATP) ratio. Changes in AMPK phosphorylation that were caused by SCD1 down- and upregulation affected NAD+ levels following changes in NAD+ -dependent deacetylase sirtuin-1 (SIRT1) activity and histone 3 (H3K9) acetylation and methylation status. Moreover, mice with muscle-targeted overexpression of SCD1 were more susceptible to high-fat diet-induced lipid accumulation and the development of insulin resistance compared with wild-type mice. These data show that SCD1 is involved in nucleotide (ATP and NAD+ ) metabolism and suggest that the SCD1-dependent regulation of muscle steatosis and insulin sensitivity are mediated by cooperation between AMPK- and SIRT1-regulated pathways. Altogether, the present study reveals a novel mechanism that links SCD1 with the maintenance of metabolic homeostasis and insulin sensitivity in skeletal muscle.
Collapse
Affiliation(s)
- Anna Dziewulska
- Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Aneta M Dobosz
- Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Dobrzyn
- Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Smolinska
- Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Kolczynska
- Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - James M Ntambi
- Departments of Biochemistry and Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Pawel Dobrzyn
- Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
37
|
Camptothecin activates SIRT1 to promote lipid catabolism through AMPK/FoxO1/ATGL pathway in C2C12 myogenic cells. Arch Pharm Res 2019; 42:672-683. [DOI: 10.1007/s12272-019-01155-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 04/15/2019] [Indexed: 01/02/2023]
|
38
|
Mohammed MA, Mahmoud MO, Awaad AS, Gamal GM, Abdelfatah D. Alpha lipoic acid protects against dexamethasone-induced metabolic abnormalities via APPL1 and PGC-1 α up regulation. Steroids 2019; 144:1-7. [PMID: 30684496 DOI: 10.1016/j.steroids.2019.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/08/2019] [Accepted: 01/17/2019] [Indexed: 11/24/2022]
Abstract
BACKGROUND Glucocorticoids (GCs) have various uses in the medicine in different specialties. However, GCs administration is usually accompanying with multiple side effects such as hyperglycemia and hyperlipidemia. Alpha lipoic acid (ALA) has been documented to posse anti-diabetic properties. AIM OF THE STUDY this study highlights the role of ALA in avoiding dexamethasone induced metabolic disturbance. MATERIALS & METHODS 30 rats were randomly divided into 5 groups: Group (1): Control group; Groups 3, 4, and 5: rats received dexamethasone 1 mg/kg/day for 10 days; Groups 2, 4, and 5: Rats received ALA 100 mg/kg/day all the duration of the study, 2 weeks before dexamethasone, or concomitant with dexamethasone respectively. For each rat, we collected blood samples for measurement of glucose, lipid profiles, adiponectin, irisin, and Phosphoinositide 3-kinase (PI3K). We also isolated gastrocnemius muscles for measurement of insulin receptor substrate-1(IRS-1), peroxisome proliferator-activated receptor γ coactivator 1 α(PGC1-α), and adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1(APPL) gene expression. RESULTS Dexamethasone administration caused hyperglycemia, hyperlipemia, decrease the level of adiponectin, irisin, and PI3K besides decreasing the gene expression of IRS-1, PGC-1 α, and APPL1. ALA administration pre or concomitant to dexamethasone avoided these results. CONCLUSION ALA can prevent metabolic abnormalities induced by dexamethasone via PGC1α and APPL1 upregulation.
Collapse
Affiliation(s)
| | - Mohamed O Mahmoud
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Egypt
| | - Ashraf Sayed Awaad
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Beni-Suef University, Egypt
| | | | - Dina Abdelfatah
- Department of Biochemistry, Faculty of Medicine, Cairo University, Egypt
| |
Collapse
|
39
|
The Immunomodulatory Effect of Alpha-Lipoic Acid in Autoimmune Diseases. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8086257. [PMID: 31016198 PMCID: PMC6446120 DOI: 10.1155/2019/8086257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 02/11/2019] [Accepted: 02/21/2019] [Indexed: 01/08/2023]
Abstract
Αlpha-lipoic acid is a naturally occurring antioxidant in human body and has been widely used as an antioxidant clinically. Accumulating evidences suggested that α-lipoic acid might have immunomodulatory effects on both adaptive and innate immune systems. This review focuses on the evidences and potential targets involved in the immunomodulatory effects of α-lipoic acid. It highlights the fact that α-lipoic acid may have beneficial effects in autoimmune diseases once the immunomodulatory effects can be confirmed by further investigation.
Collapse
|
40
|
Haghighatdoost F, Hariri M. Does alpha-lipoic acid affect lipid profile? A meta-analysis and systematic review on randomized controlled trials. Eur J Pharmacol 2019; 847:1-10. [DOI: 10.1016/j.ejphar.2019.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/22/2018] [Accepted: 01/07/2019] [Indexed: 12/29/2022]
|
41
|
Lu DL, Limbu SM, Lv HB, Ma Q, Chen LQ, Zhang ML, Du ZY. The comparisons in protective mechanisms and efficiencies among dietary α-lipoic acid, β-glucan and l-carnitine on Nile tilapia infected by Aeromonas hydrophila. FISH & SHELLFISH IMMUNOLOGY 2019; 86:785-793. [PMID: 30553889 DOI: 10.1016/j.fsi.2018.12.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/05/2018] [Accepted: 12/12/2018] [Indexed: 06/09/2023]
Abstract
Dietary α-lipoic acid (LA), β-glucan (Gluc) and l-carnitine (L-Ca) are commonly used additives to promote fish growth and stress resistance in aquaculture production. However their mechanisms and efficiencies in helping fish to resist diseases have not been compared before. In this study, we fed Nile tilapia (Oreochromis niloticus) with diets containing appropriate doses of LA, Gluc and L-Ca for five weeks and further intraperitoneally injected the fish with Aeromonas hydrophila. After dietary treatment, none of the additives affected the fish growth, but dietary Gluc and L-Ca reduced protein and lipid body contents in fish, respectively. After A. hydrophila challenge, all fish treated with the three dietary additives showed higher survival rate, but those fed on dietary L-Ca had lower survival than those fed on LA and Gluc diets, indicating high protection efficiency of LA and Gluc. The protective mechanisms of the three feed additives were quite different under A. hydrophila infection. Dietary LA induced higher total antioxidant capacity and higher mRNA expression of anti-oxidative genes than other additives in liver and also activated partly the immune function in serum and spleen. Gluc largely increased the immune function by activating the immunity enzymes in serum, inducing inflammation in liver and increasing the expression of immune genes in spleen and head kidney. Gluc also increased partly the antioxidant capacity in serum and liver and lipid catabolism in liver. L-Ca largely increased lipid catabolism in liver while it increased partly the antioxidant capacities in serum and liver. Taken together, these results indicate that, dietary LA, Gluc and L-Ca have various protective mechanisms and differ in their efficiencies on resisting A. hydrophila infection in Nile tilapia.
Collapse
Affiliation(s)
- Dong-Liang Lu
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, PR China
| | - Samwel Mchele Limbu
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, PR China; Department of Aquatic Sciences and Fisheries Technology, University of Dar es Salaam, Dar es Salaam, Tanzania
| | - Hong-Bo Lv
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, PR China
| | - Qiang Ma
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, PR China
| | - Li-Qiao Chen
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, PR China
| | - Mei-Ling Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, PR China
| | - Zhen-Yu Du
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, PR China.
| |
Collapse
|
42
|
Valdecantos MP, Pérez-Matute P, Prieto-Hontoria P, Moreno-Aliaga MJ, Martínez JA. Impact of dietary lipoic acid supplementation on liver mitochondrial bioenergetics and oxidative status on normally fed Wistar rats. Int J Food Sci Nutr 2019; 70:834-844. [PMID: 30764676 DOI: 10.1080/09637486.2019.1572716] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aim of this study was to examine the effects of α-lipoic acid (α-LA) on liver mitochondrial bioenergetics and oxidative status for 8 weeks in normal-healthy animals. A pair-fed group was included to differentiate between α-LA direct effects and those changes due to reduced food intake. α-LA decreased body weight gain, liver weight and insulin levels with no differences compared to its pair-fed group. α-LA significantly reduced energy efficiency, the activity of the electron transport chain complexes and induced a lower efficiency of oxidative phosphorylation with reduced ATP production. α-LA supplementation directly decreased plasma triglycerides (TGs), free fatty acids and ketone bodies levels. A significant reduction in hepatic TG content was also observed. A significant up-regulation of Cpt1a, Acadl and Sirt3, all β-oxidation genes, along with a significant deacetylation of the forkhead transcription factor 3a (FOXO3A) was found in α-LA-treated animals. Thus, α-LA along with a standard chow diet has direct actions on lipid metabolism and liver by modulating mitochondrial function in normal-weight rats. These results should be taken into account when α-LA is administered or recommended to a healthy population.
Collapse
Affiliation(s)
- M P Valdecantos
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM) , Madrid , Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III , Madrid , Spain.,Department of Nutrition, Food Science and Physiology, University of Navarra , Pamplona , Spain
| | - P Pérez-Matute
- Department of Nutrition, Food Science and Physiology, University of Navarra , Pamplona , Spain.,Department of Infectious Diseases, Infectious Diseases, Microbiota and Metabolism Unit, Center for Biomedical Research of La Rioja (CIBIR) , Logroño, Spain
| | - P Prieto-Hontoria
- Department of Nutrition, Food Science and Physiology, University of Navarra , Pamplona , Spain
| | - M J Moreno-Aliaga
- Department of Nutrition, Food Science and Physiology, University of Navarra , Pamplona , Spain.,Centre for Nutrition Research, University of Navarra , Pamplona , Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn) , Madrid , Spain.,IdiSNA, Navarra's Health Research Institute , Pamplona , Spain
| | - J A Martínez
- Department of Nutrition, Food Science and Physiology, University of Navarra , Pamplona , Spain.,Centre for Nutrition Research, University of Navarra , Pamplona , Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn) , Madrid , Spain.,IdiSNA, Navarra's Health Research Institute , Pamplona , Spain.,IMDEA Food Institute, CEI UAM + CSIC , Madrid , Spain
| |
Collapse
|
43
|
Lo MC, Chen MH, Hsueh YT, Kuo YT, Lee HM. Alpha-lipoic acid suppresses N ε-(carboxymethyl) lysine-induced epithelial mesenchymal transition in HK-2 human renal proximal tubule cells. Free Radic Res 2019; 52:1387-1397. [PMID: 30693839 DOI: 10.1080/10715762.2018.1489129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Nε-(carboxymethyl) lysine (CML) plays causal roles in diabetic complications. In the present study, we investigated whether CML-induced HIF-1α accumulation and epithelial-mesenchymal transition (EMT) in HK-2 renal proximal tubular epithelial cells. Treatment with CML-BSA increased reactive oxygen species (ROS) production reduced the mitochondrial membrane potential and induced mitochondrial fragmentation. Pre-treatment of cells with antioxidant, α-lipoic acid, normalised the ROS production and restored the mitochondrial membrane potential. These changes were accompanied with morphological changes of epithelial mesenchymal transition. CML-BSA increased the protein level of hypoxia-inducible factor-1α (HIF-1α), and the EMT-associated transcription factor, TWIST. These effects were reversed by α-lipoic acid. CML-BSA increased the protein levels of mesenchymal-specific markers, including vimentin, α-smooth muscle actin, which were alleviated by pre-treatment with α-lipoic acid. Our data suggest that CML-BSA induces EMT through a ROS/HIF-1α/TWIST-dependent mechanism, and that α-lipoic acid may alleviate the CML-induced EMT in renal tubular cells.
Collapse
Affiliation(s)
- Mei-Chen Lo
- a Department of Pediatrics , Shuang Ho Hospital, Taipei Medical University , Taipei , Taiwan
| | - Ming-Hong Chen
- b Department of Pathology , Saint Paul's Hospital , Tao-Yuan , Taiwan
| | - Yu-Ting Hsueh
- c Department of Medical Laboratory Sciences and Biotechnology, College of Medical Sciences and Technology , Taipei Medical University , Taipei , Taiwan
| | - Yung-Ting Kuo
- a Department of Pediatrics , Shuang Ho Hospital, Taipei Medical University , Taipei , Taiwan.,d Department of Pediatrics, School of Medicine, College of Medicine , Taipei Medical University , Taipei , Taiwan
| | - Horng-Mo Lee
- c Department of Medical Laboratory Sciences and Biotechnology, College of Medical Sciences and Technology , Taipei Medical University , Taipei , Taiwan.,e Ph.D. Program in Medical Biotechnology, College of Medical Sciences and Technology , Taipei Medical University , Taipei , Taiwan
| |
Collapse
|
44
|
Zhang Y, Lv YL, Si YN, Zhou J, Qian Y, Bao HG. α-lipoic acid attenuates spatial learning and memory impairment induced by hepatectomy. Exp Ther Med 2019; 17:2329-2333. [PMID: 30867718 DOI: 10.3892/etm.2019.7202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 11/19/2018] [Indexed: 01/07/2023] Open
Abstract
The aim of the present study was to compare the effects of α-lipoic acid (ALA) on postoperative cognitive dysfunction (POCD) between wild type (WT) and leptin receptor-deficient (db/db) mice and to elucidate the underlying mechanism of treatment with ALA. The present study compared the effects of ALA on spatial learning and memory of WT and db/db mice using a Morris water maze following hepatectomy. The expression levels of proteins, including cyclin-dependent kinase 5 (Cdk5), tau, phosphorylated tau and amyloid β (Aβ) were measured in the hippocampus. Surgery impaired postoperative cognitive function in both WT and db/db mice. Furthermore, the expression levels of Cdk5 and Aβ, and the phosphorylation of tau in the hippocampus increased after the surgery in both WT and db/db mice. The ultrastructure of hippocampal neurons and synapses was analyzed by transmission electron microscopy and the results revealed that surgery damaged the structure of neurons and synapses in both WT and db/db mice. Treatment with ALA protected the postoperative cognitive function and the structure of hippocampal neurons and synapses, and prevented the increase in protein expression levels of Cdk5 and Aβ, and the phosphorylation of tau in the hippocampus of WT but not db/db mice. The results of the present study suggest that ALA may be used for the treatment of POCD. The molecular mechanisms underlying the activity of ALA require further investigation.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Yun-Luo Lv
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Yan-Na Si
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Jianwei Zhou
- Deparment of Molecular Cell Biology and Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Yanning Qian
- Department of Anesthesiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hong-Guang Bao
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| |
Collapse
|
45
|
Yang B, Shi MQ, Li ZH, Shi L, Wang AM, Guo XJ, Li D. Effects of n-3 fatty acid supplements on cardiometabolic profiles in hypertensive patients with abdominal obesity in Inner Mongolia: a randomized controlled trial. Food Funct 2019; 10:1661-1670. [DOI: 10.1039/c8fo01707g] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Daily supplementation with n-3 fatty acid (FA) has been believed to be an adjunct or alternative to drug treatments to reduce blood pressure (BP) and triglyceride (TG) levels in western patients with high risk of cardiovascular disease.
Collapse
Affiliation(s)
- Bo Yang
- Institute of Lipids Medicine
- Wenzhou Medical University
- Wenzhou
- China
| | - Mei-qi Shi
- Department of Food Science and Nutrition
- Zhejiang University
- Hangzhou
- China
| | - Zi-hao Li
- Department of Food Science and Nutrition
- Zhejiang University
- Hangzhou
- China
| | - Lin Shi
- Ejin Horo Banner Centre for Disease Prevention and Control
- Ordos
- China
| | - Ai-min Wang
- Ejin Horo Banner Centre for Disease Prevention and Control
- Ordos
- China
| | - Xiao-juan Guo
- Institute of Lipids Medicine
- Wenzhou Medical University
- Wenzhou
- China
| | - Duo Li
- Department of Food Science and Nutrition
- Zhejiang University
- Hangzhou
- China
- Institute of Nutrition & Health
| |
Collapse
|
46
|
Akbari M, Ostadmohammadi V, Lankarani KB, Tabrizi R, Kolahdooz F, Khatibi SR, Asemi Z. The effects of alpha-lipoic acid supplementation on glucose control and lipid profiles among patients with metabolic diseases: A systematic review and meta-analysis of randomized controlled trials. Metabolism 2018; 87:56-69. [PMID: 29990473 DOI: 10.1016/j.metabol.2018.07.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 06/24/2018] [Accepted: 07/05/2018] [Indexed: 02/08/2023]
Abstract
OBJECTIVE This systematic review and meta-analysis of randomized controlled trials (RCTs) was performed to summarize the effect of alpha-lipoic acid (ALA) supplementation on glycemic control and lipid profiles among patients with metabolic diseases. METHODS We searched the following databases till October 2017: MEDLINE, EMBASE, Web of Science and Cochrane Central Register of Controlled Trials. The relevant data were extracted and assessed for quality of the studies according to the Cochrane risk of bias tool. Data were pooled using the inverse variance method and expressed as standardized mean difference (SMD) with 95% confidence intervals (95% CI). Heterogeneity between studies was assessed by the Cochran Q statistic and I-squared tests (I2). Twenty-four studies were included in the meta-analyses. RESULTS The findings of this meta-analysis showed that ALA supplementation among patients with metabolic diseases significantly decreased fasting glucose (SMD -0.54; 95% CI, -0.89, -0.19; P = 0.003), insulin (SMD -1.01; 95% CI, -1.70, -0.31; P = 0.006), homeostasis model assessment of insulin resistance (SMD -0.76; 95% CI, -1.15, -0.36; P < 0.001) and hemoglobin A1c (SMD -1.22; 95% CI, -2.01, -0.44; P = 0.002), triglycerides (SMD -0.58; 95% CI, -1.00, -0.16; P = 0.006), total- (SMD -0.64; 95% CI, -1.01, -0.27; P = 0.001), low density lipoprotein-cholesterol (SMD -0.44; 95% CI, -0.76, -0.11; P = 0.008). We found no detrimental effect of ALA supplementation on high density lipoprotein-cholesterol (HDL-cholesterol) levels (SMD 0.57; 95% CI, -0.14, 1.29; P = 0.11). CONCLUSIONS Overall, the current meta-analysis demonstrated that ALA administration may lead to an improvement in glucose homeostasis parameters and lipid profiles except HDL-cholesterol levels.
Collapse
Affiliation(s)
- Maryam Akbari
- Health Policy Research Center, Institute of Health, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahidreza Ostadmohammadi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R., Iran
| | - Kamran B Lankarani
- Health Policy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Tabrizi
- Health Policy Research Center, Institute of Health, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fariba Kolahdooz
- Indigenous and Global Health Research, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Seyed Reza Khatibi
- Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R., Iran.
| |
Collapse
|
47
|
The protein-sparing effect of α-lipoic acid in juvenile grass carp, Ctenopharyngodon idellus: effects on lipolysis, fatty acid β-oxidation and protein synthesis. Br J Nutr 2018; 120:977-987. [DOI: 10.1017/s000711451800226x] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AbstractTo investigate the protein-sparing effect of α-lipoic acid (LA), experimental fish (initial body weight: 18·99 (sd 1·82) g) were fed on a 0, 600 or 1200 mg/kg α-LA diet for 56 d, and hepatocytes were treated with 20 μm compound C, the inhibitor of AMP kinase α (AMPKα), treated for 30 min before α-LA treatment for 24 h. LA significantly decreased lipid content of the whole body and other tissues (P<0·05), and it also promoted protein deposition in vivo (P<0·05). Further, dietary LA significantly decreased the TAG content of serum and increased the NEFA content of serum (P<0·05); however, there were no significant differences among all groups in the hepatopancreas and muscle (P>0·05). Consistent with results from the experiment in vitro, LA activated phosphorylation of AMPKα and notably increased the protein content of adipose TAG lipase in intraperitoneal fat, hepatopancreas and muscle in vivo (P<0·05). Meanwhile, LA significantly up-regulated the mRNA expression of genes involved in fatty acid β-oxidation in the same three areas, and LA also obviously down-regulated the mRNA expression of genes involved in amino acid catabolism in muscle (P<0·05). Besides, it was observed that LA significantly activated the mammalian target of rapamycin (mTOR) pathway in muscle of experimental fish (P<0·05). LA could promote lipolysis and fatty acid β-oxidation via increasing energy supply from lipid catabolism, and then, it could economise on the protein from energy production to increase protein deposition in grass carp. Besides, LA might directly promote protein synthesis through activating the mTOR pathway.
Collapse
|
48
|
Zhu Y, Huang JJ, Zhang XX, Yan Y, Yin XW, Ping G, Jiang WM. Qing Gan Zi Shen Tang alleviates adipose tissue dysfunction with up-regulation of SIRT1 in spontaneously hypertensive rat. Biomed Pharmacother 2018; 105:246-255. [DOI: 10.1016/j.biopha.2018.05.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 04/30/2018] [Accepted: 05/07/2018] [Indexed: 11/17/2022] Open
|
49
|
Guo WW, Wang X, Chen XQ, Ba YY, Zhang N, Xu RR, Zhao WW, Wu X. Flavonones from Penthorum chinense Ameliorate Hepatic Steatosis by Activating the SIRT1/AMPK Pathway in HepG2 Cells. Int J Mol Sci 2018; 19:ijms19092555. [PMID: 30154382 PMCID: PMC6165420 DOI: 10.3390/ijms19092555] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/04/2018] [Accepted: 08/22/2018] [Indexed: 12/13/2022] Open
Abstract
Pinocembrin-7-O-β-d-glucoside (PCBG), pinocembrin (PCB), and 5-methoxy-pinocembrin-7-O-β-d-glucoside (MPG) are three flavonones isolated from Penthorum chinense Pursh (P. chinense). The effects of the three flavonones on hepatic steatosis and their molecular mechanisms in HepG2 cells were investigated in this study for the first time. A model of hepatic steatosis in HepG2 cells was induced by free fatty acid (FFA), and co-treated with the three flavonones as mentioned. Intracellular lipid droplets were detected by Oil Red O staining. PCB, PCBG, and MPG suppressed oxidative stress by decreasing malondialdehyde (MDA) levels and increasing superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities. The levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were ameliorated. Moreover, these flavonones enhanced the phosphorylation of AMP-activated protein kinase (AMPK) and the expression of silent mating type information regulation 2 homolog 1 (SIRT1) and peroxisome proliferator-activated receptor α (PPARα), and reduced the expression of sterol regulatory element binding protein-1c (SREBP1c) and the downstream targets fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC), and stearoyl-CoA desaturase 1 (SCD1). Molecular docking was used to predict the interaction and combination patterns between the three flavonones and the enzymes above. The results revealed that the SIRT1/AMPK pathway is involved in the functions of the three flavonones, and the most effective flavonone against hepatic steatosis might be PCBG, followed by MPG and PCB. Therefore, the three flavonones from P. chinense were found to exert preventive effects against hepatic steatosis by regulating the SIRT1/AMPK pathway.
Collapse
Affiliation(s)
- Wei-Wei Guo
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| | - Xing Wang
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| | - Xiao-Qing Chen
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| | - Yin-Ying Ba
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| | - Nan Zhang
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| | - Rong-Rong Xu
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| | - Wen-Wen Zhao
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| | - Xia Wu
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| |
Collapse
|
50
|
Mousavi SM, Shab-Bidar S, Kord-Varkaneh H, Khorshidi M, Djafarian K. Effect of alpha-lipoic acid supplementation on lipid profile: A systematic review and meta-analysis of controlled clinical trials. Nutrition 2018; 59:121-130. [PMID: 30471524 DOI: 10.1016/j.nut.2018.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 06/28/2018] [Accepted: 08/02/2018] [Indexed: 12/22/2022]
Abstract
Several studies have shown the effect of alpha-lipoic acid (ALA) on lipid profile. However, findings remain controversial. This systematic review and meta-analysis was conducted to systematically summarize the available clinical trials that examined the effects ALA supplementation on the lipid profile of adults. A systematic search through PubMed and Scopus was done for studies published in English up to April 2017. Effect sizes were combined with fixed- or random-effects analysis, where appropriate. Between-study heterogeneity was evaluated by Cochran's Q test and I2. Eleven clinical trials with 452 adults (51.5% women, 48.5% men) were included in this meta-analysis. Combining effect sizes of 10 studies on serum triacylglycerol (TG) concentrations revealed a significant effect of ALA supplementation on serum TG compared with the placebo group (weighted mean difference [WMD], -29.185 mg/dL; 95% confidence interval [CI], -51.454 to -6.916; P = 0.010). We also found significant changes in serum total cholesterol and low-density lipoprotein (WMD, -10.683 mg/dL; 95% CI, -19.816 to -1.550; P = 0.022, WMD, -12.906 mg/dL; 95% CI, -22.133 to -3.679; P = 0.006, respectively). Significant changes were not observed in serum high-density lipoprotein (WMD, -0.092 mg/dL; 95% CI, -3.014 to 2.831; P = 0.025). Supplementation dosage and body mass index were potential sources of heterogeneity, in which those with body mass index >30 kg/m2 who received >600 mg/d ALA showed better improvements in lipid profile. Our findings showed that supplementation with ALA significantly decreased the serum concentrations of TG, total cholesterol, and low-density lipoprotein but did not affect serum levels of high-density lipoprotein in adults.
Collapse
Affiliation(s)
- Seyed Mohammad Mousavi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran; Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Sakineh Shab-Bidar
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Hamed Kord-Varkaneh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Masoud Khorshidi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Kurosh Djafarian
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|