1
|
Sinke L, Beekman M, Raz Y, Gehrmann T, Moustakas I, Boulinguiez A, Lakenberg N, Suchiman E, Bogaards FA, Bizzarri D, van den Akker EB, Waldenberger M, Butler‐Browne G, Trollet C, de Groot CPGM, Heijmans BT, Slagboom PE. Tissue-specific methylomic responses to a lifestyle intervention in older adults associate with metabolic and physiological health improvements. Aging Cell 2025; 24:e14431. [PMID: 39618079 PMCID: PMC11984676 DOI: 10.1111/acel.14431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/24/2024] [Accepted: 11/14/2024] [Indexed: 04/12/2025] Open
Abstract
Across the lifespan, diet and physical activity profiles substantially influence immunometabolic health. DNA methylation, as a tissue-specific marker sensitive to behavioral change, may mediate these effects through modulation of transcription factor binding and subsequent gene expression. Despite this, few human studies have profiled DNA methylation and gene expression simultaneously in multiple tissues or examined how molecular levels react and interact in response to lifestyle changes. The Growing Old Together (GOTO) study is a 13-week lifestyle intervention in older adults, which imparted health benefits to participants. Here, we characterize the DNA methylation response to this intervention at over 750 thousand CpGs in muscle, adipose, and blood. Differentially methylated sites are enriched for active chromatin states, located close to relevant transcription factor binding sites, and associated with changing expression of insulin sensitivity genes and health parameters. In addition, measures of biological age are consistently reduced, with decreases in grimAge associated with observed health improvements. Taken together, our results identify responsive molecular markers and demonstrate their potential to measure progression and finetune treatment of age-related risks and diseases.
Collapse
Affiliation(s)
- Lucy Sinke
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
| | - Marian Beekman
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
| | - Yotam Raz
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
| | - Thies Gehrmann
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
- Department of Bioscience Engineering, Research Group Environmental Ecology and Applied MicrobiologyUniversity of AntwerpAntwerpBelgium
| | - Ioannis Moustakas
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
- Sequencing Analysis Support Core, Department of Biomedical Data SciencesLeiden University Medical CenterLeidenThe Netherlands
| | - Alexis Boulinguiez
- Myology Center for Research, U974Sorbonne Université, INSERM, AIM, GH Pitié Salpêtrière Bat BabinskiParisFrance
| | - Nico Lakenberg
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
| | - Eka Suchiman
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
| | - Fatih A. Bogaards
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
- Division of Human NutritionWageningen University and ResearchWageningenThe Netherlands
| | - Daniele Bizzarri
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
- Delft Bioinformatics Lab, Pattern Recognition and BioinformaticsDelftThe Netherlands
| | - Erik B. van den Akker
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
- Delft Bioinformatics Lab, Pattern Recognition and BioinformaticsDelftThe Netherlands
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Institute of EpidemiologyHelmholtz Munich, German Research Center for Environmental HealthNeuherbergGermany
- German Center for Cardiovascular Research (DZHK)Partner Site Munich Heart AllianceMunichGermany
| | - Gillian Butler‐Browne
- Myology Center for Research, U974Sorbonne Université, INSERM, AIM, GH Pitié Salpêtrière Bat BabinskiParisFrance
| | - Capucine Trollet
- Myology Center for Research, U974Sorbonne Université, INSERM, AIM, GH Pitié Salpêtrière Bat BabinskiParisFrance
| | - C. P. G. M. de Groot
- Division of Human NutritionWageningen University and ResearchWageningenThe Netherlands
| | - Bastiaan T. Heijmans
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
| | - P. Eline Slagboom
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
| |
Collapse
|
2
|
Zakaria N, Menze ET, Elsherbiny DA, Tadros MG, George MY. Lycopene mitigates paclitaxel-induced cognitive impairment in mice; Insights into Nrf2/HO-1, NF-κB/NLRP3, and GRP-78/ATF-6 axes. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111262. [PMID: 39848561 DOI: 10.1016/j.pnpbp.2025.111262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
Chemotherapy-induced cognitive impairment, referred to as "chemobrain", is widely acknowledged as a significant adverse effect of cancer therapy. Paclitaxel, a chemotherapeutic drug, has been reported to cause cognitive impairment clinically and in animal models. However, the precise mechanisms are not fully understood. The current study explored the potential neuroprotective effect of lycopene in paclitaxel-induced cognitive impairment in mice and its potential underlying mechanisms. Mice were randomly allocated into six groups: control, paclitaxel-treated (10 mg/kg), lycopene-treated (5, 10, and 20 mg/kg) + paclitaxel, and lycopene alone-treated (20 mg/kg) groups. The effect of lycopene treatment on behavioral function and histological examination was assessed. Lycopene (20 mg/kg) was selected for additional investigation into the underlying mechanisms. Lycopene treatment counteracted paclitaxel-induced oxidative stress by reducing lipid peroxidation and enhancing catalase levels. Additionally, lycopene-treated mice demonstrated a significant elevation in nuclear factor erythroid 2-related factor 2 with no significant effect on hemeoxygenase-1. Moreover, paclitaxel administration elevated endoplasmic reticulum stress markers; glucose-regulated protein78, activating Transcription Factor 6, C/EBP homologous protein, and apoptosis marker annexin V which were significantly reduced by lycopene treatment. Furthermore, lycopene mitigated paclitaxel-induced neuroinflammation through the reduction of the levels of the NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome axis markers; nuclear factor-κB, NLRP3, caspase-1, interleukin-1β, and interleukin-18. Our study findings may provide new evidence that lycopene mitigates paclitaxel-induced cognitive impairment in mice by reversing oxidative stress, endoplasmic reticulum stress, and inflammatory mechanisms.
Collapse
Affiliation(s)
- Nora Zakaria
- Armed Forces Medical Complex- Kobry El-Qobba, Ministry of Defense, Kobry El-Qobba, Cairo 11766, Egypt
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Doaa A Elsherbiny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Mariane G Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt.
| |
Collapse
|
3
|
Liu Y, Liu J, Ren R, Xin Z, Luo Y, Chen Y, Huang C, Liu Y, Yang T, Wang X. Short-term and long-term high-fat diet promote metabolic disorder through reprogramming mRNA m 6A in white adipose tissue by gut microbiota. MICROBIOME 2025; 13:75. [PMID: 40091072 PMCID: PMC11912683 DOI: 10.1186/s40168-025-02047-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 01/22/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Although short-term high-fat diet (S-HFD) and long-term high-fat diet (L-HFD) induce metabolic disorder, the underlying epigenetic mechanism is still unclear. RESULTS Here, we found that both 4 days of S-HFD and 10 weeks of L-HFD increased mRNA m6A level in epididymal white adipose tissue (eWAT) and impaired metabolic health. Interestingly, S-HFD activated transposable elements (TEs), especially endogenous retroviruses (ERVs) in eWAT, while L-HFD activated long interspersed elements (LINEs). Subsequently, we demonstrated that both S-HFD and L-HFD increased m6A level of Ehmt2 and decreased EHMT2 protein expression and H3K9me2 level, accounting for activation of ERVs and LINEs. Overexpression of EHMT2 in eWAT or inhibition of ERVs and LINEs by antiviral therapy improved metabolic health under HFD feeding. Notably, we found that both short-term and long-term HFD feeding increased Fimicutes/Bacteroidota ratio and decreased the gut microbiome health index. Fecal microbiota transplantation (FMT) experiments demonstrated that gut microbiota from S-HFD and L-HFD was responsible for increased m6A level in eWAT, resulting in glucose intolerance and insulin insensitivity. Furthermore, we identified that both S-HFD and L-HFD increased the abundance of the gut microbial metabolite homogentisic acid (HGA), and HGA level was positively correlated with unclassified_f__Lachnospiraceae which was both increased in S-HFD and L-HFD feeding mice. Administration of HGA increased the m6A level of Ehmt2 and decreased the EHMT2 protein expression and H3K9me2 level in eWAT, leading to metabolic disorder in mice. CONCLUSIONS Together, this study reveals a novel mechanism that S-HFD and L-HFD induce metabolism disorder through gut microbiota-HGA-m6A-Ehmt2-ERV/LINE signaling. These findings may provide a novel insight for prevention and treatment of metabolism disorder upon short-term or long-term dietary fat intake. Video Abstract.
Collapse
Affiliation(s)
- Youhua Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Jiaqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Ruiti Ren
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Zimeng Xin
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Yaojun Luo
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Yushi Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Chaoqun Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Yuxi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Tongyudan Yang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China.
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China.
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Turner LA, Easton AA, Ferguson MM, Danzmann RG. Differences in gene expression between high and low tolerance rainbow trout (Oncorhynchus mykiss) to acute thermal stress. PLoS One 2025; 20:e0312694. [PMID: 39775350 PMCID: PMC11709236 DOI: 10.1371/journal.pone.0312694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 10/10/2024] [Indexed: 01/11/2025] Open
Abstract
Understanding the mechanisms that underlie the adaptive response of ectotherms to rising temperatures is key to mitigate the effects of climate change. We assessed the molecular and physiological processes that differentiate between rainbow trout (Oncorhynchus mykiss) with high and low tolerance to acute thermal stress. To achieve our goal, we used a critical thermal maximum trial in two strains of rainbow trout to elicit loss of equilibrium responses to identify high and low tolerance fish. We then compared the hepatic transcriptome profiles of high and low tolerance fish relative to untreated controls common to both strains to uncover patterns of differential gene expression and to gain a broad perspective on the interacting gene pathways and functional processes involved. We observed some of the classic responses to increased temperature (e.g., induction of heat shock proteins) but these responses were not the defining factors that differentiated high and low tolerance fish. Instead, high tolerance fish appeared to suppress growth-related functions, enhance certain autophagy components, better regulate neurodegenerative processes, and enhance stress-related protein synthesis, specifically spliceosomal complex activities, mRNA regulation, and protein processing through post-translational processes, relative to low tolerance fish. In contrast, low tolerance fish had higher transcript diversity and demonstrated elevated developmental, cytoskeletal, and morphogenic, as well as lipid and carbohydrate metabolic processes, relative to high tolerance fish. Our results suggest that high tolerance fish engaged in processes that supported the prevention of further damage by enhancing repair pathways, whereas low tolerance fish were more focused on replacing damaged cells and their structures.
Collapse
Affiliation(s)
- Leah A. Turner
- Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada
| | - Anne A. Easton
- Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada
- Ontario Aquaculture Research Centre, University of Guelph, Elora, Ontario, Canada
| | - Moira M. Ferguson
- Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada
| | - Roy G. Danzmann
- Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
5
|
Zhang L, Xiang Y, Cao C, Tan J, Li F, Yang X. Ciliary neurotrophic factor promotes the development of homocysteine-induced vascular endothelial injury through inflammation mediated by the JAK2/STAT3 signaling pathway. Exp Cell Res 2024; 440:114103. [PMID: 38848951 DOI: 10.1016/j.yexcr.2024.114103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/22/2024] [Accepted: 05/26/2024] [Indexed: 06/09/2024]
Abstract
Elevated homocysteine (Hcy) levels have been recognized as significant risk factor for cardiovascular and cerebrovascular diseases, closely related to endothelial injury. While expression of Ciliary Neurotrophic Factor (CNTF) significantly increases during Hcy-induced vascular endothelial cell injury, the precise molecular pathways through which CNTF operates remain to be clarified. To induce vascular endothelial cell injury, human umbilical vein endothelial cells (HUVECs) were treated with Hcy. Cell viability and apoptosis in HUVECs were assessed using the CCK-8 assay and flow cytometry. Western blot analysis determined the expression levels of the JAK2-STAT3 pathway, inflammation-related factors (IL-1β, NLRP3, ICAM-1, VCAM-1), and apoptosis-related factors (cleaved Caspase-3 and Bax). Immunofluorescence staining and western blotting were employed to examine CD31 and α-SMA expression. Knockdown of CNTF was achieved using lentiviral interference, and its effects on inflammation and cell injury were evaluated. Chromatin immunoprecipitation (ChIP) and dual luciferase reporter analysis were conducted to investigate the interaction between the MAFK and CNTF promoters. Our results indicated that Hcy induced high expression of CNTF and activated the JAK2-STAT3 signaling pathway, thereby upregulating factors associated with inflammation and cell apoptosis. Inhibiting CNTF alleviated Hcy-induced inflammation and cell injury. MAFK was identified as a transcription factor promoting CNTF transcription, and its overexpression exacerbated inflammation and cell injury in Hcy-treated HUVECs through the CNTF-JAK2-STAT3 axis, which could be reversed by knocking down CNTF. Activation of MAFK leads to CNTF upregulation, which activates the JAK2-STAT3 signaling pathway, regulating inflammation and inducing injury in Hcy-exposed vascular endothelial cells. Targeting CNTF or its upstream regulator MAFK may represent potential therapeutic strategies for mitigating endothelial dysfunction associated with hyperhomocysteinemia and cardiovascular diseases.
Collapse
Affiliation(s)
- Lijuan Zhang
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China
| | - Yan Xiang
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China; Ouyang Road Community Health Service Center, Hongkou District, Shanghai, China
| | - Chengxiu Cao
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China
| | - Jiaorong Tan
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China
| | - Fei Li
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China
| | - Xin Yang
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China.
| |
Collapse
|
6
|
Saha S. An Overview of Therapeutic Targeting of Nrf2 Signaling Pathway in Rheumatoid Arthritis. ACS OMEGA 2024; 9:10049-10057. [PMID: 38463248 PMCID: PMC10918843 DOI: 10.1021/acsomega.4c00163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 03/12/2024]
Abstract
Rheumatoid arthritis (RA), an autoimmune condition that has a significant inflammatory component and is exacerbated by dysregulated redox-dependent signaling pathways. In RA, the corelationship between oxidative stress and inflammation appears to be regulated by the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. Furthermore, it has been shown that transcriptional pathways involving Nrf2 and NFκB significantly interact under conditions of oxidative stress and inflammation. Because pathologic cells in RA have a higher chance of surviving, Nrf2's influence on concomitant pathologic mechanisms in the disease is explained by its interaction with key redox-sensitive inflammatory pathways. The current review not only updates knowledge about Nrf2's function in RA but also highlights the complex interactions between Nrf2 and other redox-sensitive transcription factors, which are essential to the self-sustaining inflammatory processes that define RA. This paper also reviews the candidates for treating RA through Nrf2 activation. Finally, future directions for pharmacologic Nrf2 activation in RA are suggested.
Collapse
Affiliation(s)
- Sarmistha Saha
- Department of Biotechnology,
Institute of Applied Sciences & Humanities, GLA University, Mathura 281406, Uttar Pradesh, India
| |
Collapse
|
7
|
Lu YY, Tsai HP, Tsai TH, Miao HC, Zhang ZH, Wu CH. RTA-408 Regulates p-NF-κB/TSLP/STAT5 Signaling to Ameliorate Nociceptive Hypersensitivity in Chronic Constriction Injury Rats. Mol Neurobiol 2024; 61:1714-1725. [PMID: 37773082 DOI: 10.1007/s12035-023-03660-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/15/2023] [Indexed: 09/30/2023]
Abstract
Neuropathic pain following nerve injury is a complex condition, which often puts a negative impact on life and remains a sustained problem. To make pain management better is of great significance and unmet need. RTA 408 (Omaveloxone) is a traditional Asian medicine with a valid anti-inflammatory property. Thus, we aim to investigate the therapeutic effect of RTA-408 on mechanical allodynia in chronic constriction injury (CCI) rats as well as the underlying mechanisms. Neuropathic pain was induced by using CCI of the rats' sciatic nerve (SN) and the behavior testing was measured by calibrated forceps testing. Activation of Nrf-2, the phosphorylation of nuclear factor-κB (NF-κB), and the inflammatory response were assessed by western blots. The number of apoptotic neurons and degree of glial cell reaction were examined by immunofluorescence assay. RTA-408 exerts an analgesic effect on CCI rats. RTA-408 reduces neuronal apoptosis and glial cell activation by increasing Nrf-2 expression and decreasing the inflammatory response (TNF-α/ p-NF-κB/ TSLP/ STAT5). These data suggest that RTA-408 is a candidate with potential to reduce nociceptive hypersensitivity after CCI by targeting TSLP/STAT5 signaling.
Collapse
Affiliation(s)
- Ying-Yi Lu
- Department of Dermatology, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
- Department of Post-Baccalaureate Medicine, School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
- Shu-Zen Junior College of Medicine and Management, Kaohsiung, 821, Taiwan
| | - Hung-Pei Tsai
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Tai-Hsin Tsai
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Hsiao-Chien Miao
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Zi-Hao Zhang
- Department of Neurosurgery, Xinle City Hospital, Xinle, Hebei, 050700, People's Republic of China
| | - Chieh-Hsin Wu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Center for Big Data Research, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
8
|
Liu CX, Guo XY, Zhou YB, Wang H. Therapeutic Role of Chinese Medicine Targeting Nrf2/HO-1 Signaling Pathway in Myocardial Ischemia/Reperfusion Injury. Chin J Integr Med 2024:10.1007/s11655-024-3657-0. [PMID: 38329655 DOI: 10.1007/s11655-024-3657-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2023] [Indexed: 02/09/2024]
Abstract
Acute myocardial infarction (AMI), characterized by high incidence and mortality rates, poses a significant public health threat. Reperfusion therapy, though the preferred treatment for AMI, often exacerbates cardiac damage, leading to myocardial ischemia/reperfusion injury (MI/RI). Consequently, the development of strategies to reduce MI/RI is an urgent priority in cardiovascular therapy. Chinese medicine, recognized for its multi-component, multi-pathway, and multi-target capabilities, provides a novel approach for alleviating MI/RI. A key area of interest is the nuclear factor E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. This pathway is instrumental in regulating inflammatory responses, oxidative stress, apoptosis, endoplasmic reticulum stress, and ferroptosis in MI/RI. This paper presents a comprehensive overview of the Nrf2/HO-1 signaling pathway's structure and its influence on MI/RI. Additionally, it reviews the latest research on leveraging Chinese medicine to modulate the Nrf2/HO-1 pathway in MI/RI treatment.
Collapse
Affiliation(s)
- Chang-Xing Liu
- First Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xin-Yi Guo
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610036, China
| | - Ya-Bin Zhou
- Department of Cardiology, the First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - He Wang
- Department of Cardiology, the First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
9
|
Lv P, Han P, Cui Y, Chen Q, Cao W. Quercetin attenuates inflammation in LPS-induced lung epithelial cells via the Nrf2 signaling pathway. Immun Inflamm Dis 2024; 12:e1185. [PMID: 38353312 PMCID: PMC10865417 DOI: 10.1002/iid3.1185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 09/28/2023] [Accepted: 01/23/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Pneumonia is the leading cause of death among children under five, and kill almost two million children each year. Quercetin, a flavonoid polyphenolic compound, exerts many beneficial biological activities, including anti-inflammatory functions. Our study aimed to investigate the possibility of quercetin as a therapeutic agent for pneumonia and its role in the inflammatory response induced by lipopolysaccharide (LPS). METHODS LPS induced human alveolar epithelial cell A549 as a lung inflammation model in vitro. The effects of quercetin on the production of cytokines and the expression of related-proteins were detected by Enzyme-Linked ImmunoSorbent Assay and Western Blot, respectively. Cell Counting Kit-8 assay was used to detect cell viability. flow cytometry was used to measure cell apoptosis. NO levels were also analyzed through NO kit. RESULTS Our results found that quercetin attenuated the release of IL-1β, IL-6, PGE2, and nitrite in LPS-induced A549 cells. In addition, quercetin inhibits cell apoptosis and relieves ROS generation in LPS-induced A549 cells. Quercetin also inhibits LPS-induced NF-κB activation. They have upregulated the expression of nuclear factor erythroid 2 (Nrf2) and HO-1. CONCLUSION In conclusion, these results suggested that quercetin attenuates LPS-induced inflammation in A549 by activating the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Pengju Lv
- Translational Medical CenterZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
| | - Pengli Han
- Translational Medical CenterZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
| | - Yuanbo Cui
- Translational Medical CenterZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
| | - Qiusheng Chen
- Department of Pulmonary and Critical Care MedicineZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
| | - Wei Cao
- Translational Medical CenterZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
10
|
Chen M, Zhang Y, Adams T, Ji D, Jiang W, Wain LV, Cho M, Kaminski N, Zhao H. Integrative analyses for the identification of idiopathic pulmonary fibrosis-associated genes and shared loci with other diseases. Thorax 2023; 78:792-798. [PMID: 36216496 PMCID: PMC10083187 DOI: 10.1136/thorax-2021-217703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/16/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND Although genome-wide association studies (GWAS) have identified many genomic regions associated with idiopathic pulmonary fibrosis (IPF), the causal genes and functions remain largely unknown. Many single-cell expression data have become available for IPF, and there is increasing evidence suggesting a shared genetic basis between IPF and other diseases. METHODS We conducted integrative analyses to improve the power of GWAS. First, we calculated global and local genetic correlations to identify IPF genetically associated traits and local regions. Then, we prioritised candidate genes contributing to local genetic correlation. Second, we performed transcriptome-wide association analysis (TWAS) of 44 tissues to identify candidate genes whose genetically predicted expression level is associated with IPF. To replicate our findings and investigate the regulatory role of the transcription factors (TF) in identified candidate genes, we first conducted the heritability enrichment analysis in TF binding sites. Then, we examined the enrichment of the TF target genes in cell-type-specific differentially expressed genes (DEGs) identified from single-cell expression data of IPF and healthy lung samples. FINDINGS We identified 12 candidate genes across 13 genomic regions using local genetic correlation, including the POT1 locus (p value=0.00041), which contained variants with protective effects on lung cancer but increasing IPF risk. We identified another 13 novel genes using TWAS. Two TFs, MAFK and SMAD2, showed significant enrichment in both partitioned heritability and cell-type-specific DEGs. INTERPRETATION Our integrative analysis identified new genes for IPF susceptibility and expanded the understanding of the complex genetic architecture and disease mechanism of IPF.
Collapse
Affiliation(s)
- Ming Chen
- Biostatistics, Yale University School of Public Health, New Haven, Connecticut, USA
| | - Yiliang Zhang
- Biostatistics, Yale University School of Public Health, New Haven, Connecticut, USA
| | - Taylor Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dingjue Ji
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, USA
| | - Wei Jiang
- Biostatistics, Yale University School of Public Health, New Haven, Connecticut, USA
| | - Louise V Wain
- National Institute for Health Research, Leicester Respiratory Biomedical Research Centre, Glenfield Hospital, Leicester, UK
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Michael Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Hongyu Zhao
- Biostatistics, Yale University School of Public Health, New Haven, Connecticut, USA
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
11
|
Duan X, Hu M, Yang L, Zhang S, Wang B, Li T, Tan Y, Li Y, Liu X, Zhan Z. IRG1 prevents excessive inflammatory responses and cardiac dysfunction after myocardial injury. Biochem Pharmacol 2023; 213:115614. [PMID: 37209857 DOI: 10.1016/j.bcp.2023.115614] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Acute myocardial infarction (MI) and chemotherapeutic drug administration can induce myocardial damage and cardiomyocyte cell death, and trigger the release of damage-associated molecular patterns (DAMPs) that initiate the aseptic inflammatory response. The moderate inflammatory response is beneficial for repairing damaged myocardium, while an excessive inflammatory response exacerbates myocardial injury, promotes scar formation, and results in a poor prognosis of cardiac diseases. Immune responsive gene 1 (IRG1) is specifically highly expressed in activated macrophages and mediates the production of tricarboxylic acid (TCA) cycle metabolite itaconate. However, the role of IRG1 in the inflammation and myocardial injury of cardiac stress-related diseases remains unknown. Here, we found that IRG1 knockout mice exhibited increased cardiac tissue inflammation and infarct size, aggravated myocardial fibrosis, and impaired cardiac function after MI and in vivo doxorubicin (Dox) administration. Mechanically, IRG1 deficiency enhanced the production of IL-6 and IL-1β by suppressing the nuclear factor red lineage 2-related factor 2 (NRF2) and activating transcription factor 3 (ATF3) pathway in cardiac macrophages. Importantly, 4-octyl itaconate (4-OI), a cell-permeable derivative of itaconate, reversed the inhibited expression of NRF2 and ATF3 caused by IRG1 deficiency. Moreover, in vivo 4-OI administration inhibited the cardiac inflammation and fibrosis, and prevented adverse ventricle remodeling in IRG1 knockout mice with MI or Dox-induced myocardial injury. Our study uncovers the critical protective role of IRG1 in suppressing inflammation and preventing cardiac dysfunction under ischemic or toxic injury conditions, providing a potential target for the treatment of myocardial injury.
Collapse
Affiliation(s)
- Xuewen Duan
- Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai 200081, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Meiling Hu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Linshan Yang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Sheng Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Bo Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Tong Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yong Tan
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yingke Li
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Xingguang Liu
- Department of Pathogen Biology, Naval Medical University, Shanghai 200433, China.
| | - Zhenzhen Zhan
- Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai 200081, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| |
Collapse
|
12
|
Peng S, Shen L, Yu X, Zhang L, Xu K, Xia Y, Zha L, Wu J, Luo H. The role of Nrf2 in the pathogenesis and treatment of ulcerative colitis. Front Immunol 2023; 14:1200111. [PMID: 37359553 PMCID: PMC10285877 DOI: 10.3389/fimmu.2023.1200111] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease involving mainly the colorectal mucosa and submucosa, the incidence of which has been on the rise in recent years. Nuclear factor erythroid 2-related factor 2 (Nrf2), known for its key function as a transcription factor, is pivotal in inducing antioxidant stress and regulating inflammatory responses. Numerous investigations have demonstrated the involvement of the Nrf2 pathway in maintaining the development and normal function of the intestine, the development of UC, and UC-related intestinal fibrosis and carcinogenesis; meanwhile, therapeutic agents targeting the Nrf2 pathway have been widely investigated. This paper reviews the research progress of the Nrf2 signaling pathway in UC.
Collapse
Affiliation(s)
- Shuai Peng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive Diseases, Wuhan, China
| | - Lei Shen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive Diseases, Wuhan, China
| | - Xiaoyun Yu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Xu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuan Xia
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive Diseases, Wuhan, China
| | - Lanlan Zha
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive Diseases, Wuhan, China
| | - Jing Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive Diseases, Wuhan, China
| | - Hesheng Luo
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive Diseases, Wuhan, China
| |
Collapse
|
13
|
Zhang J, Zhang T, Zeng S, Zhang X, Zhou F, Gillies MC, Zhu L. The Role of Nrf2/sMAF Signalling in Retina Ageing and Retinal Diseases. Biomedicines 2023; 11:1512. [PMID: 37371607 DOI: 10.3390/biomedicines11061512] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/10/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
Age-related diseases, such as Parkinson's disease, Alzheimer's disease, cardiovascular diseases, cancers, and age-related macular disease, have become increasingly prominent as the population ages. Oxygen is essential for living organisms, but it may also cause disease when it is transformed into reactive oxygen species via biological processes in cells. Most of the production of ROS occurs in mitochondrial complexes I and III. The accumulation of ROS in cells causes oxidative stress, which plays a crucial role in human ageing and many diseases. Nuclear factor-erythroid 2-related factor 2 (Nrf2) is a key antioxidant transcription factor that plays a central role in many diseases and ageing in general. It regulates many downstream antioxidative enzymes when cells are exposed to oxidative stress. A basic-region leucine zipper (bZIP) transcription factor, MAF, specifically the small MAF subfamily (sMAFs), forms heterodimers with Nrf2, which bind with Maf-recognition elements (MAREs) in response to oxidative stress. The role of this complex in the human retina remains unclear. This review summarises the current knowledge about Nrf2 and its downstream signalling, especially its cofactor-MAF, in ageing and diseases, with a focus on the retina. Since Nrf2 is the master regulator of redox homeostasis in cells, we hypothesise that targeting Nrf2 is a promising therapeutic approach for many age-related diseases.
Collapse
Affiliation(s)
- Jialing Zhang
- Save Sight Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Ting Zhang
- Save Sight Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Shaoxue Zeng
- Save Sight Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Xinyuan Zhang
- Department of Ocular Fundus Diseases, Beijing Tongren Eye Centre, Tongren Hospital, Capital Medical University, Beijing 100073, China
| | - Fanfan Zhou
- Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| | - Mark C Gillies
- Save Sight Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Ling Zhu
- Save Sight Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
14
|
Liu H, Yang R, Zhao S, Zhou F, Liu Y, Zhou Z, Chen L, Xie J. Collagen scaffolds derived from bovine skin loaded with MSC optimized M1 macrophages remodeling and chronic diabetic wounds healing. Bioeng Transl Med 2023; 8:e10467. [PMID: 37206210 PMCID: PMC10189465 DOI: 10.1002/btm2.10467] [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: 08/18/2022] [Revised: 10/25/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Owing to the persistent inflammatory microenvironment and unsubstantial dermal tissues, chronic diabetic wounds do not heal easily and their recurrence rate is high. Therefore, a dermal substitute that can induce rapid tissue regeneration and inhibit scar formation is urgently required to address this concern. In this study, we established biologically active dermal substitutes (BADS) by combining novel animal tissue-derived collagen dermal-replacement scaffolds (CDRS) and bone marrow mesenchymal stem cells (BMSCs) for the healing and recurrence treatments of chronic diabetic wounds. The collagen scaffolds derived from bovine skin (CBS) displayed good physicochemical properties and superior biocompatibility. CBS loaded with BMSCs (CBS-MCSs) could inhibit M1 macrophage polarization in vitro. Decreased MMP-9 and increased Col3 at the protein level were detected in CBS-MSCs-treated M1 macrophages, which may be attributed to the suppression of the TNF-α/NF-κB signaling pathway (downregulating phospho-IKKα/β/total IKKα/β, phospho-IκB/total IκB, and phospho-NFκB/total NFκB) in M1 macrophages. Moreover, CBS-MSCs could benefit the transformation of M1 (downregulating iNOS) to M2 (upregulating CD206) macrophages. Wound-healing evaluations demonstrated that CBS-MSCs regulated the polarization of macrophages and the balance of inflammatory factors (pro-inflammatory: IL-1β, TNF-α, and MMP-9; anti-inflammatory: IL-10 and TGF-β3) in db/db mice. Furthermore, CBS-MSCs facilitated the noncontractile and re-epithelialized processes, granulation tissue regeneration, and neovascularization of chronic diabetic wounds. Thus, CBS-MSCs have a potential value for clinical application in promoting the healing of chronic diabetic wounds and preventing the recurrence of ulcers.
Collapse
Affiliation(s)
- Hengdeng Liu
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Ronghua Yang
- Department of Burn and Plastic SurgeryGuangzhou First People's Hospital, South China University of TechnologyGuangzhouGuangdongChina
| | - Shixin Zhao
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Fei Zhou
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Yiling Liu
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Ziheng Zhou
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Lei Chen
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Julin Xie
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
15
|
Lin XL, Shi YN, Cao YL, Tan X, Zeng YL, Luo ST, Li YM, Qin L, Xia BH, Fu RG, Lin LM, Li K, Cao D, Zeng JG, Liao DF. Sanguinarine protects against indomethacin-induced small intestine injury in rats by regulating the Nrf2/NF-κB pathways. Front Pharmacol 2022; 13:960140. [PMID: 36304153 PMCID: PMC9593053 DOI: 10.3389/fphar.2022.960140] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/27/2022] [Indexed: 12/04/2022] Open
Abstract
In recent years, small intestine as a key target in the treatment of Inflammatory bowel disease caused by NSAIDs has become a hot topic. Sanguinarine (SA) is one of the main alkaloids in the Macleaya cordata extracts with strong pharmacological activity of anti-tumor, anti-inflammation and anti-oxidant. SA is reported to inhibit acetic acid-induced colitis, but it is unknown whether SA can relieve NSAIDs-induced small intestinal inflammation. Herein, we report that SA effectively reversed the inflammatory lesions induced by indomethacin (Indo) in rat small intestine and IEC-6 cells in culture. Our results showed that SA significantly relieved the symptoms and reversed the inflammatory lesions of Indo as shown in alleviation of inflammation and improvement of colon macroscopic damage index (CMDI) and tissue damage index (TDI) scores. SA decreased the levels of TNF-α, IL-6, IL-1β, MDA and LDH in small intestinal tissues and IEC-6 cells, but increased SOD activity and ZO-1 expression. Mechanistically, SA dose-dependently promoted the expression of Nrf2 and HO-1 by decreasing Keap-1 level, but inhibited p65 phosphorylation and nuclear translocation in Indo-treated rat small intestine and IEC-6 cells. Furthermore, in SA treated cells, the colocalization between p-p65 and CBP in the nucleus was decreased, while the colocalization between Nrf2 and CBP was increased, leading to the movement of gene expression in the nucleus to the direction of anti-inflammation and anti-oxidation. Nrf2 silencing blocked the effects of SA. Together our results suggest that SA can significantly prevent intestinal inflammatory lesions induced by Indo in rats and IEC-6 cells through regulation of the Nrf2 pathway and NF-κBp65 pathway.
Collapse
Affiliation(s)
- Xiu-lian Lin
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ya-ning Shi
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yu-ling Cao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xi Tan
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ya-ling Zeng
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Shi-teng Luo
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ya-mei Li
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Li Qin
- Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Bo-hou Xia
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Rong-geng Fu
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Li-mei Lin
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Kai Li
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Deliang Cao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
- *Correspondence: Deliang Cao, ; Jian-guo Zeng, ; Duan-fang Liao,
| | - Jian-guo Zeng
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, China
- *Correspondence: Deliang Cao, ; Jian-guo Zeng, ; Duan-fang Liao,
| | - Duan-fang Liao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
- *Correspondence: Deliang Cao, ; Jian-guo Zeng, ; Duan-fang Liao,
| |
Collapse
|
16
|
Loan JJM, Al-Shahi Salman R, McColl BW, Hardingham GE. Activation of Nrf2 to Optimise Immune Responses to Intracerebral Haemorrhage. Biomolecules 2022; 12:1438. [PMID: 36291647 PMCID: PMC9599325 DOI: 10.3390/biom12101438] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022] Open
Abstract
Haemorrhage into the brain parenchyma can be devastating. This manifests as spontaneous intracerebral haemorrhage (ICH) after head trauma, and in the context of vascular dementia. Randomised controlled trials have not reliably shown that haemostatic treatments aimed at limiting ICH haematoma expansion and surgical approaches to reducing haematoma volume are effective. Consequently, treatments to modulate the pathophysiological responses to ICH, which may cause secondary brain injury, are appealing. Following ICH, microglia and monocyte derived cells are recruited to the peri-haematomal environment where they phagocytose haematoma breakdown products and secrete inflammatory cytokines, which may trigger both protective and harmful responses. The transcription factor Nrf2, is activated by oxidative stress, is highly expressed by central nervous system microglia and macroglia. When active, Nrf2 induces a transcriptional programme characterised by increased expression of antioxidant, haem and heavy metal detoxification and proteostasis genes, as well as suppression of proinflammatory factors. Therefore, Nrf2 activation may facilitate adaptive-protective immune cell responses to ICH by boosting resistance to oxidative stress and heavy metal toxicity, whilst limiting harmful inflammatory signalling, which can contribute to further blood brain barrier dysfunction and cerebral oedema. In this review, we consider the responses of immune cells to ICH and how these might be modulated by Nrf2 activation. Finally, we propose potential therapeutic strategies to harness Nrf2 to improve the outcomes of patients with ICH.
Collapse
Affiliation(s)
- James J. M. Loan
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
- UK Dementia Research Institute at Edinburgh, University of Edinburgh, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | | | - Barry W. McColl
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
- UK Dementia Research Institute at Edinburgh, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Giles E. Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
- UK Dementia Research Institute at Edinburgh, University of Edinburgh, Edinburgh EH16 4SB, UK
| |
Collapse
|
17
|
Manda G, Milanesi E, Genc S, Niculite CM, Neagoe IV, Tastan B, Dragnea EM, Cuadrado A. Pros and cons of NRF2 activation as adjunctive therapy in rheumatoid arthritis. Free Radic Biol Med 2022; 190:179-201. [PMID: 35964840 DOI: 10.1016/j.freeradbiomed.2022.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 10/15/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease with an important inflammatory component accompanied by deregulated redox-dependent signaling pathways that are feeding back into inflammation. In this context, we bring into focus the transcription factor NRF2, a master redox regulator that exerts exquisite antioxidant and anti-inflammatory effects. The review does not intend to be exhaustive, but to point out arguments sustaining the rationale for applying an NRF2-directed co-treatment in RA as well as its potential limitations. The involvement of NRF2 in RA is emphasized through an analysis of publicly available transcriptomic data on NRF2 target genes and the findings from NRF2-knockout mice. The impact of NRF2 on concurrent pathologic mechanisms in RA is explained by its crosstalk with major redox-sensitive inflammatory and cell death-related pathways, in the context of the increased survival of pathologic cells in RA. The proposed adjunctive therapy targeted to NRF2 is further sustained by the existence of promising NRF2 activators that are in various stages of drug development. The interference of NRF2 with conventional anti-rheumatic therapies is discussed, including the cytoprotective effects of NRF2 for alleviating drug toxicity. From another perspective, the review presents how NRF2 activation would be decreasing the efficacy of synthetic anti-rheumatic drugs by increasing drug efflux. Future perspectives regarding pharmacologic NRF2 activation in RA are finally proposed.
Collapse
Affiliation(s)
- Gina Manda
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Elena Milanesi
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Sermin Genc
- Neurodegeneration and Neuroprotection Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey; Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Izmir, Turkey
| | - Cristina Mariana Niculite
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania; Department of Cellular and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Ionela Victoria Neagoe
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Bora Tastan
- Neurodegeneration and Neuroprotection Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Elena Mihaela Dragnea
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
18
|
Wang L, He C. Nrf2-mediated anti-inflammatory polarization of macrophages as therapeutic targets for osteoarthritis. Front Immunol 2022; 13:967193. [PMID: 36032081 PMCID: PMC9411667 DOI: 10.3389/fimmu.2022.967193] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 07/27/2022] [Indexed: 12/14/2022] Open
Abstract
Macrophages are the most abundant immune cells within the synovial joints, and also the main innate immune effector cells triggering the initial inflammatory responses in the pathological process of osteoarthritis (OA). The transition of synovial macrophages between pro-inflammatory and anti-inflammatory phenotypes can play a key role in building the intra-articular microenvironment. The pro-inflammatory cascade induced by TNF-α, IL-1β, and IL-6 is closely related to M1 macrophages, resulting in the production of pro-chondrolytic mediators. However, IL-10, IL1RA, CCL-18, IGF, and TGF are closely related to M2 macrophages, leading to the protection of cartilage and the promoted regeneration. The inhibition of NF-κB signaling pathway is central in OA treatment via controlling inflammatory responses in macrophages, while the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway appears not to attract widespread attention in the field. Nrf2 is a transcription factor encoding a large number of antioxidant enzymes. The activation of Nrf2 can have antioxidant and anti-inflammatory effects, which can also have complex crosstalk with NF-κB signaling pathway. The activation of Nrf2 can inhibit the M1 polarization and promote the M2 polarization through potential signaling transductions including TGF-β/SMAD, TLR/NF-κB, and JAK/STAT signaling pathways, with the regulation or cooperation of Notch, NLRP3, PI3K/Akt, and MAPK signaling. And the expression of heme oxygenase-1 (HO-1) and the negative regulation of Nrf2 for NF-κB can be the main mechanisms for promotion. Furthermore, the candidates of OA treatment by activating Nrf2 to promote M2 phenotype macrophages in OA are also reviewed in this work, such as itaconate and fumarate derivatives, curcumin, quercetin, melatonin, mesenchymal stem cells, and low-intensity pulsed ultrasound.
Collapse
Affiliation(s)
- Lin Wang
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chengqi He
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Chengqi He,
| |
Collapse
|
19
|
Majkutewicz I. Dimethyl fumarate: A review of preclinical efficacy in models of neurodegenerative diseases. Eur J Pharmacol 2022; 926:175025. [DOI: 10.1016/j.ejphar.2022.175025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/27/2022] [Accepted: 05/09/2022] [Indexed: 11/03/2022]
|
20
|
Sun W, Choi HS, Kim CS, Bae EH, Ma SK, Kim SW. Maslinic Acid Attenuates Ischemia/Reperfusion-Induced Acute Kidney Injury by Suppressing Inflammation and Apoptosis Through Inhibiting NF-κB and MAPK Signaling Pathway. Front Pharmacol 2022; 13:807452. [PMID: 35496304 PMCID: PMC9039024 DOI: 10.3389/fphar.2022.807452] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/03/2022] [Indexed: 12/14/2022] Open
Abstract
Inflammation and apoptosis are the major contributors to the mechanisms of acute kidney injury (AKI) due to renal ischemia-reperfusion injury (IRI). Maslinic acid (MA), a pentacyclic triterpene acid mostly found in dietary plants, the current study was to demonstrate the renoprotective effect of MA on IRI-induced AKI, and to investigate the role of inflammation and apoptosis-related signaling pathways as a molecular mechanism. C57BL/6J mice were subjected to IRI for 72 h, and MA was daily administered by intraperitoneal injection during this period. In parallel, rat renal proximal tubule cells (NRK52E) were prophylactically treated with MA and then exposed to hydrogen peroxide (H2O2). MA treatment significantly inhibited the mRNA expression of interleukin (IL-1β), tumor necrosis factor-α (TGF-α), monocyte chemoattractant protein-1 (MCP-1), and intercellular adhesion molecule-1(ICAM-1). Also, MA reduced the expression of Bax/Bcl2 ratio and cleaved caspase-3. In NRK52 cells, MA inhibited the IκBα degradation, blocked NF-κB/p65 phosphorylation, and nuclear translocation. The phosphorylation of ERK, JNK, and p38 was attenuated by MA in IRI-induced kidney injury and H2O2-stimulated NRK52 cells. The expression levels of IL-1β, MCP-1, and ICAM-1 were upregulated in H2O2-stimulated NRK52E cells, which was attenuated by NF-κB inhibitor. H2O2 treatment increased the Bax/Bcl2 ratio and cleaved caspase-3 in NRK52E cells, which was counteracted by MAPK inhibitors. Together, our data demonstrate that MA suppresses IR-induced AKI injury through NF-κB and MAPK signaling pathways and that MA is a promising agent in the treatment of kidney diseases.
Collapse
|
21
|
Tastan B, Arioz BI, Genc S. Targeting NLRP3 Inflammasome With Nrf2 Inducers in Central Nervous System Disorders. Front Immunol 2022; 13:865772. [PMID: 35418995 PMCID: PMC8995746 DOI: 10.3389/fimmu.2022.865772] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/04/2022] [Indexed: 12/15/2022] Open
Abstract
The NLRP3 inflammasome is an intracellular multiprotein complex that plays an essential role in the innate immune system by identifying and eliminating a plethora of endogenous and exogenous threats to the host. Upon activation of the NLRP3 complex, pro-inflammatory cytokines are processed and released. Furthermore, activation of the NLRP3 inflammasome complex can induce pyroptotic cell death, thereby propagating the inflammatory response. The aberrant activity and detrimental effects of NLRP3 inflammasome activation have been associated with cardiovascular, neurodegenerative, metabolic, and inflammatory diseases. Therefore, clinical strategies targeting the inhibition of the self-propelled NLRP3 inflammasome activation are required. The transcription factor Nrf2 regulates cellular stress response, controlling the redox equilibrium, metabolic programming, and inflammation. The Nrf2 pathway participates in anti-oxidative, cytoprotective, and anti-inflammatory activities. This prominent regulator, through pharmacologic activation, could provide a therapeutic strategy for the diseases to the etiology and pathogenesis of which NLRP3 inflammasome contributes. In this review, current knowledge on NLRP3 inflammasome activation and Nrf2 pathways is presented; the relationship between NLRP3 inflammasome signaling and Nrf2 pathway, as well as the pre/clinical use of Nrf2 activators against NLRP3 inflammasome activation in disorders of the central nervous system, are thoroughly described. Cumulative evidence points out therapeutic use of Nrf2 activators against NLRP3 inflammasome activation or diseases that NLRP3 inflammasome contributes to would be advantageous to prevent inflammatory conditions; however, the side effects of these molecules should be kept in mind before applying them to clinical practice.
Collapse
Affiliation(s)
- Bora Tastan
- Genc Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey,Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Burak I. Arioz
- Genc Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey,Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Sermin Genc
- Genc Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey,Department of Neuroscience, Health Sciences Institute, Dokuz Eylul University, Izmir, Turkey,*Correspondence: Sermin Genc,
| |
Collapse
|
22
|
MafK accelerates Salmonella mucosal infection through caspase-3 activation. Aging (Albany NY) 2022; 14:2287-2303. [PMID: 35260530 PMCID: PMC8954977 DOI: 10.18632/aging.203938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 02/28/2022] [Indexed: 11/25/2022]
Abstract
Gastrointestinal homeostasis is critical for maintaining host health, and is affected by many factors. A recent report showed that Musculoaponeurotic fibrosarcoma K (MafK) expression is increased in patients that have ulcerative colitis (UC). Even so, MafK’s significance in sustaining intestinal homeostasis has not been investigated. In this research, MafK overexpressing transgenic (MafK Tg) mice were found to be more susceptible to infection with Salmonella on the mucosa than the wild-type (WT) mice. Following Salmonella oral infection, MafK Tg mice suffered higher mortality and a lot more weight loss, damage to the intestines, and inflammation in the intestines than WT mice. MafK Tg mice were also unable to control Salmonella colonization and dissemination. In vivo data showed that increased MafK expression promoted epithelial cell apoptosis which was further confirmed by in vitro data. The rapid cleavage of caspase-3 in epithelial cells contributed to Salmonella dissemination and inflammation initiation. This study reveals that MafK participates in Salmonella pathogenesis acceleration by increasing caspase-3 activation.
Collapse
|
23
|
The Potential of Nrf2 Activation as a Therapeutic Target in Systemic Lupus Erythematosus. Metabolites 2022; 12:metabo12020151. [PMID: 35208225 PMCID: PMC8876688 DOI: 10.3390/metabo12020151] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/03/2022] [Accepted: 02/03/2022] [Indexed: 02/06/2023] Open
Abstract
Inflammation and oxidative stress are well established in systemic lupus erythematosus (SLE) and are critical to the pathogenesis of autoimmune diseases. The transcription factor NF-E2 related factor 2 (Nrf2) is a central regulator of cellular anti-oxidative responses, inflammation, and restoration of redox balance. Accumulating reports support an emerging role for the regulation of Nrf2 in SLE. These include findings on the development of lupus-like autoimmune nephritis and altered immune cell populations in mice lacking Nrf2, as well as decreased Nrf2 abundance in the dendritic cells of patients with SLE. Nrf2-inducing agents have been shown to alleviate oxidative and inflammatory stress and reduce tissue injury in SLE mouse models. Since Nrf2 expression can be increased in activated T cells, the precise role of Nrf2 activation in different immune cell types and their function remains to be defined. However, targeting Nrf2 for the treatment of diseases associated with oxidative stress and inflammation, such as SLE, is promising. As investigation of Nrf2-inducing agents in clinical trials grows, defining the signaling and molecular mechanisms of action and downstream effects in response to different Nrf2-inducing agents in specific cells, tissues, and diseases, will be critical for effective clinical use.
Collapse
|
24
|
Albert-Garay JS, Riesgo-Escovar JR, Salceda R. High glucose concentrations induce oxidative stress by inhibiting Nrf2 expression in rat Müller retinal cells in vitro. Sci Rep 2022; 12:1261. [PMID: 35075205 PMCID: PMC8975969 DOI: 10.1038/s41598-022-05284-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/31/2021] [Indexed: 12/11/2022] Open
Abstract
Diabetic retinopathy (DR) is a complication of diabetes. Several studies have implicated oxidative stress as a fundamental factor in the progression of the disease. The nuclear factor erythroid-2-related factor 2 (Nrf2) is one of the main regulators of redox homeostasis. Glia Müller cells (MC) maintain the structural and functional stability of the retina. The objective of this study was to evaluate the effect of high glucose concentrations on reactive oxygen species (ROS) production and Nrf2 expression levels in rat MC. MC were incubated with normal (NG; 5 mM) or high glucose (HG; 25 mM) for different times. Incubation with HG increased ROS levels from 12 to 48 h but did not affect cell viability. However, exposure to 3 h of HG caused a transient decrease Nrf2 levels. At that time, we also observed a decrease in the mRNA expression of Nrf2 target genes, glutathione levels, and catalase activity, all of which increased significantly beyond initial levels after 48 h of incubation. HG exposure leads to an increase in the p65 subunit of nuclear factor-κB (NF-kB) levels, and its target genes. These results suggest that high glucose concentrations lead to alteration of the redox regulatory capacity of Nrf2 mediated by NF-kB regulation.
Collapse
Affiliation(s)
- Jesús Silvestre Albert-Garay
- Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, CDMX, Mexico.
| | - Juan Rafael Riesgo-Escovar
- Instituto de Neurobiología, Campus UNAM Juriquilla, Universidad Nacional Autónoma de México, 76226, Querétaro, Mexico
| | - Rocío Salceda
- Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, CDMX, Mexico
| |
Collapse
|
25
|
Hu X, Liu Z, Lu Y, Chi X, Han K, Wang H, Wang Y, Ma L, Xu B. Glucose metabolism enhancement by 10-hydroxy-2-decenoic acid via the PI3K/AKT signaling pathway in high-fat-diet/streptozotocin induced type 2 diabetic mice. Food Funct 2022; 13:9931-9946. [DOI: 10.1039/d1fo03818d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Here, we used high fat diet (HFD) combined with streptozotocin (STZ) injection to establish a diabetes model, with the aim of exploring the hypoglycemic effects of 10-hydroxy-2-decenoic acid (10-HDA), and...
Collapse
|
26
|
Petpiroon N, Rosena A, Pimtong W, Charoenlappanit S, Koobkokkruad T, Roytrakul S, Aueviriyavit S. Protective effects of Thai silk sericins and their related mechanisms on UVA-induced phototoxicity and melanogenesis: Investigation in primary melanocyte cells using a proteomic approach. Int J Biol Macromol 2021; 201:75-84. [PMID: 34968545 DOI: 10.1016/j.ijbiomac.2021.12.061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 11/20/2021] [Accepted: 12/09/2021] [Indexed: 11/05/2022]
Abstract
UV radiation causes excess production of melanin as a result of hyperpigmentation and skin disorders. Silk sericin exhibited bioactivities to skin and inhibited UV-induced phototoxicity and melanogenesis in skin cells; however, the mechanism related to sericin against UV-induced melanogenesis has not been investigated. This study aimed to investigate the protective effects of Thai silk sericins against UVA-induced phototoxicity and melanogenesis and their related mechanisms. Thai silk sericins exhibited cytoprotective effects against UV-induced toxicity in human primary melanocytes by attenuation of cytotoxicity, intracellular ROS generation, and mitochondrial potential impairment. Pre- and post-treatment with sericin significantly inhibited melanin synthesis and tyrosinase activity against UVA exposure. In addition, sericin S2 could reduce the basal melanin content in zebrafish embryos. The proteomic analysis demonstrated that Thai silk sericins altered the protein expression in melanocytes especially proteins related to stress, inflammatory, cytokine stimulation, cell proliferation, and cell survival processes that contribute to cytoprotective effect and inhibitory effect on melanogenesis of sericin. Moreover, we demonstrated the novel mechanism of Thai silk sericins in inhibiting UVA-induced melanogenesis via increasing BMP4 expression in MAPK/ERK signaling pathway. These evidences support the potential use of Thai silk sericins in prevention of hyperpigmentation in skin disorders especially after UVA exposure.
Collapse
Affiliation(s)
- Nalinrat Petpiroon
- National Nanotechnology Center, National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Apiwan Rosena
- National Nanotechnology Center, National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Wittaya Pimtong
- National Nanotechnology Center, National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Sawanya Charoenlappanit
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Thongchai Koobkokkruad
- National Nanotechnology Center, National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Sasitorn Aueviriyavit
- National Nanotechnology Center, National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand.
| |
Collapse
|
27
|
Cong P, Wang T, Tong C, Liu Y, Shi L, Mao S, Shi X, Jin H, Liu Y, Hou M. Resveratrol ameliorates thoracic blast exposure-induced inflammation, endoplasmic reticulum stress and apoptosis in the brain through the Nrf2/Keap1 and NF-κB signaling pathway. Injury 2021; 52:2795-2802. [PMID: 34454721 DOI: 10.1016/j.injury.2021.08.019] [Citation(s) in RCA: 19] [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: 03/29/2021] [Revised: 08/04/2021] [Accepted: 08/13/2021] [Indexed: 02/02/2023]
Abstract
Blast injuries include the various types of internal and external trauma caused by the impact force of high-speed blast waves with multiple mechanisms involved. Thoracic blast exposure could induce neurotrauma as well, but effective therapies are lacking. Resveratrol is a polyphenol flavonoid secreted by plants and has been shown to provide cardiovascular protection and play anti-inflammatory, anti-oxidation and anti-cancer roles. However, the effects of resveratrol on thoracic blast exposure-induced brain injury have not been investigated. To explore this, a mouse model of thoracic blast exposure-induced brain injury was established. Sixty C57BL/6 wild type mice were randomly divided equally into four groups (one control group, one model group, and model groups with 25 or 50 mg/kg resveratrol injected intraperitoneally). As traumatic brain injury often accompanied by mental symptoms, cognitive dysfunction and anxious behavior were evaluated by Y maze, elevated plus maze and open field test. We also examined the mice for histopathological changes by hematoxylin-eosin staining; the expressions of inflammatory-related factors by ELISA; endoplasmic reticulum stress in brain tissue via the generation of reactive oxygen species (ROS) and the expressions of inositol-requiring enzyme-α (IRE-α) and C/EBP homologous protein (CHOP); apoptosis by measuring levels of Bax, p53 and Bcl-2. In addition, proteins of related pathways were also studied by western blotting. We found that resveratrol significantly reduced the levels of inflammatory-related factors, including interleukin (IL)-1β, IL-4, and high mobility group box protein 1(HMGB1), and increased the level of anti-inflammatory-related factor, IL-10, under thoracic blast exposure (P < 0.05). Cognitive dysfunction and anxious behavior were also ameliorated by resveratrol. In brain tissue, resveratrol significantly attenuated thoracic blast exposure-induced generation of ROS and expressions of IRE-α and CHOP, lowered the expressions of Bax and p53, and maintained Bcl-2 expression (P < 0.05). Additionally, resveratrol significantly ameliorated thoracic blast exposure-induced increases of Kelch-like ECH-associated protein 1 (Keap1) and nuclear factor (NF)-κB and the decrease in nuclear factor erythroid 2-related factor 2(Nrf2) expression in the brain (P < 0.05). Our results indicate that resveratrol has a protective effect on thoracic blast exposure-induced brain injury that is likely mediated through the Nrf2/Keap1 and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Peifang Cong
- College of Medicine and Biological Information Engineering, Northeastern University, No. 195, Chuangxin Road, Hunnan District, Shenyang, Liaoning Province, 110016, China.
| | - Teng Wang
- Jining No.1 people's Hospital of Shandong Province, No. 6, Jiankang Road, Jining, Shandong Province, 272011, China.
| | - Changci Tong
- Emergency Medicine Department of General Hospital of Northern theatre command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning Province, 110016, China.
| | - Ying Liu
- Emergency Medicine Department of General Hospital of Northern theatre command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning Province, 110016, China.
| | - Lin Shi
- Emergency Medicine Department of General Hospital of Northern theatre command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning Province, 110016, China.
| | - Shun Mao
- Emergency Medicine Department of General Hospital of Northern theatre command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning Province, 110016, China.
| | - Xiuyun Shi
- Emergency Medicine Department of General Hospital of Northern theatre command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning Province, 110016, China.
| | - Hongxu Jin
- Emergency Medicine Department of General Hospital of Northern theatre command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning Province, 110016, China.
| | - Yunen Liu
- Shenyang Medical College, No. 146, Huanghe North Street, Huanggu District, Shenyang, Liaoning Province, 110034, China; Emergency Medicine Department of General Hospital of Northern theatre command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning Province, 110016, China.
| | - Mingxiao Hou
- College of Medicine and Biological Information Engineering, Northeastern University, No. 195, Chuangxin Road, Hunnan District, Shenyang, Liaoning Province, 110016, China; Shenyang Medical College, No. 146, Huanghe North Street, Huanggu District, Shenyang, Liaoning Province, 110034, China; The Second Affiliated Hospital of Shenyang Medical College. The Veterans General Hospital of Liaoning Province, No.20 Beijiu Road, Heping District, Shenyang, Liaoning Province, 110001, China.
| |
Collapse
|
28
|
Fu J, Ni C, Ni H, Xu L, He Q, Pan H, Huang D, Sun Y, Luo G, Liu M, Yao M. Spinal Nrf2 translocation may inhibit neuronal NF-κB activation and alleviate allodynia in a rat model of bone cancer pain. J Neurochem 2021; 158:1110-1130. [PMID: 34254317 PMCID: PMC9292887 DOI: 10.1111/jnc.15468] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/23/2021] [Accepted: 07/02/2021] [Indexed: 01/11/2023]
Abstract
Bone cancer pain (BCP) is a clinical pathology that urgently needs to be solved, but research on the mechanism of BCP has so far achieved limited success. Nuclear factor erythroid 2 (NFE2)-related factor 2 (Nrf2) has been shown to be involved in pain, but its involvement in BCP and the specific mechanism have yet to be examined. This study aimed to test the hypothesis that BCP induces the transfer of Nrf2 from the cytoplasm to the nucleus and further promotes nuclear transcription to activate heme oxygenase-1 (HO-1) and inhibit the activation of nuclear factor-kappa B (NF-κB) signalling, ultimately regulating the neuroinflammatory response. Von-Frey was used for behavioural analysis in rats with BCP, whereas western blotting, real-time quantitative PCR (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) were used to detect molecular expression changes, and immunofluorescence was used to detect cellular localization. We demonstrated that BCP induced increased Nrf2 nuclear protein expression with decreased cytoplasmic protein expression in the spinal cord. Further increases in Nrf2 nuclear protein expression can alleviate hyperalgesia and activate HO-1 to inhibit the expression of NF-κB nuclear protein and inflammatory factors. Strikingly, intrathecal administration of the corresponding siRNA reversed the above effects. In addition, the results of double immune labelling revealed that Nrf2 and NF-κB were coexpressed in spinal cord neurons of rats with BCP. In summary, these findings suggest that the entry of Nrf2 into the nucleus promotes the expression of HO-1, inhibiting activation of the NF-κB signalling pathway, reducing neuroinflammation and ultimately exerting an anti-nociceptive effect.
Collapse
Affiliation(s)
- Jie Fu
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Chaobo Ni
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Hua‐Dong Ni
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Long‐Sheng Xu
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Qiu‐Li He
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Huan Pan
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Dong‐Dong Huang
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Yan‐Bao Sun
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Ge Luo
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Ming‐Juan Liu
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Ming Yao
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| |
Collapse
|
29
|
Hayashi T, Shibata T, Nakamura M, Sakurai N, Takano H, Ota M, Nomura-Horita T, Hayashi R, Shimasaki T, Ostuka T, Tahara T, Arisawa T. MAFK Polymorphisms Located in 3'-UTR are Associated with Severity of Atrophy and CDKN2A Methylation Status in the Gastric Mucosa. Genet Test Mol Biomarkers 2021; 25:255-262. [PMID: 33877894 DOI: 10.1089/gtmb.2020.0299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Objective: This study aimed to clarify the association of MAFK polymorphisms (rs4268033, rs3735656, and rs10226620) with the degree of gastric mucosal atrophy and CDKN2A CpG methylation status. Methods: A total of 491 subjects were enrolled in this study. Genotypes and methylation status were determined by polymerase chain reaction (PCR)-single-stranded conformation polymorphism and methylation-specific PCR (Fujita Health University, HM18-094). Methods: A total of 491 subjects were enrolled in this study. Genotypes and methylation status were determined by polymerase chain reaction (PCR)-single-stranded conformation polymorphism and methylation-specific PCR (Fujita Health University, HM18-094). Results: Either rs3735656 or rs10226620, located in the 3'-UTR of MAFK, was significantly associated with the severity of gastric mucosal atrophy using a dominant genetic model (odds ratio [OR], 2.10; p = 0.0012, and OR, 1.98; p = 0.0027, respectively). However, using a recessive genetic model, no significant association was found between three polymorphisms and gastric mucosal atrophy. The serum pepsinogen I/II ratio was significantly lower in subjects with minor alleles of rs3735656 and rs10226620 than in subjects with the wild homozygous allele (p = 0.018 and 0.013, respectively). In a subgroup including 400 of the 491 subjects, the CpG of p14ARF and p16 INK4a were methylated in 132 and 112 subjects, respectively. Fifty subjects had both CpG methylations and 206 subjects had neither methylation. When comparing the groups with both and neither methylations, there were no significant associations between three polymorphisms and CDKN2A methylation using a dominant genetic model. However, all polymorphisms investigated in this study (rs4268033, rs3735656, and rs10226620) were significantly associated with CDKN2A methylation in a recessive genetic model (OR, 3.58; p = 0.0071, OR, 4.49; p = 0.0004, and OR, 3.45; p = 0.0027, respectively). Conclusions: Our results indicate that carrying the minor allele of the MAFK polymorphisms, particularly when they are located in the 3'-UTR, has a high risk for the severity of gastric mucosal atrophy; furthermore, CDKN2A CpG methylation may develop in subjects with homozygous minor allele of these polymorphisms.
Collapse
Affiliation(s)
- Tasuku Hayashi
- Department of Gastroenterology, Kanazawa Medical University, Uchinada-machi, Japan
| | - Tomoyuki Shibata
- Department of Gastroenterology, Fujita Health University, Kutsukake-cho, Japan
| | - Masakatsu Nakamura
- Department of Gastroenterology, Kanazawa Medical University, Uchinada-machi, Japan
| | - Naoko Sakurai
- Department of Gastroenterology, Kanazawa Medical University, Uchinada-machi, Japan
| | - Hikaru Takano
- Department of Gastroenterology, Kanazawa Medical University, Uchinada-machi, Japan
| | - Masafumi Ota
- Department of Gastroenterology, Kanazawa Medical University, Uchinada-machi, Japan
| | - Tomoe Nomura-Horita
- Department of Gastroenterology, Kanazawa Medical University, Uchinada-machi, Japan
| | - Ranji Hayashi
- Department of Gastroenterology, Kanazawa Medical University, Uchinada-machi, Japan
| | - Takeo Shimasaki
- Department of Gastroenterology, Kanazawa Medical University, Uchinada-machi, Japan
| | - Toshimi Ostuka
- Department of Gastroenterology, Kanazawa Medical University, Uchinada-machi, Japan
| | - Tomomitsu Tahara
- Department of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - Tomiyasu Arisawa
- Department of Gastroenterology, Kanazawa Medical University, Uchinada-machi, Japan
| |
Collapse
|
30
|
Feng JH, Dong XW, Yu HL, Shen W, Lv XY, Wang R, Cheng XX, Xiong F, Hu XL, Wang H. Cynaroside protects the blue light-induced retinal degeneration through alleviating apoptosis and inducing autophagy in vitro and in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 88:153604. [PMID: 34130054 DOI: 10.1016/j.phymed.2021.153604] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/08/2021] [Accepted: 05/16/2021] [Indexed: 05/10/2023]
Abstract
BACKGROUND Blue light can directly penetrate the lens and reach the retina to induce retinal damage, causing dry age-related macular degeneration (dAMD). Cynaroside (Cyn), a flavonoid glycoside, was proved to alleviate the oxidative damage of retinal cells in vitro. However, whether or not Cyn also exerts protective effect on blue light-induced retinal degeneration and its mechanisms of action are unclear. PURPOSE This study aims to evaluate the protective effects of Cyn against blue-light induced retinal degeneration and its underlying mechanisms in vitro and in vivo. STUDY DESIGN/METHODS Blue light-induced N-retinylidene-N-retinylethanolamine (A2E)-laden adult retinal pigment epithelial-19 (ARPE-19) cell damage and retinal damage in SD rats were respectively used to evaluate the protective effects of Cyn on retinal degeneration in vitro and in vivo. MTT assay and AnnexinV-PI double staining assay were used to evaluate the in vitro efficacy. Histological analysis, TUNEL assay, and fundus imaging were conducted to evaluate the in vivo efficacy. ELISA assay, western blot, and immunostaining were performed to investigate the mechanisms of action of Cyn. RESULTS Cyn decreased the blue light-induced A2E-laden ARPE-19 cell damage and oxidative stress. Intravitreal injection of Cyn (2, 4 μg/eye) reversed the retinal degeneration induced by blue light in SD rats. Furthermore, Cyn inhibited the nuclear translocation of NF-κB and induced autophagy, which led to the clearance of overactivated pyrin domain containing 3 (NLRP3) inflammasome in vitro and in vivo. CONCLUSION Cyn protects against blue light-induced retinal degeneration by modulating autophagy and decreasing the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Jia-Hao Feng
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Xiao-Wei Dong
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Hao-Li Yu
- State Key Laboratory of Bioelectronics, Jiangsu Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210009, People's Republic of China
| | - Wei Shen
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Xian-Yu Lv
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Rong Wang
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Xue-Xiang Cheng
- Hubei Fenghuang Baiyunshan Pharmaceutical Co., Ltd, Macheng 438300, People's Republic of China
| | - Fei Xiong
- State Key Laboratory of Bioelectronics, Jiangsu Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210009, People's Republic of China
| | - Xiao-Long Hu
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| | - Hao Wang
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
31
|
Dai JM, Guo WN, Tan YZ, Niu KW, Zhang JJ, Liu CL, Yang XM, Tao KS, Chen ZN, Dai JY. Wogonin alleviates liver injury in sepsis through Nrf2-mediated NF-κB signalling suppression. J Cell Mol Med 2021; 25:5782-5798. [PMID: 33982381 PMCID: PMC8184690 DOI: 10.1111/jcmm.16604] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/24/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Sepsis is a life‐threatening organ dysfunction syndrome, and liver is a susceptible target organ in sepsis, because the activation of inflammatory pathways contributes to septic liver injury. Oxidative stress has been documented to participate in septic liver injury, because it not only directly induces oxidative genotoxicity, but also exacerbates inflammatory pathways to potentiate damage of liver. Therefore, to ameliorate oxidative stress is promising for protecting liver in sepsis. Wogonin is the compound extracted from the medicinal plant Scutellaria baicalensis Geogi and was found to exert therapeutic effects in multiple inflammatory diseases via alleviation of oxidative stress. However, whether wogonin is able to mitigate septic liver injury remains unknown. Herein, we firstly proved that wogonin treatment could improve survival of mice with lipopolysaccharide (LPS)‐ or caecal ligation and puncture (CLP)‐induced sepsis, together with restoration of reduced body temperature and respiratory rate, and suppression of several pro‐inflammatory cytokines in circulation. Then, we found that wogonin effectively alleviated liver injury via potentiation of the anti‐oxidative capacity. To be specific, wogonin activated Nrf2 thereby promoting expressions of anti‐oxidative enzymes including NQO‐1, GST, HO‐1, SOD1 and SOD2 in hepatocytes. Moreover, wogonin‐induced Nrf2 activation could suppress NF‐κB‐regulated up‐regulation of pro‐inflammatory cytokines. Ultimately, we provided in vivo evidence that wogonin activated Nrf2 signalling, potentiated anti‐oxidative enzymes and inhibited NF‐κB‐regulated pro‐inflammatory signalling. Taken together, this study demonstrates that wogonin can be the potential therapeutic agent for alleviating liver injury in sepsis by simultaneously ameliorating oxidative stress and inflammatory response through the activation of Nrf2.
Collapse
Affiliation(s)
- Ji-Min Dai
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Wei-Nan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yi-Zhou Tan
- Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Kun-Wei Niu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jia-Jia Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Cheng-Li Liu
- Department of Hepatobiliary Surgery, Air Force Medical Center, Beijing, China.,Fourth Military Medical University, Xi'an, China
| | - Xiang-Min Yang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Kai-Shan Tao
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhi-Nan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Jing-Yao Dai
- Department of Hepatobiliary Surgery, Air Force Medical Center, Beijing, China.,Fourth Military Medical University, Xi'an, China
| |
Collapse
|
32
|
Wang YR, Zhang XN, Meng FG, Zeng T. Targeting macrophage polarization by Nrf2 agonists for treating various xenobiotics-induced toxic responses. Toxicol Mech Methods 2021; 31:334-342. [PMID: 33627030 DOI: 10.1080/15376516.2021.1894624] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Macrophages can polarize into different phenotypes in response to different microenvironmental stimuli. Macrophage polarization has been assigned to two extreme states, namely proinflammatory M1 and anti-inflammatory M2. Accumulating evidences have demonstrated that M1 polarized macrophages contribute to various toxicants-induced deleterious effects. Switching macrophages from proinflammatory M1 phenotype toward anti-inflammatory M2 phenotype could be a promising approach for treating various inflammatory diseases. Studies in the past few decades have revealed that nuclear factor erythroid 2-related factor 2 (Nrf2) can modulate the polarization of macrophages. Specifically, activation of Nrf2 could block M1 stimuli-induced production of proinflammatory cytokines and chemokines, and shift the polarization of macrophages toward M2 by cross-talking with nuclear factor kappa-B (NF-κB), mitogen-activated protein kinases (MAPKs), peroxisome proliferator-activated receptor γ (PPARγ), and autophagy. Importantly, a great number of studies have confirmed the beneficial effects of natural and synthesized Nrf2 agonists on various inflammatory diseases; however, most of these compounds are far away from clinical application due to lack of characterization and defects of study designs. Interestingly, some endogenous Nrf2 inducers and compounds with dual activities (such as the Nrf2 inducing and CO releasing effects) exhibit potent anti-inflammatory effects, which points out an important direction for future researches.
Collapse
Affiliation(s)
- Yi-Ran Wang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiu-Ning Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Fan-Ge Meng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
33
|
Sela Y, Li J, Kuri P, Merrell AJ, Li N, Lengner C, Rompolas P, Stanger BZ. Dissecting phenotypic transitions in metastatic disease via photoconversion-based isolation. eLife 2021; 10:63270. [PMID: 33620315 PMCID: PMC7929558 DOI: 10.7554/elife.63270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 02/19/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer patients often harbor occult metastases, a potential source of relapse that is targetable only through systemic therapy. Studies of this occult fraction have been limited by a lack of tools with which to isolate discrete cells on spatial grounds. We developed PIC-IT, a photoconversion-based isolation technique allowing efficient recovery of cell clusters of any size – including single-metastatic cells – which are largely inaccessible otherwise. In a murine pancreatic cancer model, transcriptional profiling of spontaneously arising microcolonies revealed phenotypic heterogeneity, functionally reduced propensity to proliferate and enrichment for an inflammatory-response phenotype associated with NF-κB/AP-1 signaling. Pharmacological inhibition of NF-κB depleted microcolonies but had no effect on macrometastases, suggesting microcolonies are particularly dependent on this pathway. PIC-IT thus enables systematic investigation of metastatic heterogeneity. Moreover, the technique can be applied to other biological systems in which isolation and characterization of spatially distinct cell populations is not currently feasible.
Collapse
Affiliation(s)
- Yogev Sela
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, United States.,Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Jinyang Li
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, United States.,Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Paola Kuri
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, United States.,Department of Dermatology, University of Pennsylvania, Philadelphia, PA, United States
| | - Allyson J Merrell
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, United States.,Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Ning Li
- Department of Biomedical Sciences, School of Veterinary Medicine, Philadelphia, PA, United States.,Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Chris Lengner
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, United States.,Department of Biomedical Sciences, School of Veterinary Medicine, Philadelphia, PA, United States.,Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Pantelis Rompolas
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, United States.,Department of Dermatology, University of Pennsylvania, Philadelphia, PA, United States
| | - Ben Z Stanger
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, United States.,Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, United States.,Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
34
|
Gao S, Xue X, Yin J, Gao L, Li Z, Li L, Gao S, Wang S, Liang R, Xu Y, Yu C, Zhu Y. Danlou tablet inhibits the inflammatory reaction of high-fat diet-induced atherosclerosis in ApoE knockout mice with myocardial ischemia via the NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2020; 263:113158. [PMID: 32745509 DOI: 10.1016/j.jep.2020.113158] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 06/08/2020] [Accepted: 07/02/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Danlou tablet (DLT), a traditional herbal formula, has been used to treat chest discomfort (coronary atherosclerosis) in China. Although the anti-inflammatory activities of DLT have been proposed previously, the mechanisms of DLT in treating atherosclerosis with myocardial ischemia (AWMI) remain unknown. AIM OF THE STUDY Atherosclerosis can result in heart disease caused by stenosis or occlusion of the lumen, resulting in myocardial ischemia, hypoxia, or necrosis. In recent years, changes in people's diets, increased stress, and secondary fatigue and obesity etc. have resulted in increases in the number of patients with atherosclerosis. In cases where the condition has further developed, patients may suffer from myocardial ischemia, hypoxia, or necrosis. Many traditional Chinese medicine compounds have been prescribed for the treatment of AWMI. DLT has been used to treat chest discomfort (coronary atherosclerosis) in China. Based on previous research, the aim of this study was to further investigate the effect of DLT on AWMI, and describe the underlying mechanisms. MATERIALS AND METHODS To achieve this, an animal model of AWMI was established using apolipoprotein E (ApoE-/-) mice fed a high fat diet combined with isoprenaline (ISO) injection. For comparison, mouse models of only atherosclerosis and only myocardial ischemia were included. In the treatment groups, mice were treated daily with DLT at 700 mg/kg for four weeks. Echocardiographic evaluation, hematoxylin and eosin (H&E) staining, oil red O staining, ELISAs, Western blots, and immunohistochemical analyses were subsequently used to investigate the mechanism of DLT based on the NF-κB signaling pathway. RESULTS The results indicate that the use of DLT is effective, to varying degrees, for the treatment of atherosclerosis, myocardial ischemia, and AWMI in mice. After DLT treatment, the left ventricular structure and morphology of the mice, the histopathology of cardiac tissue, and atherosclerotic plaques in the aortas all improved to varying degrees. DLT could play a therapeutic role by regulating the NF-κB signaling pathway related to inflammatory factors, including TNF-α, IL-6, IL-1β, IL-8, MMP-1 and MMP-2, as well as protein expression of NF-κB p-50 and IκB-α, and positive cell expression of NF-κB p-50, IκB-α and phospho-NF-κB p-50 in the model mice. CONCLUSION These preliminary results indicate that the therapeutic efficacy of DLT on high-fat diet-induced atherosclerosis in ApoE-/- mice with myocardial ischemia could be exerted at least in part by regulating the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Shan Gao
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Xiaoxue Xue
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Jia Yin
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Lina Gao
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China; College of Pharmacy, Jining Medical University, Rizhao, PR China.
| | - Zhu Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Lin Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Shuming Gao
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Shuo Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Ru Liang
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Yilan Xu
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Chunquan Yu
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Yan Zhu
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China; Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| |
Collapse
|
35
|
A novel protective role of sacubitril/valsartan in cyclophosphamide induced lung injury in rats: impact of miRNA-150-3p on NF-κB/MAPK signaling trajectories. Sci Rep 2020; 10:13045. [PMID: 32747644 PMCID: PMC7400763 DOI: 10.1038/s41598-020-69810-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 06/30/2020] [Indexed: 12/12/2022] Open
Abstract
Cyclophosphamide (CP) is a chemotherapeutic agent that induces oxidative stress causing multiple organ damage. Sacubitril/valsartan, is a combined formulation of neprilysin inhibitor (sacubitril) and angiotensin II receptor blocker (valsartan), that induces the protective effect of brain natriuretic peptide. The aim of the current study is to investigate the prophylactic impacts of sacubitril/valsartan versus valsartan against CP-induced lung toxicity in rats. Rats were assigned randomly into 6 groups; control; received corn oil (2 ml/kg/day; p.o. for 6 days), sacubitril/valsartan (30 mg/kg; p.o. for 6 days), valsartan (15 mg/kg; p.o. for 6 days), CP (200 mg/kg; i.p. on day 5), sacubitril/valsartan + CP (30 mg/kg; p.o. for 6 days, 200 mg/kg; i.p. single dose on day 5, respectively), valsartan + CP (15 mg/kg; p.o. for 6 days, 200 mg/kg; i.p. single dose on day 5, respectively). Both sacubitril/valsartan and valsartan produced a significant decrease in the inflammation and fibrosis markers in the BALF, in comparison with the CP group. Both sacubitril/valsartan and valsartan produced an apparent decrease in the relative genes expression of miR-150-3p and NF-κB, as well as a significant decrease in the relative expression of P38 and ERK1/2 MAPKs and an increase in the relative gene expression of Nrf-2, compared to CP group. Intriguingly, sacubitril/valsartan , showed subtle superiority in almost all investigated parameters, compared to valsartan. In conclusion, sacubitril/valsartan effectively abrogated the CP induced lung inflammation and fibrosis, providing a potential promising protection that could be linked to their ability to inhibit miR-150-3p via inhibition of NF-κB and MAPK signaling pathways.
Collapse
|
36
|
Yadav S, Singh M, Singh SN, Kumar B. Tanshinone IIA pretreatment promotes cell survival in human lung epithelial cells under hypoxia via AP-1-Nrf2 transcription factor. Cell Stress Chaperones 2020; 25:427-440. [PMID: 32144684 PMCID: PMC7193010 DOI: 10.1007/s12192-020-01083-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/11/2020] [Accepted: 02/19/2020] [Indexed: 12/27/2022] Open
Abstract
Activator protein-1 (AP-1) plays a decisive role in cell proliferation, apoptosis, and inflammation under hypoxia; thus, AP-1 subunits or dimers could be modulated for a desired phenomenon in a cell using a suitable compound of therapeutic potential. Herein, we used Tanshinone-IIA as an AP-1 (subunits) modulator, and the purpose of the study was to investigate the signaling mechanism exhibited by Tan-IIA in facilitating tolerance to hypoxia. A549 cells were pretreated with Tan-IIA and exposed to hypoxia for 6, 12, 24, and 48 h. Biochemical and molecular parameters were assessed in order to trace the signaling pathway. Tan-IIA attenuated hypoxia-induced oxidative stress by modulating the expression of AP-1 subunits (via. MAPK) and Nrf2 transcription factor, which in turn were responsible for maintaining the higher levels of antioxidant enzymes and genes (HO). Tan-IIA increased the cell survival. This could be attributed to an increased NO level via iNOS gene and activated JNK, ERK pathway that induced c-jun/c-fos, c-jun/fosB, junD/c-fos, and junD/fosB heterodimers. This in turn leads to the cell cycle progression by activating cyclins (D and B). This was further confirmed by the lower levels of p53 and their downstream genes (p16, p21, p27). In addition, Tan-IIA decreased pro-inflammatory cytokine levels by inhibiting the formation of junB/fra-1 heterodimer regulated by p38. Tan-IIA increased cell survival to hypoxia by maintaining the higher levels of cellular iNOS, HO-1, jun-D, c-jun, fos B via Nrf2-AP-1.
Collapse
Affiliation(s)
- Seema Yadav
- Experimental Biology Division, Defence Institute of Physiology and Allied Science, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110 054, India
| | - Mrinalini Singh
- Experimental Biology Division, Defence Institute of Physiology and Allied Science, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110 054, India.
| | - Som Nath Singh
- Experimental Biology Division, Defence Institute of Physiology and Allied Science, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110 054, India
| | - Bhuvnesh Kumar
- Experimental Biology Division, Defence Institute of Physiology and Allied Science, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110 054, India
| |
Collapse
|
37
|
Zhang J, Cui H, Namani A, Yao J, Deng H, Tang X, Wang XJ. Transcriptomic profiling identifies a critical role of Nrf2 in regulating the inflammatory response to fly ash particles in mouse lung. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 190:110132. [PMID: 31918253 DOI: 10.1016/j.ecoenv.2019.110132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 12/17/2019] [Accepted: 12/23/2019] [Indexed: 06/10/2023]
Abstract
Exposure to combustion-derived nanoparticles is recognized as a major health hazard, but the molecular responses are still insufficiently described. The transcription factor erythroid 2-related factor 2 (Nrf2, also known as NFE2L2) is a master regulator of the pulmonary defense system against insults by particulate matter. However, its downstream molecular processes are not fully characterized. In the current study, BALB/c wild-type (WT) and Nrf2-/- mice were exposed by intranasal administration to fly ash particles (F3-S; 20 mg/kg BW), which were collected from a municipal waste incinerator in China, for three consecutive days. Using a comparative transcriptomics approach, the pulmonary global gene expression profiles to F3-S exposure were characterized for both genotypes. The preponderance of the differentially-expressed genes (DEGs) in WT mice induced by the fly ash particles, was related to inflammation. Functional enrichment and molecular pathway mapping of the DEGs specific to Nrf2-/- mice exposed to the particles revealed that all of the top 10 perturbed molecular pathways were associated with the inflammatory response. Our study identified a transcriptional signature related to the initial pulmonary injury in mouse upon fly ash exposure, and suggests an anti-inflammatory role of Nrf2 in protecting the lung against such exposure.
Collapse
Affiliation(s)
- Jingwen Zhang
- Department of Pharmacology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Huiling Cui
- Department of Pharmacology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Akhileshwar Namani
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Jun Yao
- Department of Pharmacology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Hong Deng
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Xiuwen Tang
- Department of Biochemistry and Department of Thoracic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Xiu Jun Wang
- Department of Pharmacology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, PR China.
| |
Collapse
|
38
|
Caracci F, Harary J, Simkovic S, Pasinetti GM. Grape-Derived Polyphenols Ameliorate Stress-Induced Depression by Regulating Synaptic Plasticity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:1808-1815. [PMID: 31532659 DOI: 10.1021/acs.jafc.9b01970] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Major depressive disorder (MDD) is associated with stress-induced immune dysregulation and reduced brain-derived neurotrophic factor (BDNF) levels in sensitive brain regions associated with depression. Elevated levels of proinflammatory cytokines and reduced BDNF levels lead to impaired synaptic plasticity mechanisms that contribute to the pathophysiology of MDD. There is accumulating evidence that the administration of polyphenols at doses ranging from 5 to 180 mg/kg of body weight can normalize elevated levels of proinflammatory cytokines and abnormal levels of BDNF and, thus, restore impaired synaptic plasticity mechanisms that mediate depressive behavior in animal models of stress. This review will focus on the mechanisms by which grape-derived polyphenols normalize impaired synaptic plasticity and reduce depressive behavior in animal models of stress.
Collapse
Affiliation(s)
- Francesca Caracci
- Department of Neurology , Icahn School of Medicine at Mount Sinai , 1 Gustave L. Levy Place , Box 1137, New York , New York 10029 , United States
| | - Joyce Harary
- Department of Neurology , Icahn School of Medicine at Mount Sinai , 1 Gustave L. Levy Place , Box 1137, New York , New York 10029 , United States
| | - Sherry Simkovic
- Department of Neurology , Icahn School of Medicine at Mount Sinai , 1 Gustave L. Levy Place , Box 1137, New York , New York 10029 , United States
| | - Giulio Maria Pasinetti
- Department of Neurology , Icahn School of Medicine at Mount Sinai , 1 Gustave L. Levy Place , Box 1137, New York , New York 10029 , United States
- Geriatrics Research, Education and Clinical Center , JJ Peters VA Medical Center , Bronx , New York 10468 , United States
| |
Collapse
|
39
|
Liu Y, Deng J, Fan D. Ginsenoside Rk3 ameliorates high-fat-diet/streptozocin induced type 2 diabetes mellitus in mice via the AMPK/Akt signaling pathway. Food Funct 2020; 10:2538-2551. [PMID: 30993294 DOI: 10.1039/c9fo00095j] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ginsenoside Rk3 (G-Rk3) is a main active ingredient of ginsenosides. Several recent studies demonstrated that ginsenosides have potential anti-type 2 diabetes mellitus (T2DM) properties. To evaluate the anti-T2DM effect of G-Rk3 and verify its potential mechanism, a high-fat-diet/streptozocin (HFD/STZ) induced model of T2DM in C57BL/6 mice and a high glucose induced insulin resistance model of HepG2 cells were applied in this research. Our analysis indicated that G-Rk3 reduced HFD/STZ induced hyperglycemia, and serum insulin and inflammation levels, and ameliorated glucose tolerance and insulin resistance, and prevented liver histological changes. Furthermore, it also significantly reduced lipid accumulation as shown by lower TG, LDL-C and TC serum concentrations and Oil Red O staining in liver tissues. The hypoglycemic effect of G-Rk3 seemed to be partially mediated via the inhibition of hepatic gluconeogenesis, which was supported by the activated p-Akt, p-FoxO1 and GLUT2 and inhibited FoxO1, PEPCK and G6pase protein expressions in the liver as well as increased glucose uptake in high glucose induced HepG2 cells. The gene expressions of hepatic gluconeogenesis were also down-regulated by G-Rk3 in HFD/STZ induced T2DM mice. In addition, G-Rk3 suppressed HFD/STZ induced lipid accumulation by regulating related gene and protein expressions such as p-ACC, FAS and SREBP-1, which are the downstream targets of AMPK. AMPK and Akt inhibitors significantly reversed G-Rk3 mediated hepatic gluconeogenesis and lipid accumulation. Thus, our study is the first to illustrate that G-Rk3 mediates hepatic gluconeogenesis and lipid accumulation via activating the AMPK/Akt signaling pathway in HFD/STZ induced T2DM mice.
Collapse
Affiliation(s)
- Yao Liu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China.
| | | | | |
Collapse
|
40
|
Wang R, Luo Y, Lu Y, Wang D, Wang T, Pu W, Wang Y. Maggot Extracts Alleviate Inflammation and Oxidative Stress in Acute Experimental Colitis via the Activation of Nrf2. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4703253. [PMID: 31827675 PMCID: PMC6885191 DOI: 10.1155/2019/4703253] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/23/2019] [Accepted: 10/14/2019] [Indexed: 12/12/2022]
Abstract
Ulcerative colitis (UC) is a common chronic remitting disease driven through altered immune responses with production of inflammatory cytokines. Oxidant/antioxidant balance is also suggested to be an important factor for the recurrence and progression of UC. Maggots are known as a traditional Chinese medicine also known as "wu gu chong." NF-E2-related factor-2 (Nrf2) transcription factor regulates the oxidative stress response and also represses inflammation. The aim of this study was to investigate the effects of maggot extracts on the amelioration of inflammation and oxidative stress in a mouse model of dextran sulfate sodium- (DSS-) induced colitis and evaluate if the maggot extracts could repress inflammation and oxidative stress using RAW 264.7 macrophages stimulated by lipopolysaccharide (LPS). In the present study, we found that the maggot extracts significantly prevented the loss of body weight and shortening of colon length in UC induced by DSS. Furthermore, DSS-induced expression of proinflammatory cytokines at both mRNA and protein levels in the colon was also attenuated by the maggot extracts. In addition, the maggot extracts could significantly suppress the expression of interleukin- (IL-) 1β, IL-6, TNF-α, NFκB p65, p-IκB, p22-phox, and gp91-phox in LPS-stimulated RAW 264.7 cells and colonic tissues. The maggot extracts increased the level of Nrf2 and prevented the degradation of Nrf2 through downregulating the expression of Keap1, which resulted in augmented levels of HO-1, SOD, and GSH-Px and reduced levels of MPO and MDA. However, after administering an Nrf2 inhibitor (ML385) to block the Nrf2/HO-1 pathway, we failed to observe the protective effects of the maggot extracts in mice with colitis and RAW 264.7 cells. Taken together, our data for the first time confirmed that the maggot extracts ameliorated inflammation and oxidative stress in experimental colitis via modulation of the Nrf2/HO-1 pathway. This study sheds light on the possible development of an effective therapeutic strategy for inflammatory bowel diseases.
Collapse
Affiliation(s)
- Rong Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Yongzheng Luo
- School of Chemistry and Life Sciences, Nanjing University Jinling College, 210089, China
| | - Yadong Lu
- Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Daojuan Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Tingyu Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Wenyuan Pu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Yong Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| |
Collapse
|
41
|
The Role of Nrf2 Activity in Cancer Development and Progression. Cancers (Basel) 2019; 11:cancers11111755. [PMID: 31717324 PMCID: PMC6896028 DOI: 10.3390/cancers11111755] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/03/2019] [Accepted: 11/05/2019] [Indexed: 12/15/2022] Open
Abstract
Nrf2 is a transcription factor that stimulates the expression of genes which have antioxidant response element-like sequences in their promoter. Nrf2 is a cellular protector, and this principle applies to both normal cells and malignant cells. While healthy cells are protected from DNA damage induced by reactive oxygen species, malignant cells are defended against chemo- or radiotherapy. Through our literature search, we found that Nrf2 activates several oncogenes unrelated to the antioxidant activity, such as Matrix metallopeptidase 9 (MMP-9), B-cell lymphoma 2 (BCL-2), B-cell lymphoma-extra large (BCL-xL), Tumour Necrosis Factor α (TNF-α), and Vascular endothelial growth factor A (VEGF-A). We also did a brief analysis of The Cancer Genome Atlas (TCGA) data of lung adenocarcinoma concerning the effects of radiation therapy and found that the therapy-induced Nrf2 activation is not universal. For instance, in the case of recurrent disease and radiotherapy, we observed that, for the majority of Nrf2-targeted genes, there is no change in expression level. This proves that the universal, axiomatic rationale that Nrf2 is activated as a response to chemo- and radiation therapy is wrong, and that each scenario should be carefully evaluated with the help of Nrf2-targeted genes. Moreover, there were nine genes involved in lipid peroxidation, which showed underexpression in the case of new radiation therapy: ADH1A, ALDH3A1, ALDH3A2, ADH1B, GPX2, ADH1C, ALDH6A1, AKR1C3, and NQO1. This may relate to the fact that, while some studies reported the co-activation of Nrf2 and other oncogenic signaling pathways such as Phosphoinositide 3-kinases (PI3K), mitogen-activated protein kinase (MAPK), and Notch1, other reported the inverse correlation between Nrf2 and the tumor-promoter Transcription Factor (TF), Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Lastly, Nrf2 establishes its activity through interactions at multiple levels with various microRNAs. MiR-155, miR-144, miR-28, miR-365-1, miR-93, miR-153, miR-27a, miR-142, miR-29-b1, miR-340, and miR-34a, either through direct repression of Nrf2 messenger RNA (mRNA) in a Kelch-like ECH-associated protein 1 (Keap1)-independent manner or by enhancing the Keap1 cellular level, inhibit the Nrf2 activity. Keap1–Nrf2 interaction leads to the repression of miR-181c, which is involved in the Nuclear factor kappa light chain enhancer of activated B cells (NF-κB) signaling pathway. Nrf2’s role in cancer prevention, diagnosis, prognosis, and therapy is still in its infancy, and the future strategic planning of Nrf2-based oncological approaches should also consider the complex interaction between Nrf2 and its various activators and inhibitors.
Collapse
|
42
|
Crosstalk of toll-like receptors signaling and Nrf2 pathway for regulation of inflammation. Biomed Pharmacother 2018; 108:1866-1878. [PMID: 30372892 DOI: 10.1016/j.biopha.2018.10.019] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 09/09/2018] [Accepted: 10/03/2018] [Indexed: 12/20/2022] Open
Abstract
Inflammation as a second line of defense of innate immunity plays a crucial role in eliminating invading pathogens (bacteria, viruses, fungi as well as other parasites). The inflammatory response may also activate adaptive immune system involving lymphocytes to mount either antibody dependent or cell-mediated immune responses to clear pathogenic insult. However, if continued, the inflammatory processes may become uncontrolled culminating in cellular injury and tissue destruction, thereby manifesting itself in chronic form. The chronic inflammation has been associated with numerous human pathological conditions like allergies and autoimmune diseases, atherosclerosis, arthritis, Alzheimer's disease, cancer, obesity, type 2 diabetes, schizophrenia, neuro-degenerative diseases and numerous others. The dysregulated inflammatory process is associated with overproduction of free radicals leading to oxidative stress and activation of different cell signaling pathways. The regulation of inflammation by TLR signaling as well as Nrf2 pathways separately is widely documented. Since both these major signaling pathways modulate inflammation, they may crosstalk to bring about coordinated inflammatory responses. The linkage between TLR signaling and Nrf2-Keap1 pathway may serve as a bridge between immune regulation and oxidative stress responses through regulation of inflammation. Also, inflammation is reportedly responsible for the plethora of diseased conditions; a study of its regulation by targeting the TLR-Nrf2 cross-talks may also be beneficial for the development of therapeutic therapies or prophylactic treatments. Hence, present review focuses on the crosstalk between TLR signaling and Nrf2 pathway with respect to their role in modulation of inflammation in normal as well as pathologic conditions.
Collapse
|
43
|
Kim J, Jin HL, Jang DS, Jeong KW, Choung SY. Quercetin-3-O-α-l-arabinopyranoside protects against retinal cell death via blue light-induced damage in human RPE cells and Balb-c mice. Food Funct 2018. [PMID: 29541735 DOI: 10.1039/c7fo01958k] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Age-related macular degeneration (AMD) is among the increasing number of diseases causing irreversible blindness in the elderly. Dry AMD is characterized by the accumulation of lipofuscin in retinal pigment epithelium (RPE) cells. N-Retinylidene-N-retinylethanolamine (A2E), a component of lipofuscin, is oxidized to oxo-A2E under blue light illumination, leading to retinal cell death. The aim of this study was to investigate the protective effect and mechanism of quercetin-3-O-α-l-arabinopyranoside (QA) against blue light (BL)-induced damage in both RPE cells and mice models. Treatment by QA inhibited A2E uptake in RPE cells, as determined by a decrease in fluorescence intensity. QA also protected A2E-laden RPE cells against BL-induced apoptosis. QA inhibited C3 complement activation and poly (ADP-ribose) polymerase (PARP) cleavage, as determined by western blotting. QA showed an inhibitory effect on AP1 and NF-kB activity as estimated in a reporter gene assay. In addition, QA activated the gene expression of aryl hydrocarbon receptor target genes (CYP1A1, CYP1B1) in TCDD-treated RPE cells. In the mice model, oral administration of QA protected against retinal degeneration induced by BL exposure as determined by histological analyses (thickness of retinal layers and immunostaining for caspase-3). In addition, QA inhibited apoptosis and inflammation via inhibition of NF-kB p65 translocation, C3 activation, and PARP cleavage. Collectively, these results revealed the protective mechanism of QA against BL-induced retinal damage both in vitro and in vivo.
Collapse
Affiliation(s)
- Jun Kim
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | | | | | | | | |
Collapse
|
44
|
STIM- and Orai-mediated calcium entry controls NF-κB activity and function in lymphocytes. Cell Calcium 2018; 74:131-143. [PMID: 30048879 DOI: 10.1016/j.ceca.2018.07.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 12/24/2022]
Abstract
The central role of Ca2+ signaling in the development of functional immunity and tolerance is well established. These signals are initiated by antigen binding to cognate receptors on lymphocytes that trigger store operated Ca2+ entry (SOCE). The underlying mechanism of SOCE in lymphocytes involves TCR and BCR mediated activation of Stromal Interaction Molecule 1 and 2 (STIM1/2) molecules embedded in the ER membrane leading to their activation of Orai channels in the plasma membrane. STIM/Orai dependent Ca2+ signals guide key antigen induced lymphocyte development and function principally through direct regulation of Ca2+ dependent transcription factors. The role of Ca2+ signaling in NFAT activation and signaling is well known and has been studied extensively, but a wide appreciation and mechanistic understanding of how Ca2+ signals also shape the activation and specificity of NF-κB dependent gene expression has lagged. Here we discuss and interpret what is known about Ca2+ dependent mechanisms of NF-kB activation, including what is known and the gaps in our understanding of how these signals control lymphocyte development and function.
Collapse
|
45
|
Ferrándiz ML, Nacher-Juan J, Alcaraz MJ. Nrf2 as a therapeutic target for rheumatic diseases. Biochem Pharmacol 2018; 152:338-346. [PMID: 29660314 DOI: 10.1016/j.bcp.2018.04.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/11/2018] [Indexed: 12/21/2022]
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a master regulator of cellular protective processes. Rheumatic diseases are chronic conditions characterized by inflammation, pain, tissue damage and limitations in function. Main examples are rheumatoid arthritis, systemic lupus erythematosus, osteoarthritis and osteoporosis. Their high prevalence constitutes a major health problem with an important social and economic impact. A wide range of evidence indicates that Nrf2 may control different mechanisms involved in the physiopathology of rheumatic conditions. Therefore, the appropriate expression and balance of Nrf2 is necessary for regulation of oxidative stress, inflammation, immune responses, and cartilage and bone metabolism. Numerous studies have demonstrated that Nrf2 deficiency aggravates the disease in experimental models while Nrf2 activation results in immunoregulatory and anti-inflammatory effects. These reports reinforce the increasing interest in the pharmacologic regulation of Nrf2 and its potential applications. Nevertheless, a majority of Nrf2 inducers are electrophilic molecules which may present off-target effects. In recent years, novel strategies have been sought to modulate the Nrf2 pathway which has emerged as a therapeutic target in rheumatic conditions.
Collapse
Affiliation(s)
- María Luisa Ferrándiz
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain.
| | - Josep Nacher-Juan
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain.
| | - Maria José Alcaraz
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain.
| |
Collapse
|
46
|
Dimethyl fumarate attenuates reactive microglia and long-term memory deficits following systemic immune challenge. J Neuroinflammation 2018; 15:100. [PMID: 29598822 PMCID: PMC5877396 DOI: 10.1186/s12974-018-1125-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 03/09/2018] [Indexed: 02/08/2023] Open
Abstract
Background Systemic inflammation is associated with increased cognitive decline and risk for Alzheimer’s disease. Microglia (MG) activated during systemic inflammation can cause exaggerated neuroinflammatory responses and trigger progressive neurodegeneration. Dimethyl fumarate (DMF) is a FDA-approved therapy for multiple sclerosis. The immunomodulatory and anti-oxidant properties of DMF prompted us to investigate whether DMF has translational potential for the treatment of cognitive impairment associated with systemic inflammation. Methods Primary murine MG cultures were stimulated with lipopolysaccharide (LPS) in the absence or presence of DMF. MG cultured from nuclear factor (erythroid-derived 2)-like 2-deficient (Nrf2−/−) mice were used to examine mechanisms of DMF actions. Conditioned media generated from LPS-primed MG were used to treat hippocampal neuron cultures. Adult C57BL/6 and Nrf2−/− mice were subjected to peripheral LPS challenge. Acute neuroinflammation, long-term memory function, and reactive astrogliosis were examined to assess therapeutic effects of DMF. Results DMF suppressed inflammatory activation of MG induced by LPS. DMF suppressed NF-κB activity through Nrf2-depedent and Nrf2-independent mechanisms in MG. DMF treatment reduced MG-mediated toxicity towards neurons. DMF suppressed brain-derived inflammatory cytokines in mice following peripheral LPS challenge. The suppressive effect of DMF on neuroinflammation was blunted in Nrf2−/− mice. Importantly, DMF treatment alleviated long-term memory deficits and sustained reactive astrogliosis induced by peripheral LPS challenge. DMF might mitigate neurotoxic astrocytes associated with neuroinflammation. Conclusions DMF treatment might protect neurons against toxic microenvironments produced by reactive MG and astrocytes associated with systemic inflammation. Electronic supplementary material The online version of this article (10.1186/s12974-018-1125-5) contains supplementary material, which is available to authorized users.
Collapse
|
47
|
Lecoq L, Raiola L, Chabot PR, Cyr N, Arseneault G, Legault P, Omichinski JG. Structural characterization of interactions between transactivation domain 1 of the p65 subunit of NF-κB and transcription regulatory factors. Nucleic Acids Res 2017; 45:5564-5576. [PMID: 28334776 PMCID: PMC5435986 DOI: 10.1093/nar/gkx146] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 02/25/2017] [Indexed: 01/27/2023] Open
Abstract
p65 is a member of the NF-κB family of transcriptional regulatory proteins that functions as the activating component of the p65-p50 heterodimer. Through its acidic transactivation domain (TAD), p65 has the capacity to form interactions with several different transcriptional regulatory proteins, including TFIIB, TFIIH, CREB-binding protein (CBP)/p300 and TAFII31. Like other acidic TADs, the p65 TAD contains two subdomains (p65TA1 and p65TA2) that interact with different regulatory factors depending on the target gene. Despite its role in controlling numerous NF-κB target genes, there are no high-resolution structures of p65TA1 bound to a target transcriptional regulatory factor. In this work, we characterize the interaction of p65TA1 with two factors, the Tfb1/p62 subunit of TFIIH and the KIX domain of CBP. In these complexes, p65TA1 transitions into a helical conformation that includes its characteristic ΦXXΦΦ motif (Φ = hydrophobic amino acid). Structural and functional studies demonstrate that the two binding interfaces are primarily stabilized by three hydrophobic amino acids within the ΦXXΦΦ motif and these residues are also crucial to its ability to activate transcription. Taken together, the results provide an atomic level description of how p65TA1 is able to bind different transcriptional regulatory factors needed to activate NF-κB target genes.
Collapse
Affiliation(s)
- Lauriane Lecoq
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Luca Raiola
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Philippe R Chabot
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Normand Cyr
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Geneviève Arseneault
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Pascale Legault
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - James G Omichinski
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| |
Collapse
|
48
|
Niacin and Selenium Attenuate Sepsis-Induced Lung Injury by Up-Regulating Nuclear Factor Erythroid 2-Related Factor 2 Signaling. Crit Care Med 2017; 44:e370-82. [PMID: 26646455 DOI: 10.1097/ccm.0000000000001422] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVES To determine whether the combination therapy of niacin and selenium attenuates lung injury and improves survival during sepsis in rats and whether its benefits are associated with the activation of the glutathione redox cycle and up-regulation of nuclear factor erythroid 2-related factor 2. DESIGN Prospective laboratory study. SETTING University laboratory. SUBJECTS Human lung microvascular endothelial cells and male Sprague-Dawley rats (n = 291). INTERVENTION In lipopolysaccharide-exposed cells, the dose-related effects of niacin and selenium were assessed, and the therapeutic effects of the combination therapy of niacin (0.9 mM) and selenium (1.5 μM) were evaluated. The role of nuclear factor erythroid 2-related factor 2 was determined using nuclear factor erythroid 2-related factor 2 knockdown cells. In endotoxemic and cecal ligation and puncture with antibiotics rats, the therapeutic effects of the posttreatments of clinically relevant doses of niacin (360 mg/kg) and selenium (60 μg/kg) were evaluated. MEASUREMENTS AND MAIN RESULTS Combination therapy reduced the hydrogen peroxide level via the synergistic activation of the glutathione redox cycle, which involves niacin-induced increases in glutathione reductase activity, and reduced the glutathione level and a selenium-induced increase in glutathione peroxidase activity. Combination therapy contributed to the up-regulation of nuclear factor erythroid 2-related factor 2, enhancement of glutathione synthesis, and down-regulation of nuclear factor κB signaling, but nuclear factor erythroid 2-related factor 2 knockdown inhibited the enhancement of glutathione synthesis and down-regulation of the nuclear factor κB pathway. The therapeutic effects of combination therapy on endotoxemic rats were consistent with those on lipopolysaccharide-exposed cells. In addition, the posttreatment of combination therapy attenuated lung injury and improved survival in endotoxemic and cecal ligation and puncture with antibiotics rats. However, individual therapies of niacin or selenium failed to achieve these benefits. CONCLUSIONS The combination therapy of niacin and selenium attenuated lung injury and improved survival during sepsis. Its therapeutic benefits were associated with the synergistic activation of the glutathione redox cycle, reduction of hydrogen peroxide level, and up-regulation of nuclear factor erythroid 2-related factor 2.
Collapse
|
49
|
Forno E, Sordillo J, Brehm J, Chen W, Benos T, Yan Q, Avila L, Soto-Quirós M, Cloutier MM, Colón-Semidey A, Alvarez M, Acosta-Pérez E, Weiss ST, Litonjua AA, Canino G, Celedón JC. Genome-wide interaction study of dust mite allergen on lung function in children with asthma. J Allergy Clin Immunol 2017; 140:996-1003.e7. [PMID: 28167095 DOI: 10.1016/j.jaci.2016.12.967] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 11/25/2016] [Accepted: 12/12/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND Childhood asthma is likely the result of gene-by-environment (G × E) interactions. Dust mite is a known risk factor for asthma morbidity. Yet, there have been no genome-wide G × E studies of dust mite allergen on asthma-related phenotypes. OBJECTIVE We sought to identify genetic variants whose effects on lung function in children with asthma are modified by the level of dust mite allergen exposure. METHODS A genome-wide interaction analysis of dust mite allergen level and lung function was performed in a cohort of Puerto Rican children with asthma (Puerto Rico Genetics of Asthma and Lifestyle [PRGOAL]). Replication was attempted in 2 independent cohorts, the Childhood Asthma Management Program (CAMP) and the Genetics of Asthma in Costa Rica Study. RESULTS Single nucleotide polymorphism (SNP) rs117902240 showed a significant interaction effect on FEV1 with dust mite allergen level in PRGOAL (interaction P = 3.1 × 10-8), and replicated in the same direction in CAMP white children and CAMP Hispanic children (combined interaction P = .0065 for replication cohorts and 7.4 × 10-9 for all cohorts). Rs117902240 was positively associated with FEV1 in children exposed to low dust mite allergen levels, but negatively associated with FEV1 in children exposed to high levels. This SNP is on chromosome 8q24, adjacent to a binding site for CCAAT/enhancer-binding protein beta, a transcription factor that forms part of the IL-17 signaling pathway. None of the SNPs identified for FEV1/forced vital capacity replicated in the independent cohorts. CONCLUSIONS Dust mite allergen exposure modifies the estimated effect of rs117902240 on FEV1 in children with asthma. Analysis of existing data suggests that this SNP may have transcription factor regulatory functions.
Collapse
Affiliation(s)
- Erick Forno
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pa; University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Joanne Sordillo
- Channing Division of Network Medicine, Department of Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, Mass
| | - John Brehm
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pa; University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Wei Chen
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pa; University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Takis Benos
- University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Qi Yan
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pa; University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Lydiana Avila
- Department of Pediatric Pulmonology, Hospital Nacional de Niños, San José, Costa Rica
| | - Manuel Soto-Quirós
- Department of Pediatric Pulmonology, Hospital Nacional de Niños, San José, Costa Rica
| | - Michelle M Cloutier
- Department of Pediatrics, University of Connecticut Health Center, Farmington, Conn
| | | | - Maria Alvarez
- Department of Pediatrics, University of Puerto Rico, San Juan, Puerto Rico
| | - Edna Acosta-Pérez
- Behavioral Sciences Research Institute, University of Puerto Rico, San Juan, Puerto Rico
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, Mass
| | - Augusto A Litonjua
- Channing Division of Network Medicine, Department of Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, Mass
| | - Glorisa Canino
- Behavioral Sciences Research Institute, University of Puerto Rico, San Juan, Puerto Rico
| | - Juan C Celedón
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pa; University of Pittsburgh School of Medicine, Pittsburgh, Pa.
| |
Collapse
|
50
|
Liu D, Wang Y, Ma S, Sun H, Shi W, Feng X. Individual and combined use of ginsenoside F2 and cyanidin-3-O-glucoside attenuates H 2O 2-induced apoptosis in HEK-293 cells via the NF-κB pathway. RSC Adv 2017; 7:41713-41722. [DOI: 10.1039/c7ra04689h] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
Ginsenoside F2 and cyanidin-3-O-glucoside synergistically inhibited H2O2-induced apoptosis in HEK-293 cells through mitochondria-mediated apoptotic and NF-κB pathways.
Collapse
Affiliation(s)
- Di Liu
- Department of Pathogenic Biology
- Jilin Medical University
- Jilin 132013
- China
| | - Ying Wang
- College of Food Science and Engineering
- Jilin University
- Changchun 130062
- China
| | - Shuang Ma
- College of Food Science and Engineering
- Jilin University
- Changchun 130062
- China
| | - Hongyu Sun
- Department of Pathogenic Biology
- Jilin Medical University
- Jilin 132013
- China
| | - Wenyan Shi
- Department of Pathogenic Biology
- Jilin Medical University
- Jilin 132013
- China
| | - Xianmin Feng
- Department of Pathogenic Biology
- Jilin Medical University
- Jilin 132013
- China
| |
Collapse
|