1
|
de Lima J, Leite JA, Basso PJ, Ghirotto B, Martins da Silva E, Menezes-Silva L, Hiyane MI, Goes CP, Coutinho LL, de Andrade Oliveira V, Olsen Saraiva Câmara N. Sirtuin 1 regulates the phenotype and functions of dendritic cells through Ido1 pathway in obesity. Cell Death Dis 2024; 15:757. [PMID: 39424786 PMCID: PMC11489582 DOI: 10.1038/s41419-024-07125-3] [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: 05/26/2024] [Revised: 09/09/2024] [Accepted: 10/01/2024] [Indexed: 10/21/2024]
Abstract
Sirtuin 1 (SIRT1) is a class III histone deacetylase (HDAC3) that plays a crucial role in regulating the activation and differentiation of dendritic cells (DCs) as well as controlling the polarization and activation of T cells. Obesity, a chronic inflammatory condition, is characterized by the activation of immune cells in various tissues. We hypothesized that SIRT1 might influence the phenotype and functions of DCs through the Ido1 pathway, ultimately leading to the polarization towards pro-inflammatory T cells in obesity. In our study, we observed that SIRT1 activity was reduced in bone marrow-derived DCs (BMDCs) from obese animals. These BMDCs exhibited elevated oxidative phosphorylation (OXPHOS) and increased extracellular acidification rates (ECAR), along with enhanced expression of class II MHC, CD86, and CD40, and elevated secretion of IL-12p40, while the production of TGF-β was reduced. The kynurenine pathway activity was decreased in BMDCs from obese animals, particularly under SIRT1 inhibition. SIRT1 positively regulated the expression of Ido1 in DCs in a PPARγ-dependent manner. To support these findings, ATAC-seq analysis revealed that BMDCs from obese mice had differentially regulated open chromatin regions compared to those from lean mice, with reduced chromatin accessibility at the Sirt1 genomic locus in BMDCs from obese WT mice. Gene Ontology (GO) enrichment analysis indicated that BMDCs from obese animals had disrupted metabolic pathways, including those related to GTPase activity and insulin response. Differential expression analysis showed reduced levels of Pparg and Sirt1 in BMDCs from obese mice, which was challenged and confirmed using BMDCs from mice with conditional knockout of Sirt1 in dendritic cells (SIRT1∆). This study highlights that SIRT1 controls the metabolism and functions of DCs through modulation of the kynurenine pathway, with significant implications for obesity-related inflammation.
Collapse
Affiliation(s)
- Jean de Lima
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jefferson Antônio Leite
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Paulo José Basso
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Bruno Ghirotto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Eloisa Martins da Silva
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luisa Menezes-Silva
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Meire Ioshie Hiyane
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carolina Purcell Goes
- Department of Animal Science, Luiz de Queiroz College of Agriculture (ESALQ), University of São Paulo, Piracicaba, Brazil
| | - Luiz Lehmann Coutinho
- Department of Animal Science, Luiz de Queiroz College of Agriculture (ESALQ), University of São Paulo, Piracicaba, Brazil
| | - Vinicius de Andrade Oliveira
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
- Center for Natural and Human Sciences, Federal University of ABC, Santo André, Brazil.
| | | |
Collapse
|
2
|
Zhou X, Fu Y, Chen J, Liu P. Progress in clinical and basic research of fuzheng Huayu formula for the treatment of liver fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:118018. [PMID: 38453100 DOI: 10.1016/j.jep.2024.118018] [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: 07/07/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine has great potential and advantages in the treatment of liver fibrosis, with Fuzheng Huayu formula (FZHY) serving as a prime example due to its remarkable efficacy in delaying and reversing liver fibrosis while simultaneously improving clinical symptoms for patients. AIM OF THE REVIEW In this paper, we present a comprehensive review of recent studies on the therapeutic potential of FZHY and its components/ingredients in the treatment of liver fibrosis and cirrhosis, with the aim of providing insights for future research endeavors. MATERIALS AND METHODS A comprehensive literature search was conducted on FZHY, TCM319, traditional Chinese medicine 319, liver fibrosis and cirrhosis using multiple internationally recognized databases including PubMed, Embase, Springer, Web of science, SciVerse ScienceDirect, Clinical Trails. Gov, CNKI, Wanfang, and VIP. RESULTS FZHY is widely used clinically for liver fibrosis and cirrhosis caused by various chronic liver diseases, with the effects of improving serum liver function, liver pathological histology, serological indices related to liver fibrosis, decreasing liver stiffness values and portal hypertension, as well as reducing the incidence of hepatocellular carcinoma and morbidity/mortality in patients with cirrhosis. Numerous in vivo and in vitro experiments have demonstrated that FZHY possesses anti-fibrotic effects by inhibiting hepatic stellate cell activation, reducing inflammation, protecting hepatocytes, inhibiting hepatic sinusoidal capillarization and angiogenesis, promoting extracellular matrix degradation, and facilitating liver regeneration. In recent years, there has been a growing focus on investigating the primary active components/ingredients of FZHY, and significant strides have been made in comprehending their synergistic mechanisms that enhance efficacy. CONCLUSION FZHY is a safe and effective drug for treating liver fibrosis. Future research on FZHY should focus on its active components/ingredients and their synergistic effects, as well as the development of modern cocktail drugs based on its components/ingredients. This will facilitate a more comprehensive understanding of the molecular mechanisms and targets of FZHY in treating liver fibrosis, thereby further guide clinical applications and drug development.
Collapse
Affiliation(s)
- Xiaoxi Zhou
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yadong Fu
- Institute of Interdisciplinary Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory of Cell Biology, Center for Excellence in Molecular and Cellular Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jiamei Chen
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Ping Liu
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Institute of Interdisciplinary Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
3
|
Zhu X, Yang Y, Feng D, Wang O, Chen J, Wang J, Wang B, Liu Y, Edenfield BH, Haddock AN, Wang Y, Patel T, Bi Y, Ji B. Albumin promoter-driven FlpO expression induces efficient genetic recombination in mouse liver. Am J Physiol Gastrointest Liver Physiol 2024; 326:G495-G503. [PMID: 38469630 PMCID: PMC11376971 DOI: 10.1152/ajpgi.00263.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 03/13/2024]
Abstract
Tissue-specific gene manipulations are widely used in genetically engineered mouse models. A single recombinase system, such as the one using Alb-Cre, has been commonly used for liver-specific genetic manipulations. However, most diseases are complex, involving multiple genetic changes and various cell types. A dual recombinase system is required for conditionally modifying different genes sequentially in the same cell or inducing genetic changes in different cell types within the same organism. A FlpO cDNA was inserted between the last exon and 3'-UTR of the mouse albumin gene in a bacterial artificial chromosome (BAC-Alb-FlpO). The founders were crossed with various reporter mice to examine the efficiency of recombination. Liver cancer tumorigenesis was investigated by crossing the FlpO mice with FSF-KrasG12D mice and p53frt mice (KPF mice). BAC-Alb-FlpO mice exhibited highly efficient recombination capability in both hepatocytes and intrahepatic cholangiocytes. No recombination was observed in the duodenum and pancreatic cells. BAC-Alb-FlpO-mediated liver-specific expression of mutant KrasG12D and conditional deletion of p53 gene caused the development of liver cancer. Remarkably, liver cancer in these KPF mice manifested a distinctive mixed hepatocellular carcinoma and cholangiocarcinoma phenotype. A highly efficient and liver-specific BAC-Alb-FlpO mouse model was developed. In combination with other Cre lines, different genes can be manipulated sequentially in the same cell, or distinct genetic changes can be induced in different cell types of the same organism.NEW & NOTEWORTHY A liver-specific Alb-FlpO mouse line was generated. By coupling it with other existing CreERT or Cre lines, the dual recombinase approach can enable sequential gene modifications within the same cell or across various cell types in an organism for liver research through temporal and spatial gene manipulations.
Collapse
Affiliation(s)
- Xiaohui Zhu
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States
| | - Yan Yang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States
| | - Dongfeng Feng
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States
| | - Oliver Wang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States
| | - Jiaxiang Chen
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States
| | - Jiale Wang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States
| | - Bin Wang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States
| | - Yang Liu
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States
| | - Brandy H Edenfield
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States
| | - Ashley N Haddock
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States
| | - Ying Wang
- Departments of Cardiovascular Diseases and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States
| | - Tushar Patel
- Department of Transplantation, Mayo Clinic, Jacksonville, Florida, United States
| | - Yan Bi
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida, United States
| | - Baoan Ji
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States
| |
Collapse
|
4
|
You J, Li Y, Chong W. The role and therapeutic potential of SIRTs in sepsis. Front Immunol 2024; 15:1394925. [PMID: 38690282 PMCID: PMC11058839 DOI: 10.3389/fimmu.2024.1394925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by the host's dysfunctional response to infection. Abnormal activation of the immune system and disturbance of energy metabolism play a key role in the development of sepsis. In recent years, the Sirtuins (SIRTs) family has been found to play an important role in the pathogenesis of sepsis. SIRTs, as a class of histone deacetylases (HDACs), are widely involved in cellular inflammation regulation, energy metabolism and oxidative stress. The effects of SIRTs on immune cells are mainly reflected in the regulation of inflammatory pathways. This regulation helps balance the inflammatory response and may lessen cell damage and organ dysfunction in sepsis. In terms of energy metabolism, SIRTs can play a role in immunophenotypic transformation by regulating cell metabolism, improve mitochondrial function, increase energy production, and maintain cell energy balance. SIRTs also regulate the production of reactive oxygen species (ROS), protecting cells from oxidative stress damage by activating antioxidant defense pathways and maintaining a balance between oxidants and reducing agents. Current studies have shown that several potential drugs, such as Resveratrol and melatonin, can enhance the activity of SIRT. It can help to reduce inflammatory response, improve energy metabolism and reduce oxidative stress, showing potential clinical application prospects for the treatment of sepsis. This review focuses on the regulation of SIRT on inflammatory response, energy metabolism and oxidative stress of immune cells, as well as its important influence on multiple organ dysfunction in sepsis, and discusses and summarizes the effects of related drugs and compounds on reducing multiple organ damage in sepsis through the pathway involving SIRTs. SIRTs may become a new target for the treatment of sepsis and its resulting organ dysfunction, providing new ideas and possibilities for the treatment of this life-threatening disease.
Collapse
Affiliation(s)
- Jiaqi You
- Department of Emergency, The First Hospital of China Medical University, Shenyang, China
| | - Yilin Li
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Wei Chong
- Department of Emergency, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Lu H. Inflammatory liver diseases and susceptibility to sepsis. Clin Sci (Lond) 2024; 138:435-487. [PMID: 38571396 DOI: 10.1042/cs20230522] [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: 09/03/2023] [Revised: 01/09/2024] [Accepted: 03/12/2024] [Indexed: 04/05/2024]
Abstract
Patients with inflammatory liver diseases, particularly alcohol-associated liver disease and metabolic dysfunction-associated fatty liver disease (MAFLD), have higher incidence of infections and mortality rate due to sepsis. The current focus in the development of drugs for MAFLD is the resolution of non-alcoholic steatohepatitis and prevention of progression to cirrhosis. In patients with cirrhosis or alcoholic hepatitis, sepsis is a major cause of death. As the metabolic center and a key immune tissue, liver is the guardian, modifier, and target of sepsis. Septic patients with liver dysfunction have the highest mortality rate compared with other organ dysfunctions. In addition to maintaining metabolic homeostasis, the liver produces and secretes hepatokines and acute phase proteins (APPs) essential in tissue protection, immunomodulation, and coagulation. Inflammatory liver diseases cause profound metabolic disorder and impairment of energy metabolism, liver regeneration, and production/secretion of APPs and hepatokines. Herein, the author reviews the roles of (1) disorders in the metabolism of glucose, fatty acids, ketone bodies, and amino acids as well as the clearance of ammonia and lactate in the pathogenesis of inflammatory liver diseases and sepsis; (2) cytokines/chemokines in inflammatory liver diseases and sepsis; (3) APPs and hepatokines in the protection against tissue injury and infections; and (4) major nuclear receptors/signaling pathways underlying the metabolic disorders and tissue injuries as well as the major drug targets for inflammatory liver diseases and sepsis. Approaches that focus on the liver dysfunction and regeneration will not only treat inflammatory liver diseases but also prevent the development of severe infections and sepsis.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, U.S.A
| |
Collapse
|
6
|
Wang M, Zhao J, Chen J, Long T, Xu M, Luo T, Che Q, He Y, Xu D. The role of sirtuin1 in liver injury: molecular mechanisms and novel therapeutic target. PeerJ 2024; 12:e17094. [PMID: 38563003 PMCID: PMC10984179 DOI: 10.7717/peerj.17094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 02/20/2024] [Indexed: 04/04/2024] Open
Abstract
Liver disease is a common and serious threat to human health. The progression of liver diseases is influenced by many physiologic processes, including oxidative stress, inflammation, bile acid metabolism, and autophagy. Various factors lead to the dysfunction of these processes and basing on the different pathogeny, pathology, clinical manifestation, and pathogenesis, liver diseases are grouped into different categories. Specifically, Sirtuin1 (SIRT1), a member of the sirtuin protein family, has been extensively studied in the context of liver injury in recent years and are confirmed the significant role in liver disease. SIRT1 has been found to play a critical role in regulating key processes in liver injury. Further, SIRT1 seems to cause divers outcomes in different types of liver diseases. Recent studies have showed some therapeutic strategies involving modulating SIRT1, which may bring a novel therapeutic target. To elucidate the mechanisms underlying the role of sirtuin1 in liver injury and its potentiality as a therapeutic target, this review outlines the key signaling pathways associated with sirtuin1 and liver injury, and discusses recent advances in therapeutic strategies targeting sirtuin1 in liver diseases.
Collapse
Affiliation(s)
- Mufei Wang
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
- Department of Infectious Diseases, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Juanjuan Zhao
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jiuxia Chen
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Teng Long
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Mengwei Xu
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Tingting Luo
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Qingya Che
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yihuai He
- Department of Infectious Diseases, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Delin Xu
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
7
|
Li S. The β-adrenergic hypothesis of synaptic and microglial impairment in Alzheimer's disease. J Neurochem 2023; 165:289-302. [PMID: 36799441 DOI: 10.1111/jnc.15782] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease originating partly from amyloid β protein-induced synaptic failure. As damaging of noradrenergic neurons in the locus coeruleus (LC) occurs at the prodromal stage of AD, activation of adrenergic receptors could serve as the first line of defense against the onset of the disease. Activation of β2 -ARs strengthens long-term potentiation (LTP) and synaptic activity, thus improving learning and memory. Physical stimulation of animals exposed to an enriched environment (EE) leads to the activation of β2 -ARs and prevents synaptic dysfunction. EE also suppresses neuroinflammation, suggesting that β2 -AR agonists may play a neuroprotective role. The β2 -AR agonists used for respiratory diseases have been shown to have an anti-inflammatory effect. Epidemiological studies further support the beneficial effects of β2 -AR agonists on several neurodegenerative diseases. Thus, I propose that β2 -AR agonists may provide therapeutic value in combination with novel treatments for AD.
Collapse
Affiliation(s)
- Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Curcuma aromatica Salisb. Protects from Acetaminophen-Induced Hepatotoxicity by Regulating the Sirt1/HO-1 Signaling Pathway. Nutrients 2023; 15:nu15040808. [PMID: 36839166 PMCID: PMC9964786 DOI: 10.3390/nu15040808] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/16/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Acetaminophen (APAP) overdose-induced hepatotoxicity reduces the activity of sirtuin-1 (Sirt1) along with heme oxygenase 1 (HO-1) and promotes inflammatory responses and oxidative stress. Although the extract of Curcuma aromatica Salisb. (CAS) possesses hepatoprotective properties, scientific evidence on whether CAS prevents hepatotoxicity and the underlying molecular mechanisms are lacking. Here, we hypothesized that CAS ameliorates hepatotoxicity by inhibiting inflammation and oxidative stress via Sirt1/HO-1 signaling. CAS pretreatment at doses of 200 and 400 μg/mL significantly increased cell viability in APAP-treated primary hepatocytes. The expression of inducible nitric oxide synthase (iNOS) substantially increased after APAP treatment; however, this expression significantly decreased in cells pretreated with 100, 200, and 400 µg/mL CAS. CAS increased Sirt1 and HO-1 levels in APAP-treated hepatocytes in a dose-dependent manner. When CAS was orally administered to mice at doses of 20 or 100 mg/kg for 7 days, the APAP-induced increase in serum aspartate aminotransferase and alanine aminotransferase levels was inhibited. Moreover, CAS decreased IL-6, TNF-α, and IL-1β, increased IL-10, suppressed ROS generation, increased glutathione levels, inhibited iNOS and cyclooxygenase-2, and enhanced Sirt1 and HO-1 in the mouse model of APAP-induced hepatotoxicity. These findings suggest that CAS could be used as a natural hepatoprotective drug to treat APAP-induced injury.
Collapse
|
9
|
Wu QJ, Zhang TN, Chen HH, Yu XF, Lv JL, Liu YY, Liu YS, Zheng G, Zhao JQ, Wei YF, Guo JY, Liu FH, Chang Q, Zhang YX, Liu CG, Zhao YH. The sirtuin family in health and disease. Signal Transduct Target Ther 2022; 7:402. [PMID: 36581622 PMCID: PMC9797940 DOI: 10.1038/s41392-022-01257-8] [Citation(s) in RCA: 352] [Impact Index Per Article: 117.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/10/2022] [Accepted: 11/18/2022] [Indexed: 12/30/2022] Open
Abstract
Sirtuins (SIRTs) are nicotine adenine dinucleotide(+)-dependent histone deacetylases regulating critical signaling pathways in prokaryotes and eukaryotes, and are involved in numerous biological processes. Currently, seven mammalian homologs of yeast Sir2 named SIRT1 to SIRT7 have been identified. Increasing evidence has suggested the vital roles of seven members of the SIRT family in health and disease conditions. Notably, this protein family plays a variety of important roles in cellular biology such as inflammation, metabolism, oxidative stress, and apoptosis, etc., thus, it is considered a potential therapeutic target for different kinds of pathologies including cancer, cardiovascular disease, respiratory disease, and other conditions. Moreover, identification of SIRT modulators and exploring the functions of these different modulators have prompted increased efforts to discover new small molecules, which can modify SIRT activity. Furthermore, several randomized controlled trials have indicated that different interventions might affect the expression of SIRT protein in human samples, and supplementation of SIRT modulators might have diverse impact on physiological function in different participants. In this review, we introduce the history and structure of the SIRT protein family, discuss the molecular mechanisms and biological functions of seven members of the SIRT protein family, elaborate on the regulatory roles of SIRTs in human disease, summarize SIRT inhibitors and activators, and review related clinical studies.
Collapse
Affiliation(s)
- Qi-Jun Wu
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tie-Ning Zhang
- grid.412467.20000 0004 1806 3501Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Huan-Huan Chen
- grid.412467.20000 0004 1806 3501Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xue-Fei Yu
- grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jia-Le Lv
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu-Yang Liu
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ya-Shu Liu
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Gang Zheng
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun-Qi Zhao
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi-Fan Wei
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jing-Yi Guo
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fang-Hua Liu
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qing Chang
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi-Xiao Zhang
- grid.412467.20000 0004 1806 3501Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cai-Gang Liu
- grid.412467.20000 0004 1806 3501Department of Cancer, Breast Cancer Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu-Hong Zhao
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Wang M, Liu H, Zhang X, Zhao W, Li D, Xu C, Wu Z, Xie F, Li X. Lack of Mof reduces acute liver injury by enhancing transcriptional activation of Igf1. J Cell Physiol 2021; 236:6559-6570. [PMID: 33634483 DOI: 10.1002/jcp.30332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/25/2021] [Accepted: 02/08/2021] [Indexed: 12/30/2022]
Abstract
Acute liver injury (ALI) is a rapid pathological process that may cause severe liver disease and may even be life-threatening. During ALI, the function of males absent on the first (MOF) has not yet been elucidated. In this study, we unveiled the expression pattern of MOF during carbon tetrachloride (CCl4 )-induced ALI and role of MOF in the regulation of liver regeneration. In the process of ALI, MOF is significantly overexpressed in the liver injury area. Knockdown of Mof attenuated CCl4 -induced ALI, and promoted liver cell proliferation, hepatic stellate cell activation and aggregation to the injured area, and liver fibrosis. Simultaneously, overexpression of Mof aggravated liver dysfunction caused by ALI. By directly binding to the promoter, MOF suppressed the transcriptional activation of Igf1. Knockdown of Mof promotes the expression of Igf1 and activates the Insulin-like growth factor 1 signaling pathway in the liver. Through this pathway, Knockdown of Mof reduces CCl4 -induced ALI and promotes liver regeneration. Our results provide the first demonstration for MOF contributing to ALI. Further understanding of the role of MOF in ALI may lead to new therapeutic strategies for ALI.
Collapse
Affiliation(s)
- Meng Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
- Department of Cell and Neurobiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Haoyu Liu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
| | - Xu Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
| | - Wenbo Zhao
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Danyang Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
- Department of Rehabilitation, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Chengpeng Xu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
| | - Zhen Wu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
| | - Fei Xie
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
| | - Xiangzhi Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
| |
Collapse
|
11
|
Duan X, Sun W, Sun H, Zhang L. Perfluorooctane sulfonate continual exposure impairs glucose-stimulated insulin secretion via SIRT1-induced upregulation of UCP2 expression. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 278:116840. [PMID: 33689947 DOI: 10.1016/j.envpol.2021.116840] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 06/12/2023]
Abstract
Per- and polyfluoroalkyl substances (PFASs) are environmentally and biologically persistent anthropogenic chemicals linked to adverse health outcomes. Epidemiological data have revealed association between exposure to specific PFAS and disruption of insulin level in bodies. However, the effect of PFASs on insulin secretion and the responsible molecular mechanism are poorly understood. In the present study, we used perfluorooctane sulfonate (PFOS) as a representative PFAS family member to investigate its effect on the insulin secretion in mouse pancreatic β cells (β-TC-6). Our results showed that exposure to PFOS inhibited silent information regulator 1 (SIRT1) activity, and molecular simulation showed PFOS could fit into the pocket overlapped with the nicotinamide adenine dinucleotide (NAD+) binding cavity in SIRT1. PFOS exposure upregulated uncoupling protein 2 (UCP2) expression, and this upregulation was blunted in the presence of Ex-527, a SIRT1 specific inhibitor. The mitochondria membrane potential (ΔΨm), as well as the glucose-induced ATP production and Ca2+ influx decreased under PFOS treatment. PFOS continual exposure (48 h) impaired glucose stimulated insulin secretion (GSIS), while the gene expression of insulin was not significantly altered. Importantly, the SIRT1 activator and UCP2 inhibitor could partly reverse the PFOS-induced impairment of GSIS. Taken together, the results suggested that PFOS continual exposure could inhibit SIRT1 activity, and the SIRT1-UCP2 pathway mediated, at least partially, the PFOS induced GSIS impairment.
Collapse
Affiliation(s)
- Xiaoyu Duan
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China
| | - Weijie Sun
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China
| | - Hongwen Sun
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China
| | - Lianying Zhang
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China; School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin, 300384, China.
| |
Collapse
|
12
|
The Anti-Inflammatory Effect of Zhibaidihuang Decoction on Recurrent Oral Ulcer with Sirt1 as the Key Regulatory Target. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8886699. [PMID: 34007301 PMCID: PMC8110403 DOI: 10.1155/2021/8886699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/10/2021] [Accepted: 04/19/2021] [Indexed: 01/07/2023]
Abstract
The syndrome of ROU is generally manifested as obvious pain, redness, and swelling of local ulceration area, accompanied by flushed face, red eyes, sore throat, and swollen gums. Traditional Chinese medicine (TCM) doctors believe that "yin deficiency" is one causative factor of ROU. Zhibaidihuang decoction (ZBDHD) is a prescriptively developed receipt, where Anemarrhena asphodeloides and Phellodendri amurensis Cortex are added in the original Liuweidihuang decoction. It is generally used for "yin deficiency" treatment. It can effectively reduce the recurrence of oral ulcers and release the severity of the disease. However, the mechanism of this activity remains to be elucidated. In this study, we found that ZBDHD has a certain therapeutic effect on the pathological changes of oral mucosa. Furthermore, the results of serum metabolomics showed ZBDHD influenced the synthesis and metabolism of certain fatty acids. The results of western blot, immunochemical, and immunofluorescence staining indicate that ZBDHD could increase the expression of Sirt1 and Foxp3 and suppress the expression and acetylation of NF-κB in oral mucosa cells. By screening active ingredients in ZBDHD, we found berberine, as well as other compounds, presenting high fitness of the Sirt1 reactive centre. Therefore, it is possible that ZBDHD can regulate the Sirt1-NF-κB pathway to improve fatty acids metabolism in the body, thereby achieving the effect of treating ROU.
Collapse
|
13
|
Zhou D, Yang F, Lin L, Tang L, Li L, Yang Y, Liu D, Zhang C, Wu T, Wei H, Zhang X, Zhang L. The sirtuin 1 activator SRT1720 alleviated endotoxin-induced fulminant hepatitis in mice. Exp Anim 2021; 70:302-310. [PMID: 33678756 PMCID: PMC8390304 DOI: 10.1538/expanim.20-0014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The metabolic sensor sirtuin 1 (SIRT1) also functions as a checkpoint in inflammation, and SRT1720 is a highly active and selective SIRT1 activator shown to
alleviate inflammatory injury in several recent experimental studies. In the present study, the potential effects and underlying mechanisms of SRT1720 on
lipopolysaccharide (LPS)-induced fulminant hepatitis in D-galactosamine (D-Gal)-sensitized mice were investigated. The results indicated that treatment with
SRT1720 inhibited LPS/D-Gal-induced elevation of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), alleviated the histological abnormalities,
suppressed the induction of tumor necrosis factor alpha (TNF-α) and IL-6, mitigated the phosphorylation of c-Jun N-terminal kinase (JNK), downregulated the
activities of caspase 8, caspase 9 and caspase 3, decreased the level of cleaved caspase 3, reduced the TUNEL-positive cells, and improved the survival rate of
the LPS/D-Gal-exposed mice. These data indicated that treatment with the SIRT1 activator SRT1720 alleviated LPS/D-Gal-induced fulminant hepatitis, which might
be attributed to the suppressive effects of SRT1720 on TNF-α production and the subsequent activation of the apoptosis cascade.
Collapse
Affiliation(s)
- Dan Zhou
- Department of Pathology, Fuling Central Hospital, 2 Gaosuntang Road, Chongqing 408099, P.R. China
| | - Feng Yang
- Department of Gynaecology and Obstetrics, Fuling Central Hospital, 2 Gaosuntang Road, Chongqing 408099, P.R. China
| | - Ling Lin
- Department of Pathophysiology, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, P.R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, P.R. China
| | - Li Tang
- Department of Pathophysiology, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, P.R. China
| | - Longjiang Li
- Department of Pathophysiology, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, P.R. China
| | - Yongqiang Yang
- Department of Pathophysiology, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, P.R. China
| | - Dingrong Liu
- Department of Pathology, Fuling Central Hospital, 2 Gaosuntang Road, Chongqing 408099, P.R. China
| | - Chong Zhang
- Department of Pathology, Fuling Central Hospital, 2 Gaosuntang Road, Chongqing 408099, P.R. China
| | - Tong Wu
- Department of Pathology, Fuling Central Hospital, 2 Gaosuntang Road, Chongqing 408099, P.R. China
| | - Huijie Wei
- Department of Pathology, Fuling Central Hospital, 2 Gaosuntang Road, Chongqing 408099, P.R. China
| | - Xiaoming Zhang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, 188 Tanhualin Road, Wuhan 430061, P.R. China.,Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, 188 Tanhualin Road, Wuhan 430061, P.R. China
| | - Li Zhang
- Department of Pathophysiology, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, P.R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, P.R. China
| |
Collapse
|
14
|
Xiao Z, Liu W, Mu YP, Zhang H, Wang XN, Zhao CQ, Chen JM, Liu P. Pharmacological Effects of Salvianolic Acid B Against Oxidative Damage. Front Pharmacol 2020; 11:572373. [PMID: 33343348 PMCID: PMC7741185 DOI: 10.3389/fphar.2020.572373] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
Salvianolic acid B (Sal B) is one of the main active ingredients of Salvia miltiorrhiza, with strong antioxidant effects. Recent findings have shown that Sal B has anti-inflammatory, anti-apoptotic, anti-fibrotic effects and can promote stem cell proliferation and differentiation, and has a beneficial effect on cardiovascular and cerebrovascular diseases, aging, and liver fibrosis. Reactive oxygen species (ROS) include oxygen free radicals and oxygen-containing non-free radicals. ROS can regulate cell proliferation, survival, death and differentiation to regulate inflammation, and immunity, while Sal B can scavenge oxygen free radicals by providing hydrogen atoms and reduce the production of oxygen free radicals and oxygen-containing non-radicals by regulating the expression of antioxidant enzymes. The many pharmacological effects of Sal B may be closely related to its elimination and inhibition of ROS generation, and Nuclear factor E2-related factor 2/Kelch-like ECH-related protein 1 may be the core link in its regulation of the expression of antioxidant enzyme to exert its antioxidant effect. What is confusing and interesting is that Sal B exhibits the opposite mechanisms in tumors. To clarify the specific target of Sal B and the correlation between its regulation of oxidative stress and energy metabolism homeostasis will help to further understand its role in different pathological conditions, and provide a scientific basis for its further clinical application and new drug development. Although Sal B has broad prospects in clinical application due to its extensive pharmacological effects, the low bioavailability is a serious obstacle to further improving its efficacy in vivo and promoting clinical application. Therefore, how to improve the availability of Sal B in vivo requires the joint efforts of many interdisciplinary subjects.
Collapse
Affiliation(s)
- Zhun Xiao
- Institute of Interdisciplinary Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Liu
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, China
| | - Yong-ping Mu
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, China
| | - Hua Zhang
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-ning Wang
- Institute of Interdisciplinary Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, China
| | - Chang-qing Zhao
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, China
| | - Jia-mei Chen
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, China
| | - Ping Liu
- Institute of Interdisciplinary Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
15
|
Ahmed N, El-Agamy DS, Mohammed GA, Abo-Haded H, Elkablawy M, Ibrahim SRM. Suppression of LPS-Induced Hepato- and Cardiotoxic Effects by Pulicaria petiolaris via NF-κB Dependent Mechanism. Cardiovasc Toxicol 2020; 20:121-129. [PMID: 31273688 DOI: 10.1007/s12012-019-09539-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recently, there is an increasing interest in searching for harmless natural products isolated from plant materials that can be used as beneficial dietary supplements and/or therapeutic drug candidates. The present study aimed to test the potential protective role of Pulicaria petiolaris (PP, Asteraceae) against hepatic and cardiotoxic effects associated with lipopolysaccharide (LPS) injection. PP was given orally for 5 days at two different doses before LPS injection. Results have shown that LPS induced remarkable hepatic and cardiac injurious effects in mice. Hepatic damage was evident through increased serum transaminases, lactate dehydrogenase (LDH), alkaline phosphatase (ALP), and activity. Estimation of high levels of serum creatine kinase-MB (CK-MB) and cardiac troponin I indicated cardiac damage. Histopathological examination of liver and heart confirmed the biochemical results. Increase in oxidative stress along with a depressed antioxidant status of liver and heart were observed in LPS-intoxicated animals. Furthermore, LPS induced activation of nuclear factor-κB (NF-κB) and subsequent elevation of inflammatory cytokines (TNF-α, IL-6). On the other hand, PP treatment successfully safeguards both organs against LPS-induced injury as indicated by the improvement of the biochemical and histopathological parameters. These results suggest that PP ameliorates LPS-induced hepatic and cardiac oxidative injurious effects via antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Nishat Ahmed
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al Madinah Al Munawwarah, 30078, Saudi Arabia
| | - Dina Saad El-Agamy
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al Madinah Al Munawwarah, 30078, Saudi Arabia
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Gamal Abdallah Mohammed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Hany Abo-Haded
- Cardiology Unit, College of Medicine, Taibah University, Al Madinah Al Munawwarah, 30078, Saudi Arabia
| | - Mohamed Elkablawy
- Department of Pathology, College of Medicine, Taibah University, Al-Madinah Al-Munawwarah, 30001, Saudi Arabia
- Department of Pathology, Faculty of Medicine, Menoufia University, Menoufia, 32511, Egypt
| | - Sabrin Ragab Mohamed Ibrahim
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, Taibah University, Al Madinah Al Munawwarah, 30078, Saudi Arabia.
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt.
| |
Collapse
|
16
|
Li Y, Xi Y, Tao G, Xu G, Yang Z, Fu X, Liang Y, Qian J, Cui Y, Jiang T. Sirtuin 1 activation alleviates primary biliary cholangitis via the blocking of the NF-κB signaling pathway. Int Immunopharmacol 2020; 83:106386. [PMID: 32193100 DOI: 10.1016/j.intimp.2020.106386] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 03/05/2020] [Accepted: 03/05/2020] [Indexed: 12/12/2022]
Abstract
This report sought to establish the mechanistic role of sirtuin-1 (Sirt1), a NAD+-dependent deacetylase in the modulation of primary biliary cholangitis (PBC) pathogenesis. 64 PBC patients (diagnosed based on practice guidelines for American Association for the Study of Liver Diseases) and 60 healthy controls were included in this study. Clinically, the mRNA expression level of Sirt1 in macrophages differentiated from peripheral blood mononuclear cells (PBMCs) of PBC subjects substantially decreased when compared with the healthy controls but not in other Sirt family genes (Sirt2-7). Consistent with clinical results, a PBC murine model showed that levels of Sirt1 significantly decreased in the liver and Kupffer cells of mice treated with polyinosinic/polycytidylic acid (poly I:C) for 16 weeks. A TAK1 inhibitor (NG25) prevented the poly I:C-induced Sirt1 protein level decreasing in Kupffer cells but not MAPK inhibitor. Sirt1 activators resveratrol (RSV) and SRT1720 (SRT) ameliorated poly I:C-induced hepatic injury observed via histopathologic analysis and decreased aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels in the PBC murine model. Furthermore, Sirt1 activators significantly reduced pro-inflammatory cytokines levels such as interleukin-1 beta (IL-1β), IL-6, interferon-gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α) in serum in poly I:C-induced mice. In addition, Sirt1 activators significantly inhibited the phosphorylated and acetylated levels of the RelA/p65 subunit of the nuclear transcription factor (NF-κB) but not the interferon regulatory factor (IRF) 3 in poly I:C-injured mice livers. Significantly, RSV improved the interaction between Sirt1 and p65, which may contribute to the decreased activity of NF-κB. In summary, the Sirt1 signaling pathway plays an essential role in the development of PBC and this may represent a novel approach and target for the treatment of PBC.
Collapse
Affiliation(s)
- Yong Li
- Department of Laboratory Medicine, First People's Hospital of Taicang, Taicang Hospital Affiliated to Suzhou University, Taicang 215400, Jiangsu, China
| | - Yanhai Xi
- Department of Spine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Guohua Tao
- Department of Laboratory Medicine, First People's Hospital of Nantong, 226001 Jiangsu, China
| | - Guohua Xu
- Department of Immunology and Microbiology, Institution of Laboratory Medicine of Changshu, Changshu 215500, Jiangsu, China
| | - Zaixing Yang
- Department of Laboratory Medicine, Huangyan Hospital of Wenzhou Medical University, Taizhou First People's Hospital, Zhejiang, China
| | - Xingli Fu
- Jiangsu University Health Science Center, Zhenjiang, Jiangsu, China
| | - Yan Liang
- Department of Laboratory Diagnostics, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Jianping Qian
- Department of Immunology and Microbiology, Institution of Laboratory Medicine of Changshu, Changshu 215500, Jiangsu, China
| | - Yanhong Cui
- Department of Immunology and Microbiology, Institution of Laboratory Medicine of Changshu, Changshu 215500, Jiangsu, China
| | - Tingwang Jiang
- Department of Immunology and Microbiology, Institution of Laboratory Medicine of Changshu, Changshu 215500, Jiangsu, China.
| |
Collapse
|
17
|
Therapeutic benefits of apocynin in mice with lipopolysaccharide/D-galactosamine-induced acute liver injury via suppression of the late stage pro-apoptotic AMPK/JNK pathway. Biomed Pharmacother 2020; 125:110020. [PMID: 32106375 DOI: 10.1016/j.biopha.2020.110020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/14/2022] Open
Abstract
The excessive generation of reactive oxygen species (ROS) plays crucial roles in the development of acute liver injury. Nicotinamide adenine dinucleotide phosphate oxidase (NOX) is responsible for the robust production of ROS under inflammatory circumstance, but the pathological roles of NOX and the pharmacological significance of NOX inhibitor in acute liver injury remains unclear. In the present study, the potential roles of NOX in acute liver injury were investigated in a mouse model with lipopolysaccharide (LPS)/D-galactosamine (D-Gal)-induced acute liver injury. The results indicated that LPS/D-Gal exposure time-dependently increased the level of ROS in liver tissue. Pretreatment with the NOX inhibitor apocynin suppressed LPS/D-Gal induced upregulation of ROS, 8-hydroxy-2-deoxyguanosine (8-OH-dG), protein carbonyl content and thiobarbituric acid reactive substances (TBARS). Pretreatment with apocynin also suppressed LPS/D-Gal-induced elevation of aminotransferase, alleviated histological abnormalities, inhibited the production of pro-inflammatory cytokine tumor necrosis factor α (TNF-α), blocked the activation of caspase cascade, reduced the count of TUNEL-positive cells and prevented LPS/D-Gal-induced mortality. Interestingly, post insult treatment with apocynin also suppressed LPS/D-Gal-induced oxidative stress, hepatocyte apoptosis, liver damage but improved the survival rate. Mechanistically, posttreatment with apocynin prohibited LPS/D-Gal-induced activation of the late stage pro-apoptotic AMP-activated protein kinase (AMPK)/c-Jun N-terminal kinase (JNK) pathway. Post-insult treatment with the antioxidant N-acetylcysteine also resulted in suppressed activation of AMPK/JNK, mitigated apoptosis and alleviated liver injury. These data suggest that NOX-derived ROS might be a crucial late stage detrimental factor in LPS/D-Gal-induced acute liver injury via promoting the activation of the pro-apoptotic AMPK/JNK pathway, and the NOX inhibitor might have important value in the pharmacological intervention of inflammation-base liver damage.
Collapse
|
18
|
Win S, Min RW, Chen CQ, Zhang J, Chen Y, Li M, Suzuki A, Abdelmalek MF, Wang Y, Aghajan M, Aung FW, Diehl AM, Davis RJ, Than TA, Kaplowitz N. Expression of mitochondrial membrane-linked SAB determines severity of sex-dependent acute liver injury. J Clin Invest 2019; 129:5278-5293. [PMID: 31487267 PMCID: PMC6877311 DOI: 10.1172/jci128289] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 08/29/2019] [Indexed: 12/15/2022] Open
Abstract
SH3 domain-binding protein that preferentially associates with Btk (SAB) is an outer-membrane docking protein for JNK-mediated impairment of mitochondrial function. Deletion of Sab in hepatocytes inhibits sustained JNK activation and cell death. The current study demonstrates that an increase in SAB expression enhanced the severity of acetaminophen-induced (APAP-induced) liver injury. Female mice were resistant to liver injury and exhibited markedly decreased hepatic SAB protein expression compared with male mice. The mechanism of SAB repression involved a pathway from ERα to p53 expression that induced miR34a-5p. miR34a-5p targeted the Sab mRNA coding region, thereby repressing SAB expression. Fulvestrant or p53 knockdown decreased miR34a-5p and increased SAB expression in female mice, leading to increased injury from APAP and TNF/galactosamine. In contrast, an ERα agonist increased p53 and miR34a-5p, which decreased SAB expression and hepatotoxicity in male mice. Hepatocyte-specific deletion of miR34a also increased the severity of liver injury in female mice, which was prevented by GalNAc-ASO knockdown of Sab. Similar to mice, premenopausal women expressed elevated levels of hepatic p53 and low levels of SAB, whereas age-matched men expressed low levels of p53 and high levels of SAB, but there was no difference in SAB expression between the sexes in the postmenopausal stage. In conclusion, SAB expression levels determined the severity of JNK-dependent liver injury. Female mice expressed low levels of hepatic SAB protein because of the ERα/p53/miR34a pathway, which repressed SAB expression and accounted for the resistance to liver injury seen in these females.
Collapse
Affiliation(s)
- Sanda Win
- USC Research Center for Liver Disease, Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
| | - Robert W.M. Min
- USC Research Center for Liver Disease, Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
| | - Christopher Q. Chen
- USC Research Center for Liver Disease, Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
| | - Jun Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yibu Chen
- USC Libraries Bioinformatics Service, Norris Medical Library, USC, Los Angeles, California, USA
| | - Meng Li
- USC Libraries Bioinformatics Service, Norris Medical Library, USC, Los Angeles, California, USA
| | - Ayako Suzuki
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Manal F. Abdelmalek
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Ying Wang
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina, USA
| | | | - Filbert W.M. Aung
- USC Research Center for Liver Disease, Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Roger J. Davis
- Howard Hughes Medical Institute and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Tin A. Than
- USC Research Center for Liver Disease, Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
| | - Neil Kaplowitz
- USC Research Center for Liver Disease, Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
| |
Collapse
|
19
|
Guo Q, Chen S, Rao X, Li Y, Pan M, Fu G, Yao Y, Gao X, Tang P, Zhou Y, Xu X, Gao J, Hua G. Inhibition of SIRT1 promotes taste bud stem cell survival and mitigates radiation-induced oral mucositis in mice. Am J Transl Res 2019; 11:4789-4799. [PMID: 31497199 PMCID: PMC6731402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/13/2019] [Indexed: 06/10/2023]
Abstract
Taste loss is one of the debilitating complications in radiation-induced oral mucositis (RIOM), as occurs in head and neck cancer patients undergoing radiotherapy. We report here a radio-mitigation effect of Sirtuin 1 (SIRT1) inhibitors in taste bud organoids and a mouse model of radiation-induced taste bud injury. The organoids, developed from circumvallate (CV) papilla, were irradiated with single dose of X-rays and inhibitors of SIRT1 or SIRT2 were added 24 h later. The survival was evaluated by measuring the number and size of regenerated organoids after irradiation (IR). Oral mucositis (OM) was induced by IR of the oral region of Lgr5-lacZ transgenic mice. The surviving Lgr5+ taste bud stem cells were identified after lacZ-staining and the mucosal ulceration on tongue dorsal surface was determined by histological methods. Results showed that SIRT1 inhibitors (nicotinamide, EX527, salermide and sirtinol), but not SIRT2 inhibitors, significantly improve taste bud organoid survival after IR. Remarkably, administration of nicotinamide (NAM), a recognized inhibitor of SIRT1 to mice 24 h after IR promotes the survival of Lgr5+ taste bud stem cells, resulting in alleviated tongue mucositis. In conclusion, SIRT1 inhibitors promote Lgr5+ taste bud stem cell survival and mitigate RIOM in mice. These observations have important implications for efforts to develop therapeutic strategies against taste dysfunction and mucosal ulceration in RIOM.
Collapse
Affiliation(s)
- Qiang Guo
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Shengzhi Chen
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Xinxin Rao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Yuanchuang Li
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Mengxue Pan
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Guoxiang Fu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Ye Yao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan UniversityShanghai 200032, China
| | - Xiaoxue Gao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Peiyuan Tang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Yi Zhou
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Xiaoya Xu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Jianjun Gao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Guoqiang Hua
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Fudan UniversityShanghai 200032, China
| |
Collapse
|
20
|
Zhai Q, Gong X, Wang C, Zhao J, Zhang H, Tian F, Chen W. Food-borne patulin toxicity is related to gut barrier disruption and can be prevented by docosahexaenoic acid and probiotic supplementation. Food Funct 2019; 10:1330-1339. [PMID: 30741300 DOI: 10.1039/c8fo02292e] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Patulin (PAT) is a mycotoxin widely found in fruits and vegetables. Several reviews and studies have hypothesized that in vivo PAT toxicity is related to gut barrier dysfunction, but evidence for this is not substantial. The goal of the present study was to further demonstrate the role of the gut barrier in food-borne PAT toxicity. In vitro assays showed that PAT exposure induced significant cell death, inhibited the mRNA expressions of tight junction proteins and increased gut permeability in Caco-2 cell monolayers. An acute PAT exposure animal trial reported for the first time an association between PAT-induced disruption of the gut barrier and endotoxemia in mice. Sub-chronic PAT exposure also inhibited the expression of ZO-1 in the gut and induced both intestinal and systematic inflammation in mice. Dietary supplements with previously reported protective effects on the gut barrier, such as docosahexaenoic acid and Lactobacillus plantarum CCFM8610, were able to recover the PAT-induced gut barrier dysfunction and significantly alleviate PAT toxicity in vivo. Another L. plantarum strain, CCFM11, with poor gut barrier modulation ability, failed to exhibit identical protective effects against PAT toxicity to L. plantarum CCFM8610. Our results indicated that PAT-induced disruption of the gut barrier and bacterial translocation may be another toxic mechanism of PAT besides its inherent cytotoxicity. Gut barrier protection may be considered an important target for the prevention of PAT toxicity.
Collapse
Affiliation(s)
- Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| | | | | | | | | | | | | |
Collapse
|
21
|
Yan T, Huang J, Nisar MF, Wan C, Huang W. The Beneficial Roles of SIRT1 in Drug-Induced Liver Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8506195. [PMID: 31354914 PMCID: PMC6636535 DOI: 10.1155/2019/8506195] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/27/2019] [Accepted: 04/24/2019] [Indexed: 02/07/2023]
Abstract
Drug-induced liver injury (DILI) is a major cause of acute liver failure (ALF) as a result of accumulated drugs in the human body metabolized into toxic agents and helps generate heavy oxidative stress, inflammation, and apoptosis, which induces necrosis in hepatocytes and ultimately damages the liver. Sirtuin 1 (SIRT1) is said to have multiple vital roles in cell proliferation, aging, and antistress systems of the human body. The levels of SIRT1 and its activation precisely modulate its critical role in the interaction between multiple step procedures of DILI. The nuclear factor kappa-light-chain-enhancer of activated B cell- (NF-κB-) mediated inflammation signaling pathway, reactive oxygen species (ROS), DNA damage, mitochondrial membrane potential collapse, and endoplasmic reticulum (ER) stress also contribute to aggravate DILI. Apoptosis is regarded as the terminal reaction followed by multiple signaling cascades including caspases, p53, and mitochondrial dysfunction which have been said to contribute in DILI. The SIRT1 activator is regarded as a potential candidate for DILI, because the former could inhibit signaling of p53, NF-κB, and ER stress. On the other hand, overexpression of SIRT1 also enhances the activation of antioxidant responses via Kelch-like ECH-associated protein 1- (Keap1-) nuclear factor- (erythroid-derived 2-) like 2 (Nrf2) signaling. The current manuscript will highlight the mechanism of DILI and the interaction of SIRT1 with various cytoplasmic factors leading to DILI along with the summary of potent SIRT1 agonists.
Collapse
Affiliation(s)
- Tingdong Yan
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jinlong Huang
- The Institute of Infection and Inflammation, Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang, Hubei 443002, China
| | - Muhammad Farrukh Nisar
- Department of Physiology and Biochemistry, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, 63100, Pakistan
| | - Chunpeng Wan
- Jiangxi Key Laboratory for Postharvest Technology and Nondestructive Testing of Fruits & Vegetables, Collaborative Innovation Center of Post-Harvest Key Technology and Quality Safety of Fruits and Vegetables, College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China
| | - Weifeng Huang
- The Institute of Infection and Inflammation, Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang, Hubei 443002, China
| |
Collapse
|
22
|
Generation of H11-albumin-rtTA Transgenic Mice: A Tool for Inducible Gene Expression in the Liver. G3-GENES GENOMES GENETICS 2019; 9:591-599. [PMID: 30591434 PMCID: PMC6385985 DOI: 10.1534/g3.118.200963] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The modification of the mouse genome by site-specific gene insertion of transgenes and other genetic elements allows the study of gene function in different developmental stages and in the pathogenesis of diseases. Here, we generated a “genomic safe harbor” Hipp11 (H11) locus-specific knock-in transgenic mouse line in which the albumin promoter is used to drive the expression of the reverse tetracycline transactivator (rtTA) in the liver. The newly generated H11-albumin-rtTA transgenic mice were bred with tetracycline-operator-Histone-2B-green fluorescent protein (TetO-H2BGFP) mice to assess inducibility and tissue-specificity. Expression of the H2BGFP fusion protein was observed exclusively upon doxycycline (Dox) induction in the liver of H11-albumin-rtTA/TetO-H2BGFP double transgenic mice. To further analyze the ability of the Dox-inducible H11-albumin-rtTA mice to implement conditional DNA recombination, H11-albumin-rtTA transgenic mice were crossed with TetO-Cre and Ai14 mice to generate H11-albumin-rtTA/TetO-Cre/Ai14 triple transgenic mice. We successfully confirmed that the Cre-mediated recombination efficiency was as strong in Dox-induced H11-albumin-rtTA /TetO-Cre/Ai14 mice as in the control albumin-Cre/A14 mice. Finally, to characterize the expression-inducing effects of Dox in H11-albumin-rtTA/TetO-H2BGFP mice in detail, we examined GFP expression in embryos at different developmental stages and found that newly conceived H11-albumin-rtTA/TetO-H2BGFP embryos of Dox-treated pregnant female mice were expressing reporter GFP by E16.5. Our study demonstrates that these new H11-albumin-rtTA transgenic mice are a powerful and efficient tool for the temporally and spatially conditional manipulation of gene expression in the liver, and illustrates how genetic crosses with these new mice enable the generation of complex multi-locus transgenic animals for mechanistic studies.
Collapse
|
23
|
Zhang HY, Wang HL, Zhong GY, Zhu JX. Molecular mechanism and research progress on pharmacology of traditional Chinese medicine in liver injury. PHARMACEUTICAL BIOLOGY 2018; 56:594-611. [PMID: 31070528 PMCID: PMC6282438 DOI: 10.1080/13880209.2018.1517185] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/27/2018] [Accepted: 08/21/2018] [Indexed: 05/09/2023]
Abstract
CONTEXT Liver disease is a common threat to human health, caused by a variety of factors that damage the liver. Recent studies have shown that active ingredients (for example: flavonoids, saponins, acids, phenols, and alkaloids) from Traditional Chinese Medicine (TCM) can have hepatoprotective benefits, which represents an attractive source of drug discovery for treating liver injury. OBJECTIVE We reviewed recent contributions on the chemically induced liver injury, immunological liver damage, alcoholic liver injury, and drug-induced liver injury, in order to summarize the research progress in molecular mechanism and pharmacology of TCM, and provides a comprehensive overview of new TCM treatment strategies for liver disease. MATERIALS AND METHODS Relevant literature was obtained from scientific databases such as Pubmed, Web of Science. and CNKI databases on ethnobotany and ethnomedicines (from January 1980 to the end of May 2018). The experimental studies involving the antihepatic injury role of the active agents from TCM and the underlying mechanisms were identified. The search terms included 'liver injury' or 'hepatic injury', and 'traditional Chinese medicine', or 'herb'. RESULTS A number of studies revealed that the active ingredients of TCM exhibit potential therapeutic benefits against liver injury, while the underlying mechanisms appear to contribute to the regulation of inflammation, oxidant stress, and pro-apoptosis signaling pathways. DISCUSSION AND CONCLUSIONS The insights provided in this review will help further exploration of botanical drugs in the development of liver injury therapy via study on the effective components of TCM.
Collapse
Affiliation(s)
- Hong Yang Zhang
- Research Center of Traditional Chinese Medicine Resources and Minority Medicine, Jiangxi University of Traditional Chinese Medicine, Nan Chang, China
| | - Hong Ling Wang
- Research Center of Traditional Chinese Medicine Resources and Minority Medicine, Jiangxi University of Traditional Chinese Medicine, Nan Chang, China
| | - Guo Yue Zhong
- Research Center of Traditional Chinese Medicine Resources and Minority Medicine, Jiangxi University of Traditional Chinese Medicine, Nan Chang, China
| | - Ji Xiao Zhu
- Research Center of Traditional Chinese Medicine Resources and Minority Medicine, Jiangxi University of Traditional Chinese Medicine, Nan Chang, China
| |
Collapse
|
24
|
Win S, Than TA, Kaplowitz N. The Regulation of JNK Signaling Pathways in Cell Death through the Interplay with Mitochondrial SAB and Upstream Post-Translational Effects. Int J Mol Sci 2018; 19:ijms19113657. [PMID: 30463289 PMCID: PMC6274687 DOI: 10.3390/ijms19113657] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/17/2018] [Accepted: 11/17/2018] [Indexed: 02/08/2023] Open
Abstract
c-Jun-N-terminal kinase (JNK) activity plays a critical role in modulating cell death, which depends on the level and duration of JNK activation. The kinase cascade from MAPkinase kinase kinase (MAP3K) to MAPkinase kinase (MAP2K) to MAPKinase (MAPK) can be regulated by a number of direct and indirect post-transcriptional modifications, including acetylation, ubiquitination, phosphorylation, and their reversals. Recently, a JNK-mitochondrial SH3-domain binding protein 5 (SH3BP5/SAB)-ROS activation loop has been elucidated, which is required to sustain JNK activity. Importantly, the level of SAB expression in the outer membrane of mitochondria is a major determinant of the set-point for sustained JNK activation. SAB is a docking protein and substrate for JNK, leading to an intramitochondrial signal transduction pathway, which impairs electron transport and promotes reactive oxygen species (ROS) release to sustain the MAPK cascade.
Collapse
Affiliation(s)
- Sanda Win
- Division of Gastrointestinal and Liver Disease, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Tin Aung Than
- Division of Gastrointestinal and Liver Disease, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Neil Kaplowitz
- Division of Gastrointestinal and Liver Disease, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
25
|
Fan K, Lin L, Ai Q, Wan J, Dai J, Liu G, Tang L, Yang Y, Ge P, Jiang R, Zhang L. Lipopolysaccharide-Induced Dephosphorylation of AMPK-Activated Protein Kinase Potentiates Inflammatory Injury via Repression of ULK1-Dependent Autophagy. Front Immunol 2018; 9:1464. [PMID: 29988556 PMCID: PMC6026648 DOI: 10.3389/fimmu.2018.01464] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 06/12/2018] [Indexed: 12/14/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is a crucial metabolic regulator with profound modulatory activities on inflammation. Although the anti-inflammatory benefits of AMPK activators were well documented in experimental studies, the pathological significance of endogenous AMPK in inflammatory disorders largely remains unknown. This study investigated the phosphorylation status of endogenous AMPK and the potential roles of AMPK in mice with lipopolysaccharide (LPS)-induced lethal inflammation. The results indicated that LPS dose-dependently decreased the phosphorylation level of AMPK and its target protein acetyl-CoA carboxylase (ACC). Reactivation of AMPK with the AMPK activator A-769662 suppressed LPS-induced elevation of interleukin 6, alleviated histological abnormalities in lung and improved the survival of LPS-challenged mice. Treatment with A-769662 restored LPS-induced suppression of autophagy, inhibition of autophagy by 3-MA reversed the beneficial effects of A-769662. Treatment with A-769662 suppressed LPS-induced activation of mammalian target of rapamycin (mTOR), co-administration of mTOR activator abolished the beneficial effects of A-769662, and the suppressive effects of A-769662 on uncoordinated-51-like kinase 1 (ULK1) phosphorylation. Inhibition of ULK1 removed the beneficial effects of A-769662. These data indicated that LPS-induced dephosphorylation of AMPK could result in weakened inhibition of mTOR and repression of ULK1-dependent autophagy, which might potentiate the development of LPS-induced inflammatory injury. These data suggest that pharmacological restoration of AMPK activation might be a beneficial approach for the intervention of inflammatory disorders.
Collapse
Affiliation(s)
- Kerui Fan
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Ling Lin
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Qing Ai
- Department of Physiology, Chongqing Medical University, Chongqing, China
| | - Jingyuan Wan
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Jie Dai
- Hospital of Chongqing University of Arts and Sciences, Chongqing, China
| | - Gang Liu
- Department of Emergency, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Li Tang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Yongqiang Yang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Pu Ge
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Rong Jiang
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Li Zhang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| |
Collapse
|
26
|
Chen S, Ye J, Chen X, Shi J, Wu W, Lin W, Lin W, Li Y, Fu H, Li S. Valproic acid attenuates traumatic spinal cord injury-induced inflammation via STAT1 and NF-κB pathway dependent of HDAC3. J Neuroinflammation 2018; 15:150. [PMID: 29776446 PMCID: PMC5960086 DOI: 10.1186/s12974-018-1193-6] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/08/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Microglial polarization with M1/M2 phenotype shifts and the subsequent neuroinflammatory responses are vital contributing factors for spinal cord injury (SCI)-induced secondary injury. Nuclear factor-κB (NF-κB) is considered the central transcription factor of inflammatory mediators, which plays a crucial role in microglial activation. Lysine acetylation of STAT1 seems necessary for NF-kB pathway activity, as it is regulated by histone deacetylases (HDACs). There have been no studies that have explained if HDAC inhibition by valproic acid (VPA) affects the NF-κB pathway via acetylation of STAT1 dependent of HDAC activity in the microglia-mediated central inflammation following SCI. We investigated the potential molecular mechanisms that focus on the phenotypic transition of microglia and the STAT1-mediated NF-κB acetylation after a VPA treatment. METHODS The Basso-Beattie-Bresnahan locomotion scale, the inclined plane test, the blood-spinal cord barrier, and Nissl staining were employed to determine the neuroprotective effects of VPA treatment after SCI. Assessment of microglia polarization and pro-inflammatory markers, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and interferon (INF)-γ was used to evaluate the neuroinflammatory responses and the anti-inflammatory effects of VPA treatment. Immunofluorescent staining and Western blot analysis were used to detect HDAC3 nuclear translocation, activity, and NF-κB signaling pathway activation to evaluate the effects of VPA treatment. The impact of STAT1 acetylation on NF-kB pathway and the interaction between STAT1 and NF-kB were assessed to evaluate anti-inflammation effects of VPA treatment and also whether these effects were dependent on a STAT1/NF-κB pathway to gain further insight into the mechanisms underlying the development of the neuroinflammatory response after SCI. RESULTS The results showed that the VPA treatment promoted the phenotypic shift of microglia from M1 to M2 phenotype and inhibited microglial activation, thus reducing the SCI-induced inflammatory factors. The VPA treatment upregulation of the acetylation of STAT1/NF-κB pathway was likely caused by the HDAC3 translocation to the nucleus and activity. These results indicated that the treatment with the VPA suppressed the expression and the activity of HDAC3 and enhanced STAT1, as well as NF-κB p65 acetylation following a SCI. The acetylation status of NF-kB p65 and the complex with NF-κB p65 and STAT1 inhibited the NF-kB p65 transcriptional activity and attenuated the microglia-mediated central inflammatory response following SCI. CONCLUSIONS These results suggested that the VPA treatment attenuated the inflammatory response by modulating microglia polarization through STAT1-mediated acetylation of the NF-κB pathway, dependent of HDAC3 activity. These effects led to neuroprotective effects following SCI.
Collapse
Affiliation(s)
- Shoubo Chen
- Department of Orthopaedics, The Second Affiliated Hospital, Fujian Medical Universityz, Quanzhou, 362000, Fujian Province, China
| | - Jingfang Ye
- Department of nursing faculty, Quanzhou Medical College, Quanzhou, 362000, Fujian Province, China
| | - Xiangrong Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian Province, China.
| | - Jinnan Shi
- Department of Orthopaedics, The Second Affiliated Hospital, Fujian Medical Universityz, Quanzhou, 362000, Fujian Province, China
| | - Wenhua Wu
- Department of Orthopaedics, The Second Affiliated Hospital, Fujian Medical Universityz, Quanzhou, 362000, Fujian Province, China
| | - Wenping Lin
- Department of Orthopaedics, The Second Affiliated Hospital, Fujian Medical Universityz, Quanzhou, 362000, Fujian Province, China
| | - Weibin Lin
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian Province, China
| | - Yasong Li
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian Province, China
| | - Huangde Fu
- Department of Neurosurgery, Affiliated Hospital of YouJiang Medical University for Nationalities, Baise, 533000, Guangxi Province, China
| | - Shun Li
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan Province, China.
| |
Collapse
|
27
|
Nakamura K, Zhang M, Kageyama S, Ke B, Fujii T, Sosa RA, Reed EF, Datta N, Zarrinpar A, Busuttil RW, Araujo JA, Kupiec-Weglinski JW. Macrophage heme oxygenase-1-SIRT1-p53 axis regulates sterile inflammation in liver ischemia-reperfusion injury. J Hepatol 2017; 67:1232-1242. [PMID: 28842295 PMCID: PMC5884687 DOI: 10.1016/j.jhep.2017.08.010] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/03/2017] [Accepted: 08/08/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Hepatic ischemia-reperfusion injury (IRI), characterized by exogenous antigen-independent local inflammation and hepatocellular death, represents a risk factor for acute and chronic rejection in liver transplantation. We aimed to investigate the molecular communication involved in the mechanism of liver IRI. METHODS We analyzed human liver transplants, primary murine macrophage cell cultures and IR-stressed livers in myeloid-specific heme oxygenase-1 (HO-1) gene mutant mice, for anti-inflammatory and cytoprotective functions of macrophage-specific HO-1/SIRT1 (sirtuin 1)/p53 (tumor suppressor protein) signaling. RESULTS Decreased HO-1 expression in human post-reperfusion liver transplant biopsies correlated with a deterioration in hepatocellular function (serum ALT; p<0.05) and inferior patient survival (p<0.05). In the low HO-1 liver transplant biopsy group, SIRT1/Arf (alternative reading frame)/p53/MDM2 (murine double minute 2) expression levels decreased (p<0.05) while cleaved caspase 3 and frequency of TUNEL+cells simultaneously increased (p<0.05). Immunofluorescence showed macrophages were the principal source of HO-1 in human and mouse IR-stressed livers. In vitro macrophage cultures revealed that HO-1 induction positively regulated SIRT1 signaling, whereas SIRT1-induced Arf inhibited ubiquitinating activity of MDM2 against p53, which in turn attenuated macrophage activation. In a murine model of hepatic warm IRI, myeloid-specific HO-1 deletion lacked SIRT1/p53, exacerbated liver inflammation and IR-hepatocellular death, whereas adjunctive SIRT1 activation restored p53 signaling and rescued livers from IR-damage. CONCLUSION This bench-to-bedside study identifies a new class of macrophages activated via the HO-1-SIRT1-p53 signaling axis in the mechanism of hepatic sterile inflammation. This mechanism could be a target for novel therapeutic strategies in liver transplant recipients. LAY SUMMARY Post-transplant low macrophage HO-1 expression in human liver transplants correlates with reduced hepatocellular function and survival. HO-1 regulates macrophage activation via the SIRT1-p53 signaling network and regulates hepatocellular death in liver ischemia-reperfusion injury. Thus targeting this pathway in liver transplant recipients could be of therapeutic benefit.
Collapse
Affiliation(s)
- Kojiro Nakamura
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Min Zhang
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Shoichi Kageyama
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Bibo Ke
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Takehiro Fujii
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Rebecca A. Sosa
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Nakul Datta
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Ali Zarrinpar
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Ronald W. Busuttil
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Jesus A. Araujo
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA,Corresponding authors. Addresses: Dumont-UCLA Transplant Center, 10833 Le Conte Ave, 77-120 CHS, Los Angeles, CA 90095, USA. Tel.: +1 (310) 825 4196; fax: +1 (310) 267 2358 (J.W. Kupiec-Weglinski) and UCLA Division of Cardiology, 10833 Le Conte Ave, CHS 43-264, Los Angeles, CA 90095, USA. Tel.: +1 (310) 825 3222; fax: +1 (310) 206 9133 (J.A. Araujo). (J.A. Araujo), (J.W. Kupiec-Weglinski)
| | - Jerzy W. Kupiec-Weglinski
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA,Corresponding authors. Addresses: Dumont-UCLA Transplant Center, 10833 Le Conte Ave, 77-120 CHS, Los Angeles, CA 90095, USA. Tel.: +1 (310) 825 4196; fax: +1 (310) 267 2358 (J.W. Kupiec-Weglinski) and UCLA Division of Cardiology, 10833 Le Conte Ave, CHS 43-264, Los Angeles, CA 90095, USA. Tel.: +1 (310) 825 3222; fax: +1 (310) 206 9133 (J.A. Araujo). (J.A. Araujo), (J.W. Kupiec-Weglinski)
| |
Collapse
|
28
|
Zhu M, Zhou X, Zhao J. Quercetin prevents alcohol-induced liver injury through targeting of PI3K/Akt/nuclear factor-κB and STAT3 signaling pathway. Exp Ther Med 2017; 14:6169-6175. [PMID: 29285175 DOI: 10.3892/etm.2017.5329] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 10/17/2017] [Indexed: 12/16/2022] Open
Abstract
Quercetin is a type of flavonoid compound, which has potent antioxidant and anti-inflammatory activities, capable of treating a variety of diseases including neurodegenerative diseases, tumors, diabetes and obesity. The present study selected alcohol-induced liver injury model mice and aimed at studying the protective role of quercetin in preventing alcohol-induced liver injury. In alcohol-induced liver injury mice treated with quercetin, it was demonstrated that levels of aspartate transaminase, alanine transaminase, total bilirubin and triglyceride were reduced. In addition to this, the activities of the antioxidant enzymes superoxide dismutase and glutathione peroxidase were increased, malondialdehyde was inhibited, and interleukin (IL)-1β, IL-6, IL-10 and inducible nitric oxide synthase were suppressed. Quercetin additionally suppressed the protein expression levels of B-cell lymphoma (Bcl)-2, Bcl-2 associated X apoptosis regulator, Caspase-3, poly ADP-ribose polymerase, and signal transducer and activator of transcription (STAT) 3 phosphorylation, nuclear factor (NF)-κB and protein kinase B (Akt) phosphorylation levels in alcohol-induced liver injured mice. These results suggested that the protective role of quercetin prevents alcohol-induced liver injury through the phosphoinositide 3-kinase/Akt/NF-κB and STAT3 pathway.
Collapse
Affiliation(s)
- Minglin Zhu
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Xuefeng Zhou
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jinping Zhao
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
29
|
Nakamura K, Kageyama S, Ke B, Fujii T, Sosa RA, Reed EF, Datta N, Zarrinpar A, Busuttil RW, Kupiec-Weglinski JW. Sirtuin 1 attenuates inflammation and hepatocellular damage in liver transplant ischemia/Reperfusion: From mouse to human. Liver Transpl 2017; 23:1282-1293. [PMID: 28719070 PMCID: PMC5705033 DOI: 10.1002/lt.24821] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/28/2017] [Accepted: 07/09/2017] [Indexed: 12/12/2022]
Abstract
Hepatic ischemia/reperfusion injury (IRI), an inevitable antigen-independent inflammation response in cadaveric liver transplantation, correlates with poor early graft function, rejection episodes, and contributes to donor organ shortage. Sirtuin 1 (SIRT1) is a histone deacetylase that may regulate inflammatory cell activity and manage liver function in IRI, though its functional role and clinical relevance remains to be elucidated. We investigated the efficacy of SIRT1 activation in a murine liver IRI model and verified the concept of putative SIRT1-mediated hepatoprotection in clinical liver transplantation. In the experimental arm, mice were subjected to 90 minutes of liver partial warm ischemia followed by 6 hours of reperfusion with or without adjunctive SIRT1 activation in vivo (resveratrol [Res]). In parallel, bone marrow-derived macrophage (BMDM) or spleen lymphocyte cultures were treated with Res. In the clinical arm, liver biopsies from 21 adult primary liver transplant patients (2 hours after reperfusion) were divided into "low" (n = 11) versus "high" (n = 10) SIRT1 expression groups, assessed by Western blots. Treatment with Res attenuated murine liver IRI while up-regulating SIRT1, suppressing leukocyte infiltration, and decreasing proinflammatory cytokine programs. SIRT1 silencing (small interfering RNA) in BMDM cultures enhanced inflammatory cytokine programs, whereas addition of Res decreased proinflammatory response in a SIRT1-dependent manner. In addition, Res decreased interferon γ production in liver-infiltrating and spleen lymphocyte cultures. Human liver transplants with high SIRT1 levels showed improved hepatocellular function and superior survival (P = 0.04), accompanied by lower proinflammatory cytokine profile. In conclusion, our translational study is the first to identify SIRT1 as a regulator of hepatocellular function in human liver transplant recipients under ischemia/reperfusion stress. By targeting innate and adaptive immune activation, manipulation of SIRT1 signaling should be considered as a novel means to combat inflammation in liver transplantation. Liver Transplantation 23 1282-1293 2017 AASLD.
Collapse
Affiliation(s)
- Kojiro Nakamura
- Dumont-UCLA Transplant Center, Department of Surgery, Division of Liver and Pancreas Transplantation, University of California, Los Angeles, Los Angeles, CA
| | - Shoichi Kageyama
- Dumont-UCLA Transplant Center, Department of Surgery, Division of Liver and Pancreas Transplantation, University of California, Los Angeles, Los Angeles, CA
| | - Bibo Ke
- Dumont-UCLA Transplant Center, Department of Surgery, Division of Liver and Pancreas Transplantation, University of California, Los Angeles, Los Angeles, CA
| | - Takehiro Fujii
- Dumont-UCLA Transplant Center, Department of Surgery, Division of Liver and Pancreas Transplantation, University of California, Los Angeles, Los Angeles, CA
| | - Rebecca A. Sosa
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA
| | - Nakul Datta
- Dumont-UCLA Transplant Center, Department of Surgery, Division of Liver and Pancreas Transplantation, University of California, Los Angeles, Los Angeles, CA
| | - Ali Zarrinpar
- Dumont-UCLA Transplant Center, Department of Surgery, Division of Liver and Pancreas Transplantation, University of California, Los Angeles, Los Angeles, CA
| | - Ronald W. Busuttil
- Dumont-UCLA Transplant Center, Department of Surgery, Division of Liver and Pancreas Transplantation, University of California, Los Angeles, Los Angeles, CA
| | - Jerzy W. Kupiec-Weglinski
- Dumont-UCLA Transplant Center, Department of Surgery, Division of Liver and Pancreas Transplantation, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
30
|
Huang J, Tian R, Yang Y, Jiang R, Dai J, Tang L, Zhang L. The SIRT1 inhibitor EX-527 suppresses mTOR activation and alleviates acute lung injury in mice with endotoxiemia. Innate Immun 2017; 23:678-686. [PMID: 28952835 DOI: 10.1177/1753425917733531] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
It is generally regarded that Sirtuin 1 (SIRT1), a longevity factor in mammals, acts as a negative regulator of inflammation. However, recent studies also found that SIRT1 might be a detrimental factor under certain inflammatory circumstance. In this study, the potential pathophysiological roles and the underlying mechanisms of SIRT1 in a mouse model with endotoxemia-associated acute lung injury were investigated. The results indicated that treatment with the selective SIRT1 inhibitor EX-527 suppressed LPS-induced elevation of TNF-α and IL-6 in plasma. Treatment with EX-527 attenuated LPS-induced histological abnormalities in lung tissue, which was accompanied with decreased myeloperoxidase level and suppressed induction of tissue factor and plasminogen activator inhibitor-1. Treatment with EX-527 also suppressed LPS-induced phosphorylation of eukaryotic translation initiation factor-binding protein 1 (4E-BP1). Co-administration of a mammalian target of rapamycin (mTOR) activator 3-benzyl-5-[(2-nitrophenoxy) methyl]-dihydrofuran-2 (3H)-one (3BDO) abolished the inhibitory effects of EX-527 on 4E-BP1 phosphorylation. Meanwhile, the inhibitory effects of EX-527 on IL-6 induction and the beneficial effects of EX-527 on lung injury were partially reversed by 3BDO. This study suggests that selective inhibition of SIRT1 by EX-527 might alleviate endotoxemia-associated acute lung injury partially via suppression of mTOR, which implies that SIRT1 selective inhibitors might have potential value for the pharmacological intervention of inflammatory lung injury.
Collapse
Affiliation(s)
- Jing Huang
- 1 Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Rui Tian
- 2 Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yongqiang Yang
- 1 Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Rong Jiang
- 3 Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Jie Dai
- 4 Hospital of Chongqing University of Arts and Sciences, Chongqing, China
| | - Li Tang
- 1 Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Li Zhang
- 1 Department of Pathophysiology, Chongqing Medical University, Chongqing, China.,3 Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
31
|
Guo Y, Li JX, Mao TY, Zhao WH, Liu LJ, Wang YL. Targeting Sirt1 in a rat model of high-fat diet-induced non-alcoholic fatty liver disease: Comparison of Gegen Qinlian decoction and resveratrol. Exp Ther Med 2017; 14:4279-4287. [PMID: 29104641 PMCID: PMC5658732 DOI: 10.3892/etm.2017.5076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 06/29/2017] [Indexed: 12/19/2022] Open
Abstract
The present study aimed to explore the mechanism of action of Gegen Qinlian decoction (GGQLD) in experimental non-alcoholic fatty liver disease (NAFLD). A total of 30 rats were randomly divided into five groups: The chow, model, high- and low-dose GGQLD (GGQLD-H and GGQLD-L, respectively) and resveratrol (Resl) groups, and were treated with saline, GGQLD and Resl when a model of high-fat diet (HFD)-induced NAFLD was established. Blood lipid and liver enzymes were detected following treatment for 8 weeks and liver tissue pathology was observed using Oil Red O and haematoxylin and eosin staining. Furthermore, the liver protein and mRNA expression of sirtuin (Sirt)1, peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α and nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) were measured using western blotting and reverse transcription-quantitative polymerase chain reaction. Compared with the chow group, the model group demonstrated significantly increased serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels (P<0.01). GGQLD doses and Resl attenuated the elevated serum ALT and AST levels. GGQLD-H and Resl significantly increased the serum high-density lipoprotein cholesterol level compared with that in the model group (P<0.01), while GGQLD-L and Resl significantly decreased serum low-density lipoprotein cholesterol levels (P<0.01). The GGQLDs and Resl groups revealed an evident improvement in Sirt1 protein and mRNA expression. Although GGQLD and Resl significantly decreased NF-κB gene expression compared with the model group (P<0.01), the effect on NF-κB protein expression was not significant. Furthermore, the PGC-1α gene and protein expression in the HFD rat group slightly decreased compared to the levels in the chow group, but the decrease was insignificant. However, an evident increase in PGC-1α mRNA expression was observed in the GGQLD-H group compared with the model group (P<0.01). Histological staining revealed that GGQLD and Resl decreased the lipid droplets in hepatocytes and normalized steatosis in rats fed with a HFD. The results indicated that GGQLD treatment may be a potent strategy for managing NAFLD by managing lipid metabolism and inflammatory and histological abnormalities by triggering the Sirt1 pathway.
Collapse
Affiliation(s)
- Yi Guo
- Department of Gastroenterology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, P.R. China.,Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Jun-Xiang Li
- Department of Gastroenterology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, P.R. China
| | - Tang-You Mao
- Department of Gastroenterology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, P.R. China.,Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Wei-Han Zhao
- Department of Gastroenterology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, P.R. China.,Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Li-Juan Liu
- Department of Gastroenterology of Traditional Chinese Medicine, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Yun-Liang Wang
- Department of Gastroenterology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, P.R. China
| |
Collapse
|
32
|
NAD + augmentation ameliorates acute pancreatitis through regulation of inflammasome signalling. Sci Rep 2017; 7:3006. [PMID: 28592850 PMCID: PMC5462749 DOI: 10.1038/s41598-017-03418-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 04/27/2017] [Indexed: 12/12/2022] Open
Abstract
Acute pancreatitis (AP) is a complicated disease without specific drug therapy. The cofactor nicotinamide adenine dinucleotide (NAD+) is an important regulator of cellular metabolism and homeostasis. However, it remains unclear whether modulation of NAD+ levels has an impact on caerulein-induced AP. Therefore, in this study, we investigated the effect of increased cellular NAD+ levels on caerulein-induced AP. We demonstrated for the first time that the activities and expression of SIRT1 were suppressed by reduction of intracellular NAD+ levels and the p53-microRNA-34a pathway in caerulein-induced AP. Moreover, we confirmed that the increase of cellular NAD+ by NQO1 enzymatic action using the substrate β-Lapachone suppressed caerulein-induced AP with down-regulating TLR4-mediated inflammasome signalling, and thereby reducing the inflammatory responses and pancreatic cell death. These results suggest that pharmacological stimulation of NQO1 could be a promising therapeutic strategy to protect against pathological tissue damage in AP.
Collapse
|