51
|
Haghi-Aminjan H, Farhood B, Rahimifard M, Didari T, Baeeri M, Hassani S, Hosseini R, Abdollahi M. The protective role of melatonin in chemotherapy-induced nephrotoxicity: a systematic review of non-clinical studies. Expert Opin Drug Metab Toxicol 2018; 14:937-950. [PMID: 30118646 DOI: 10.1080/17425255.2018.1513492] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 08/15/2018] [Indexed: 12/18/2022]
Abstract
BSTRACT Introduction: The aim of this study was to investigate the potential role of melatonin in the prevention of chemotherapy-induced nephrotoxicity at the preclinical level. Areas to be covered: To illuminate the possible role of melatonin in preventing chemotherapy-related nephrotoxicity, Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline was followed. A comprehensive search strategy was developed to include PubMed, Web of Science, Scopus, and Embase electronic databases from their inception to May 2018. Based on a set of prespecified inclusion and exclusion criteria, 21 non-clinical articles were ultimately included in the study. Expert opinion: Our findings clearly demonstrate that melatonin has a protective role in the prevention of chemotherapy-induced nephrotoxicity which may be caused by different chemotherapy agents such as cyclophosphamide, cisplatin, doxorubicin, methotrexate, oxaliplatin, etoposide, and daunorubicin. On the basis of current review of non-clinical studies, this protective effect of melatonin is attributed to different mechanisms such as reduction of oxidative stress, apoptosis, and inflammation. The findings presented in this review are based on non-clinical studies and thus conducting appropriate clinical trials to evaluate the real effectiveness of the concurrent use of chemotherapy agents with melatonin in the cancer patients is necessary.
Collapse
Affiliation(s)
- Hamed Haghi-Aminjan
- a Department of Toxicology and Pharmacology, Faculty of Pharmacy , Tehran University of Medical Sciences , Tehran , Iran
| | - Bagher Farhood
- b Departmentof Medical Physics and Radiology, Faculty of Paramedical Sciences , Kashan University of Medical Sciences , Kashan , Iran
| | - Mahban Rahimifard
- c Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS) , Tehran University of Medical Sciences , Tehran , Iran
| | - Tina Didari
- c Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS) , Tehran University of Medical Sciences , Tehran , Iran
| | - Maryam Baeeri
- c Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS) , Tehran University of Medical Sciences , Tehran , Iran
| | - Shokoufeh Hassani
- c Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS) , Tehran University of Medical Sciences , Tehran , Iran
| | - Rohollah Hosseini
- a Department of Toxicology and Pharmacology, Faculty of Pharmacy , Tehran University of Medical Sciences , Tehran , Iran
| | - Mohammad Abdollahi
- a Department of Toxicology and Pharmacology, Faculty of Pharmacy , Tehran University of Medical Sciences , Tehran , Iran
- c Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS) , Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
52
|
Fan EKY, Fan J. Regulation of alveolar macrophage death in acute lung inflammation. Respir Res 2018; 19:50. [PMID: 29587748 PMCID: PMC5872399 DOI: 10.1186/s12931-018-0756-5] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 03/05/2018] [Indexed: 02/07/2023] Open
Abstract
Acute lung injury (ALI) and its severe form, known as acute respiratory distress syndrome (ARDS), are caused by direct pulmonary insults and indirect systemic inflammatory responses that result from conditions such as sepsis, trauma, and major surgery. The reciprocal influences between pulmonary and systemic inflammation augments the inflammatory process in the lung and promotes the development of ALI. Emerging evidence has revealed that alveolar macrophage (AM) death plays important roles in the progression of lung inflammation through its influence on other immune cell populations in the lung. Cell death and tissue inflammation form a positive feedback cycle, ultimately leading to exaggerated inflammation and development of disease. Pharmacological manipulation of AM death signals may serve as a logical therapeutic strategy for ALI/ARDS. This review will focus on recent advances in the regulation and underlying mechanisms of AM death as well as the influence of AM death on the development of ALI.
Collapse
Affiliation(s)
- Erica K Y Fan
- Kenneth P. Dietrich School of Arts & Sciences, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
53
|
Ammal Kaidery N, Thomas B. Current perspective of mitochondrial biology in Parkinson's disease. Neurochem Int 2018; 117:91-113. [PMID: 29550604 DOI: 10.1016/j.neuint.2018.03.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative movement disorder characterized by preferential loss of dopaminergic neurons of the substantia nigra pars compacta and the presence of Lewy bodies containing α-synuclein. Although the cause of PD remains elusive, remarkable advances have been made in understanding the possible causative mechanisms of PD pathogenesis. An explosion of discoveries during the past two decades has led to the identification of several autosomal dominant and recessive genes that cause familial forms of PD. The investigations of these familial PD gene products have shed considerable insights into the molecular pathogenesis of the more common sporadic PD. A growing body of evidence suggests that the etiology of PD is multifactorial and involves a complex interplay between genetic and environmental factors. Substantial evidence from human tissues, genetic and toxin-induced animal and cellular models indicates that mitochondrial dysfunction plays a central role in the pathophysiology of PD. Deficits in mitochondrial functions due to bioenergetics defects, alterations in the mitochondrial DNA, generation of reactive oxygen species, aberrant calcium homeostasis, and anomalies in mitochondrial dynamics and quality control are implicated in the underlying mechanisms of neuronal cell death in PD. In this review, we discuss how familial PD-linked genes and environmental factors interface the pathways regulating mitochondrial functions and thereby potentially converge both familial and sporadic PD at the level of mitochondrial integrity. We also provide an overview of the status of therapeutic strategies targeting mitochondrial dysfunction in PD. Unraveling potential pathways that influence mitochondrial homeostasis in PD may hold the key to therapeutic intervention for this debilitating neurodegenerative movement disorder.
Collapse
Affiliation(s)
| | - Bobby Thomas
- Departments of Pharmacology and Toxicology, Augusta, GA 30912, United States; Neurology Medical College of Georgia, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
54
|
Hennig P, Garstkiewicz M, Grossi S, Di Filippo M, French LE, Beer HD. The Crosstalk between Nrf2 and Inflammasomes. Int J Mol Sci 2018; 19:ijms19020562. [PMID: 29438305 PMCID: PMC5855784 DOI: 10.3390/ijms19020562] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 01/03/2023] Open
Abstract
The Nrf2 (nuclear factor E2-related factor or nuclear factor (erythroid-derived 2)-like 2) transcription factor is a key player in cytoprotection and activated in stress conditions caused by reactive oxygen species (ROS) or electrophiles. Inflammasomes represent central regulators of inflammation. Upon detection of various stress factors, assembly of the inflamasome protein complex results in activation and secretion of proinflammatory cytokines. In addition, inflammasome activation causes pyroptosis, a lytic form of cell death, which supports inflammation. There is growing evidence of a crosstalk between the Nrf2 and inflammasome pathways at different levels. For example, Nrf2 activating compounds inhibit inflammasomes and consequently inflammation. This review summarizes what is known about the complex and predominantly antagonistic relationship of both stress-activated pathways.
Collapse
Affiliation(s)
- Paulina Hennig
- Department of Dermatology, University Hospital of Zurich, Gloriastrasse 31, F30, CH-8091 Zurich, Switzerland.
| | - Martha Garstkiewicz
- Department of Dermatology, University Hospital of Zurich, Gloriastrasse 31, F30, CH-8091 Zurich, Switzerland.
| | - Serena Grossi
- Department of Dermatology, University Hospital of Zurich, Gloriastrasse 31, F30, CH-8091 Zurich, Switzerland.
| | - Michela Di Filippo
- Department of Dermatology, University Hospital of Zurich, Gloriastrasse 31, F30, CH-8091 Zurich, Switzerland.
| | - Lars E French
- Department of Dermatology, University Hospital of Zurich, Gloriastrasse 31, F30, CH-8091 Zurich, Switzerland.
- Faculty of Medicine, University of Zurich, CH-8091 Zurich, Switzerland.
| | - Hans-Dietmar Beer
- Department of Dermatology, University Hospital of Zurich, Gloriastrasse 31, F30, CH-8091 Zurich, Switzerland.
- Faculty of Medicine, University of Zurich, CH-8091 Zurich, Switzerland.
| |
Collapse
|
55
|
Jiang D, Chen S, Sun R, Zhang X, Wang D. The NLRP3 inflammasome: Role in metabolic disorders and regulation by metabolic pathways. Cancer Lett 2018; 419:8-19. [PMID: 29339210 DOI: 10.1016/j.canlet.2018.01.034] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 01/06/2018] [Accepted: 01/09/2018] [Indexed: 12/14/2022]
Abstract
Inflammasomes are large multimolecular complexes present in the cytosol of stimulated immune cells; they mediate the activation of caspase-1, leading to cellular pyroptosis. So far, a variety of studies on inflammasomes have emerged, and the best-studied is the NLRP3 inflammasome that is involved in many inflammatory responses. Furthermore, its relationship with metabolism is gaining increasing attention in this field. In this review, we discuss the importance of the NLRP3 inflammasome in metabolic disorders and its close association with metabolic pathways.
Collapse
Affiliation(s)
| | | | | | - Xue Zhang
- Department of Pathology and Pathophysiology, China.
| | - Di Wang
- Institute of Immunology, China.
| |
Collapse
|
56
|
Yang Y, Zhang X, Xu M, Wu X, Zhao F, Zhao C. Quercetin attenuates collagen-induced arthritis by restoration of Th17/Treg balance and activation of Heme Oxygenase 1-mediated anti-inflammatory effect. Int Immunopharmacol 2018; 54:153-162. [DOI: 10.1016/j.intimp.2017.11.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 11/03/2017] [Accepted: 11/09/2017] [Indexed: 10/18/2022]
|
57
|
Castelblanco M, Lugrin J, Ehirchiou D, Nasi S, Ishii I, So A, Martinon F, Busso N. Hydrogen sulfide inhibits NLRP3 inflammasome activation and reduces cytokine production both in vitro and in a mouse model of inflammation. J Biol Chem 2017; 293:2546-2557. [PMID: 29279328 DOI: 10.1074/jbc.m117.806869] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/17/2017] [Indexed: 12/13/2022] Open
Abstract
A variety of stimuli, including monosodium urate (MSU) crystals, activate the NLRP3 inflammasome, and this activation involves several molecular mechanisms including xanthine oxidase (XO) up-regulation and mitochondrial dysfunction. Upon oligomerization of apoptosis-associated speck-like protein containing a CARD (ASC), caspase-1 becomes active and cleaves the proinflammatory cytokine IL-1β into its active secreted form. Hydrogen sulfide (H2S), a gasotransmitter mainly produced by cystathionine γ-lyase (CSE) in macrophages, could modulate inflammation. Here, we sought to investigate the effects of exogenous and endogenous H2S on NLRP3 inflammasome activation in vitro and in vivo Primed bone marrow-derived macrophages (BMDM) isolated from wildtype (wt) or CSE-deficient mice and human macrophages (THP1 cells and primary macrophages), were stimulated with MSU crystals in the presence or absence of a H2S donor, sodium thiosulfate (STS) or GYY4137 (GYY). In murine and human macrophages in vitro, both STS and GYY inhibited MSU crystal-induced IL-1β secretion in a dose-dependent manner. Moreover, the H2S donors inhibited MSU crystal-induced XO/caspase-1 activities, mitochondrial reactive oxygen species (ROS) generation, and ASC oligomerization. Accordingly, IL-1β secretion and XO/caspase-1 activities were higher in CSE-deficient BMDMs than in wt BMDMs. For in vivo studies, we experimentally induced peritonitis by intraperitoneal injection of MSU crystals into mice. GYY pretreatment ameliorated inflammation, evidenced by decreased IL-6/monocyte chemoattractant protein-1 (MCP-1) released into peritoneal lavages. Taken together, our results suggest that both exogenous (via H2S donors) and endogenous (via CSE) H2S production may represent approaches for managing, for example, acute gout or other inflammation conditions.
Collapse
Affiliation(s)
| | - Jérôme Lugrin
- the Department of Biochemistry, University of Lausanne, Epalinges 155 1066, Switzerland and
| | - Driss Ehirchiou
- the Department of Biochemistry, University of Lausanne, Epalinges 155 1066, Switzerland and
| | - Sonia Nasi
- From the Service of Rheumatology, DAL, Lausanne University Hospital (CHUV) and
| | - Isao Ishii
- Showa Pharmaceutical University, Tokyo 194-8543, Japan
| | - Alexander So
- From the Service of Rheumatology, DAL, Lausanne University Hospital (CHUV) and
| | - Fabio Martinon
- the Department of Biochemistry, University of Lausanne, Epalinges 155 1066, Switzerland and
| | - Nathalie Busso
- From the Service of Rheumatology, DAL, Lausanne University Hospital (CHUV) and
| |
Collapse
|
58
|
The role of Nrf2 in NLRP3 inflammasome activation. Cell Mol Immunol 2017; 14:1011-1012. [PMID: 29129911 DOI: 10.1038/cmi.2017.114] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 09/10/2017] [Indexed: 11/08/2022] Open
|
59
|
Cleophas MC, Joosten LA, Stamp LK, Dalbeth N, Woodward OM, Merriman TR. ABCG2 polymorphisms in gout: insights into disease susceptibility and treatment approaches. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2017; 10:129-142. [PMID: 28461764 PMCID: PMC5404803 DOI: 10.2147/pgpm.s105854] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
As a result of the association of a common polymorphism (rs2231142, Q141K) in the ATP-binding cassette G2 (ABCG2) transporter with serum urate concentration in a genome-wide association study, it was revealed that ABCG2 is an important uric acid transporter. This review discusses the relevance of ABCG2 polymorphisms in gout, possible etiological mechanisms, and treatment approaches. The 141K ABCG2 urate-increasing variant causes instability in the nucleotide-binding domain, leading to decreased surface expression and function. Trafficking of the protein to the cell membrane is altered, and instead, there is an increased ubiquitin-mediated proteasomal degradation of the variant protein as well as sequestration into aggresomes. In humans, this leads to decreased uric acid excretion through both the kidney and the gut with the potential for a subsequent compensatory increase in renal urinary excretion. Not only does the 141K polymorphism in ABCG2 lead to hyperuricemia through renal overload and renal underexcretion, but emerging evidence indicates that it also increases the risk of acute gout in the presence of hyperuricemia, early onset of gout, tophi formation, and a poor response to allopurinol. In addition, there is some evidence that ABCG2 dysfunction may promote renal dysfunction in chronic kidney disease patients, increase systemic inflammatory responses, and decrease cellular autophagic responses to stress. These results suggest multiple benefits in restoring ABCG2 function. It has been shown that decreased ABCG2 141K surface expression and function can be restored with colchicine and other small molecule correctors. However, caution should be exercised in any application of these approaches given the role of surface ABCG2 in drug resistance.
Collapse
Affiliation(s)
- M C Cleophas
- Department of Internal Medicine.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - L A Joosten
- Department of Internal Medicine.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - L K Stamp
- Department of Medicine, University of Otago Christchurch, Christchurch
| | - N Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - O M Woodward
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
60
|
|
61
|
Liu X, Zhang X, Ding Y, Zhou W, Tao L, Lu P, Wang Y, Hu R. Nuclear Factor E2-Related Factor-2 Negatively Regulates NLRP3 Inflammasome Activity by Inhibiting Reactive Oxygen Species-Induced NLRP3 Priming. Antioxid Redox Signal 2017; 26:28-43. [PMID: 27308893 PMCID: PMC5198158 DOI: 10.1089/ars.2015.6615] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS The NLRP3 inflammasome is a multiprotein complex that protects hosts against a variety of pathogens. However, the molecular mechanisms of modulating NLRP3 inflammasome activation, especially at the priming step, are still poorly understood. This study was designed to elucidate the negative regulation of nuclear factor E2-related factor-2 (Nrf2) on the activation of NLRP3 inflammasome. RESULTS We reported that Nrf2 activation inhibited NLRP3 expression, caspase-1 cleavage, and subsequent IL-1β generation. Compared with normal cells, Nrf2-deficient cells showed upregulated cleaved caspase-1, which were attributed to the increased transcription of NLRP3 caused by excess reactive oxygen species (ROS). Furthermore, priming of the NLRP3 inflammasome was sensitive to the exogenous ROS levels induced by H2O2 or rotenone. Combined with adenosine triphosphate, rotenone triggered higher activity of the NLRP3 inflammasome compared with lipopolysaccharide, suggesting that ROS promoted the priming step. In addition, Nrf2-induced NQO1 was involved in the inhibition of the NLRP3 inflammasome. In an in vivo alum-induced peritonitis mouse model, Nrf2 activation suppressed typical IL-1 signaling-dependent inflammation, whereas Nrf2-/- mice exhibited a significant increase in the recruitment of immune cell and the generation of IL-1β compared with wild-type mice. INNOVATION We elucidated the effects and possible mechanisms of Nrf2 activation-induced NQO1 expression on NLRP3 inflammasome inactivation and established a novel regulatory role of the Nrf2 pathway in ROS-induced NLRP3 priming. CONCLUSIONS We demonstrated Nrf2 negatively regulating NLRP3 inflammasome activity by inhibiting the priming step and suggested that Nrf2 could be a potential target for some uncontrolled inflammasome activation-associated diseases. Antioxid. Redox Signal. 26, 28-43.
Collapse
Affiliation(s)
- Xiuting Liu
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing, China
| | - Xin Zhang
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing, China
| | - Yang Ding
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing, China
| | - Wei Zhou
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing, China
| | - Lei Tao
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing, China
| | - Ping Lu
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing, China
| | - Yajing Wang
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing, China
| | - Rong Hu
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing, China
| |
Collapse
|
62
|
Role of Uric Acid Metabolism-Related Inflammation in the Pathogenesis of Metabolic Syndrome Components Such as Atherosclerosis and Nonalcoholic Steatohepatitis. Mediators Inflamm 2016; 2016:8603164. [PMID: 28070145 PMCID: PMC5192336 DOI: 10.1155/2016/8603164] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 11/03/2016] [Accepted: 11/15/2016] [Indexed: 02/07/2023] Open
Abstract
Uric acid (UA) is the end product of purine metabolism and can reportedly act as an antioxidant. However, recently, numerous clinical and basic research approaches have revealed close associations of hyperuricemia with several disorders, particularly those comprising the metabolic syndrome. In this review, we first outline the two molecular mechanisms underlying inflammation occurrence in relation to UA metabolism; one is inflammasome activation by UA crystallization and the other involves superoxide free radicals generated by xanthine oxidase (XO). Importantly, recent studies have demonstrated the therapeutic or preventive effects of XO inhibitors against atherosclerosis and nonalcoholic steatohepatitis, which were not previously considered to be related, at least not directly, to hyperuricemia. Such beneficial effects of XO inhibitors have been reported for other organs including the kidneys and the heart. Thus, a major portion of this review focuses on the relationships between UA metabolism and the development of atherosclerosis, nonalcoholic steatohepatitis, and related disorders. Although further studies are necessary, XO inhibitors are a potentially novel strategy for reducing the risk of many forms of organ failure characteristic of the metabolic syndrome.
Collapse
|
63
|
Rebamipide Suppresses Monosodium Urate Crystal-Induced Interleukin-1β Production Through Regulation of Oxidative Stress and Caspase-1 in THP-1 Cells. Inflammation 2016; 39:473-482. [PMID: 26454448 DOI: 10.1007/s10753-015-0271-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This study investigated the effect of rebamipide on activation of the NLRP3 inflammasome and generation of reactive oxygen species (ROS) in monosodium urate (MSU) crystal-induced interleukin-1β (IL-1β) production. Human monocyte cell line THP-1 and human umbilical venous endothelial cells (HUVECs) were used to assess the inflammatory response to MSU crystals. NADP/NADPH activity assays were used as a marker of ROS generation. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting were performed to evaluate levels of IL-1β, caspase-1, NLRP3, associated speck-like protein (ASC), nuclear factor-κB (NF-κB), p65, IκBα, intercellular adhesion molecule 1 (ICAM-1), and vascular cell adhesion molecule 1 (VCAM-1). Experimental pharmaceuticals included rebamipide, colchicine, dexamethasone, and ascorbic acid. In THP-1 cells, treatment with MSU crystals increased NADP/NADPH ratios and IL-1β expression, and both of these responses were potently inhibited by addition of rebamipide. Rebamipide also attenuated enhanced expression of caspase-1 gene by MSU crystals (p < 0.05). Western blotting demonstrated that MSU crystals stimulated caspase-1 but not NLRP3 and ASC activation. Similarly, MSU crystals activated the NF-κB pathway, which in turn was blocked by rebamipide. Stimulation of HUVECs with MSU crystals increased expression of VCAM-1 and ICAM-1, which were markedly inhibited by both rebamipide and dexamethasone. This study demonstrated that rebamipide inhibits IL-1β activation through suppression of ROS-mediated NF-κB signaling pathways and caspase-1 activation in MSU crystal-induced inflammation.
Collapse
|
64
|
Zheng X, Bao W, Yang J, Zhang T, Sun D, Liang Y, Li S, Wang Y, Feng X, Hao H, Wang Z. Focal Adhesion Kinase Directly Interacts with TSC2 Through Its FAT Domain and Regulates Cell Proliferation in Cashmere Goat Fetal Fibroblasts. DNA Cell Biol 2016; 35:480-8. [PMID: 27380318 DOI: 10.1089/dna.2015.3033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Focal adhesion kinase (FAK) is a cytoplasmic nonreceptor tyrosine kinase that senses a variety of extracellular signals, such as growth factors and integrins, to control the process of cell proliferation and metabolism. We cloned three goat FAK transcript variants (KM655805, KM658268, and KM658269) that encode 1052, 1006, and 962 amino-acid residue proteins. Bioinformatics analysis indicated that the putative FAK protein contains an FERM domain, a PTK domain, two Proline-rich regions, and a focal adhesion-targeting (FAT) domain. All the three transcript variants of FAK were detected in seven different goat tissues, and variant 1 had the most accumulation whereas variant 2 and variant 3 had lower accumulation. Treatment of goat fetal fibroblasts (GFbs) with a specific FAK inhibitor, TAE226, inhibited cell proliferation (p < 0.05) and induced damage to the cell morphology in a dose- and time-dependent manner. Further research demonstrated that FAK directly interacted with TSC2 (Tuberous sclerosis 2) tuberin domain through its C-terminus, which contains the complete FAT domain. In conclusion, our results indicated that FAK may be widely expressed in Cashmere goat tissues and its products participate in the mammalian target of rapamycin signaling pathway and cell proliferation through a direct interaction with TSC2 in GFBs.
Collapse
Affiliation(s)
- Xu Zheng
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China .,2 Hulunbeier Municipal People's Hospital , Hailaer, People's Republic of China
| | - Wenlei Bao
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Jiaofu Yang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Tao Zhang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Dongsheng Sun
- 3 Department of Oncology, Kailuan General Hospital , Tangshan, People's Republic of China
| | - Yan Liang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Shuyu Li
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Yanfeng Wang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Xue Feng
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Huifang Hao
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Zhigang Wang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| |
Collapse
|
65
|
Zamudio-Cuevas Y, Martínez-Flores K, Fernández-Torres J, Loissell-Baltazar YA, Medina-Luna D, López-Macay A, Camacho-Galindo J, Hernández-Díaz C, Santamaría-Olmedo MG, López-Villegas EO, Oliviero F, Scanu A, Cerna-Cortés JF, Gutierrez M, Pineda C, López-Reyes A. Monosodium urate crystals induce oxidative stress in human synoviocytes. Arthritis Res Ther 2016; 18:117. [PMID: 27209322 PMCID: PMC4875700 DOI: 10.1186/s13075-016-1012-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 05/03/2016] [Indexed: 12/11/2022] Open
Abstract
Background Gout is the most common inflammatory arthropathy of metabolic origin and it is characterized by intense inflammation, the underlying mechanisms of which are unknown. The aim of this study was to evaluate the oxidative stress in human fibroblast-like synoviocytes (FLS) exposed to monosodium urate (MSU) crystals, which trigger an inflammatory process. Methods Human FLS isolated from synovial tissue explants were stimulated with MSU crystals (75 μg/mL) for 24 h. Cellular viability was evaluated by crystal violet staining, apoptosis was assessed using Annexin V, and the cellular content of reactive oxygen species (ROS) and nitrogen species (RNS) (O2-, H2O2, NO) was assessed with image-based cytometry and fluorometric methods. In order to determine protein oxidation levels, protein carbonyls were detected through oxyblot analysis, and cell ultrastructural changes were assessed by transmission electron microscopy. Results The viability of FLS exposed to MSU crystals decreased by 30 % (P < 0.05), while apoptosis increased by 42 % (P = 0.01). FLS stimulated with MSU crystals exhibited a 2.1-fold increase in H2O2 content and a 1.5-fold increase in O2- and NO levels. Oxyblots revealed that the spots obtained from FLS protein lysates exposed to MSU crystals exhibited protein carbonyl immunoreactivity, which reflects the presence of oxidatively modified proteins. Concomitantly, MSU crystals triggered the induction of changes in the morphostructure of FLS, such as the thickening and discontinuity of the endoplasmic reticulum, and the formation of vacuoles and misfolded glycoproteins. Conclusions Our results prove that MSU crystals induce the release of ROS and RNS in FLS, subsequently oxidizing proteins and altering the cellular oxidative state of the endoplasmic reticulum, which results in FLS apoptosis. Electronic supplementary material The online version of this article (doi:10.1186/s13075-016-1012-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yessica Zamudio-Cuevas
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calzada México-Xochimilco 289, Tlalpan, 14389, Mexico City, Mexico.,Laboratorio de Microbiología Molecular, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Prolongación de Carpio y Plan de Ayala S/N Col. Casco de Santo Tomas, Miguel Hidalgo, 11340, Mexico City, Mexico
| | - Karina Martínez-Flores
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calzada México-Xochimilco 289, Tlalpan, 14389, Mexico City, Mexico
| | - Javier Fernández-Torres
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calzada México-Xochimilco 289, Tlalpan, 14389, Mexico City, Mexico.,Biological and Health Sciences PhD program, Universidad Autónoma Metropolitana, Avenida San Rafael Atlixco 186, Iztapalapa, 09340, Mexico City, Mexico
| | - Yahir A Loissell-Baltazar
- Laboratorio de Microbiología Molecular, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Prolongación de Carpio y Plan de Ayala S/N Col. Casco de Santo Tomas, Miguel Hidalgo, 11340, Mexico City, Mexico
| | - Daniel Medina-Luna
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calzada México-Xochimilco 289, Tlalpan, 14389, Mexico City, Mexico
| | - Ambar López-Macay
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calzada México-Xochimilco 289, Tlalpan, 14389, Mexico City, Mexico
| | - Javier Camacho-Galindo
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calzada México-Xochimilco 289, Tlalpan, 14389, Mexico City, Mexico
| | - Cristina Hernández-Díaz
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calzada México-Xochimilco 289, Tlalpan, 14389, Mexico City, Mexico
| | - Mónica G Santamaría-Olmedo
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calzada México-Xochimilco 289, Tlalpan, 14389, Mexico City, Mexico
| | - Edgar Oliver López-Villegas
- Laboratorio Central de Microscopía, Departamento de Investigación, ENCB, IPN, Prolongación de Carpio y Plan de Ayala S/N Col. Santo Tomás, Miguel Hidalgo, 11340, Mexico City, Mexico
| | - Francesca Oliviero
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Via Giustiniani, 2, Padova, 35128, Italy
| | - Anna Scanu
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Via Giustiniani, 2, Padova, 35128, Italy
| | - Jorge Francisco Cerna-Cortés
- Laboratorio de Microbiología Molecular, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Prolongación de Carpio y Plan de Ayala S/N Col. Casco de Santo Tomas, Miguel Hidalgo, 11340, Mexico City, Mexico
| | - Marwin Gutierrez
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calzada México-Xochimilco 289, Tlalpan, 14389, Mexico City, Mexico
| | - Carlos Pineda
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calzada México-Xochimilco 289, Tlalpan, 14389, Mexico City, Mexico
| | - Alberto López-Reyes
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calzada México-Xochimilco 289, Tlalpan, 14389, Mexico City, Mexico.
| |
Collapse
|
66
|
Shi YC, Lin KS, Jhai YF, Lee BH, Han Y, Cui Z, Hsu WH, Wu SC. Miracle Fruit (Synsepalum dulcificum) Exhibits as a Novel Anti-Hyperuricaemia Agent. Molecules 2016; 21:140. [PMID: 26821007 PMCID: PMC6274476 DOI: 10.3390/molecules21020140] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 01/15/2016] [Accepted: 01/20/2016] [Indexed: 01/20/2023] Open
Abstract
Miracle fruit (Synsepalum dulcificum) belongs to the Sapotaceae family. It can change flavors on taste buds, transforming acidic tastes to sweet. We evaluated various miracle fruit extracts, including water, butanol, ethyl acetate (EA), and hexane fractions, to determine its antioxidant effects. These extracts isolated from miracle fruit exerted potential for reduction of uric acid and inhibited xanthine oxidase activity in vitro and in monosodiumurate (MSU)-treated RAW264.7 macrophages. Moreover, we also found that the butanol extracts of miracle fruit attenuated oxonic acid potassium salt-induced hyperuricaemia in ICR mice by lowering serum uric acid levels and activating hepatic xanthine oxidase. These effects were equal to those of allopurinol, suggesting that the butanol extract of miracle fruit could be developed as a novel anti-hyperuricaemia agent or health food.
Collapse
Affiliation(s)
- Yeu-Ching Shi
- Taiwan Indigena Botanica Co., Ltd., Taipei 11458, Taiwan.
| | - Kai-Sian Lin
- Department of Food Sciences, National Chiayi University, Chiayi 60004, Taiwan.
| | - Yi-Fen Jhai
- Department of Food Sciences, National Chiayi University, Chiayi 60004, Taiwan.
| | - Bao-Hong Lee
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11042, Taiwan.
- Department of Traditional Chinese Medicine, Taipei Medical University Hospital, Taipei 10042, Taiwan.
| | - Yifan Han
- Department of Oral Pathology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Zhibin Cui
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA.
| | - Wei-Hsuan Hsu
- Biochemical Process Technology Department, Center of Excellence for Drug Development, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu 30058, Taiwan.
| | - She-Ching Wu
- Department of Food Sciences, National Chiayi University, Chiayi 60004, Taiwan.
| |
Collapse
|
67
|
Reactive oxygen species and mitochondria: A nexus of cellular homeostasis. Redox Biol 2015; 6:472-485. [PMID: 26432659 PMCID: PMC4596921 DOI: 10.1016/j.redox.2015.09.005] [Citation(s) in RCA: 781] [Impact Index Per Article: 78.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/08/2015] [Accepted: 09/09/2015] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are integral components of multiple cellular pathways even though excessive or inappropriately localized ROS damage cells. ROS function as anti-microbial effector molecules and as signaling molecules that regulate such processes as NF-kB transcriptional activity, the production of DNA-based neutrophil extracellular traps (NETs), and autophagy. The main sources of cellular ROS are mitochondria and NADPH oxidases (NOXs). In contrast to NOX-generated ROS, ROS produced in the mitochondria (mtROS) were initially considered to be unwanted by-products of oxidative metabolism. Increasing evidence indicates that mtROS have been incorporated into signaling pathways including those regulating immune responses and autophagy. As metabolic hubs, mitochondria facilitate crosstalk between the metabolic state of the cell with these pathways. Mitochondria and ROS are thus a nexus of multiple pathways that determine the response of cells to disruptions in cellular homeostasis such as infection, sterile damage, and metabolic imbalance. In this review, we discuss the roles of mitochondria in the generation of ROS-derived anti-microbial effectors, the interplay of mitochondria and ROS with autophagy and the formation of DNA extracellular traps, and activation of the NLRP3 inflammasome by ROS and mitochondria.
Collapse
|
68
|
Jhang JJ, Lu CC, Ho CY, Cheng YT, Yen GC. Protective Effects of Catechin against Monosodium Urate-Induced Inflammation through the Modulation of NLRP3 Inflammasome Activation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:7343-7352. [PMID: 26234731 DOI: 10.1021/acs.jafc.5b02605] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Gouty inflammation results from the stimulation of monosodium urate (MSU). Interleukin-1β (IL-1β) secretion is the primary clinical manifestation of MSU attack, and MSU activates IL-1β through a nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome. This study investigated the protective effect and underlying mechanism of naturally occurring phenolic compounds on MSU-induced inflammation in vivo and in vitro. A screening of phenolic compounds revealed that gallic acid and catechin exhibited the most potent free radical scavenging activities. Subcutaneous injection of gallic acid or catechin significantly reduced MSU-induced IL-1β and IL-6 secretion in C57BL/6 mice. However, only catechin inhibited MSU-induced IL-1β secretion and NLRP3 inflammasome activation in MSU-challenged THP-1 cells. MSU-triggered mitochondrial reactive oxygen species (MtROS) production and intracellular calcium levels were significantly decreased by treatment with catechin in THP-1 cells. Catechin treatment also up-regulated Bcl-2 levels and restored MSU-induced mitochondrial transmembrane potential impairment. These results indicate that the protective effects of catechin on MSU-induced IL-1β secretion are associated with modulation of mitochondrial damage. It also suggests that catechin has the potential to protect gout attack by modulation of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Jhih-Jia Jhang
- Department of Food Science and Biotechnology, National Chung Hsing University , 250 Kuokuang Road, Taichung 40227, Taiwan
| | - Chi-Cheng Lu
- Department of Food Science and Biotechnology, National Chung Hsing University , 250 Kuokuang Road, Taichung 40227, Taiwan
| | - Cheng-Ying Ho
- Department of Food Science and Biotechnology, National Chung Hsing University , 250 Kuokuang Road, Taichung 40227, Taiwan
| | - Yu-Ting Cheng
- Department of Food Science and Biotechnology, National Chung Hsing University , 250 Kuokuang Road, Taichung 40227, Taiwan
| | - Gow-Chin Yen
- Department of Food Science and Biotechnology, National Chung Hsing University , 250 Kuokuang Road, Taichung 40227, Taiwan
- Agricultural Biotechnology Center, National Chung Hsing University , Taichung 40227, Taiwan
| |
Collapse
|
69
|
Bao W, Wang Y, Fu Y, Jia X, Li J, Vangan N, Bao L, Hao H, Wang Z. mTORC1 Regulates Flagellin-Induced Inflammatory Response in Macrophages. PLoS One 2015; 10:e0125910. [PMID: 25942007 PMCID: PMC4420466 DOI: 10.1371/journal.pone.0125910] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 03/25/2015] [Indexed: 11/28/2022] Open
Abstract
Bacterial flagellin triggers inflammatory responses. Phosphoinositide 3-kinase (PI3K) and mammalian target of rapamycin (mTOR) regulate the production of pro- and anti-inflammatory cytokines that are induced by extrinsic antigens, but the function of mTORC1 in flagellin-induced inflammatory response is unknown. The purpose of this study was to examine the role and the mechanism of PI3K/Akt/mTOR pathway in flagellin-induced cytokine expression in mouse macrophages. We observed that flagellin upregulated TNF-α time- and dose-dependently. Flagellin stimulated rapid (<15 min) PI3K/Akt/mTOR phosphorylation that was mediated by TLR5. Inhibition of PI3K with LY294002 and wortmannin, and of mTORC1 with rapamycin decreased flagellin-induced TNF-α and IL-6 expression and cell proliferation. The activation of NF-κB p65 and STAT3 was regulated by mTORC1 via degradation of IκBα and phosphorylation of STAT3 in response to flagellin, respectively. Thus, the PI3K/Akt/mTORC1 pathway regulates the innate immune response to bacterial flagellin. Rapamycin is potential therapy that can regulate host defense against pathogenic infections.
Collapse
Affiliation(s)
- Wenlei Bao
- College of Life Science, Inner Mongolia University, Hohhot, China
| | - Yanfeng Wang
- College of Life Science, Inner Mongolia University, Hohhot, China
| | - Yuting Fu
- College of Life Science, Inner Mongolia University, Hohhot, China
| | - Xiaoyang Jia
- College of Life Science, Inner Mongolia University, Hohhot, China
| | - Jiaxin Li
- College of Life Science, Inner Mongolia University, Hohhot, China
| | | | - Lili Bao
- College of Life Science, Inner Mongolia University, Hohhot, China
- College of Basic Medical Science, Inner Mongolia Medical University, Hohhot, China
| | - Huifang Hao
- College of Life Science, Inner Mongolia University, Hohhot, China
| | - Zhigang Wang
- College of Life Science, Inner Mongolia University, Hohhot, China
- * E-mail:
| |
Collapse
|
70
|
Molecular basis of oxidative stress in gouty arthropathy. Clin Rheumatol 2015; 34:1667-72. [PMID: 25854697 DOI: 10.1007/s10067-015-2933-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 03/24/2015] [Accepted: 03/25/2015] [Indexed: 02/04/2023]
Abstract
Gout is a disorder of urate metabolism in which persistent high urate levels in the extracellular fluids result in the deposition of monosodium urate (MSU) crystal in joints and periarticular tissues. In recent years, this disease represents an increasingly common health problem, so the pace of investigation in the field has accelerated tremendously. New research advances in the pathogenesis of hyperuricemia and in the understanding of how MSU crystals induce an acute gouty attack have been focused in this review on the processes of inflammation and involvement of the innate immune response; in addition, we discuss new knowledge about the role of the reactive oxygen species in establishing oxidative stress in MSU crystal-induced arthritis.
Collapse
|
71
|
Effects of red wine on postprandial stress: potential implication in non-alcoholic fatty liver disease development. Eur J Nutr 2015; 54:497-507. [PMID: 25772634 DOI: 10.1007/s00394-015-0877-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/06/2015] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Red wine consumption is considered to be protective against oxidative stress. Diet strongly influences non-alcoholic fatty liver disease, which is associated with oxidative stress and is considered the hepatic manifestation of the metabolic syndrome. METHODS We reviewed the available evidence that investigated the effects of red wine on the postprandial-induced metabolic and oxidative stress in humans. RESULTS After red wine consumption with meal, despite the improvement in non-enzymatic antioxidant capacity and lipoperoxidation markers, the influence of confounding factors such as uric acid should be taken into account. Both uric acid and triglycerides increases, induced by ethanol, could cause liver damage. On the other hand, further researches are required in order to understand the meaning of the induction of antioxidant enzymes by red wine and red wine polyphenols in the context of non-alcoholic fatty liver disease. CONCLUSION In conclusion, inconsistent and contrasting findings exist regarding the potential benefits of red wine consumption against postprandial stress.
Collapse
|