1
|
Yao Z, Lu Y, Wang P, Chen Z, Zhou L, Sang X, Yang Q, Wang K, Hao M, Cao G. The role of JNK signaling pathway in organ fibrosis. J Adv Res 2024:S2090-1232(24)00431-4. [PMID: 39366483 DOI: 10.1016/j.jare.2024.09.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Fibrosis is a tissue damage repair response caused by multiple pathogenic factors which could occur in almost every apparatus and leading to the tissue structure damage, physiological abnormality, and even organ failure until death. Up to now, there is still no specific drugs or strategies can effectively block or changeover tissue fibrosis. JNKs, a subset of mitogen-activated protein kinases (MAPK), have been reported that participates in various biological processes, such as genetic expression, DNA damage, and cell activation/proliferation/death pathways. Increasing studies indicated that abnormal regulation of JNK signal pathway has strongly associated with tissue fibrosis. AIM OF REVIEW This review designed to sum up the molecular mechanism progresses in the role of JNK signal pathway in organ fibrosis, hoping to provide a novel therapy strategy to tackle tissue fibrosis. KEY SCIENTIFIC CONCEPTS OF REVIEW Recent evidence shows that JNK signaling pathway could modulates inflammation, immunoreaction, oxidative stress and Multiple cell biological functions in organ fibrosis. Therefore, targeting the JNK pathway may be a useful strategy in cure fibrosis.
Collapse
Affiliation(s)
- Zhouhui Yao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yandan Lu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Pingping Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ziyan Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Licheng Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xianan Sang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qiao Yang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Kuilong Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Min Hao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Songyang Research Institute of Zhejiang Chinese Medical University, Songyang, 323400, China.
| | - Gang Cao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
2
|
Wenbo Z, Jianwei H, Hua L, Lei T, Guijuan C, Mengfei T. The potential of flavonoids in hepatic fibrosis: A comprehensive review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155932. [PMID: 39146877 DOI: 10.1016/j.phymed.2024.155932] [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: 04/07/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Hepatic fibrosis is a pathophysiological process of extracellular matrix abnormal deposition induced by multiple pathogenic factors. Currently, there is still a lack of effective and non-toxic drugs for treating fibrosis in clinic. Flavonoids are polyphenolic compounds synthesized in plants and modern pharmacological studies confirmed flavonoids exhibit potent hepatoprotective effect. PURPOSE Summarize literature to elaborate the mechanism of HF and evaluate the potential of flavonoids in HF, aiming to provide a new perspective for future research. METHODS The literatures about hepatic fibrosis and flavonoids are collected via a series of scientific search engines including Google Scholar, Elsevier, PubMed, CNKI, WanFang, SciFinder and Web of Science database. The key words are "flavonoids", "hepatic fibrosis", "pharmacokinetic", "toxicity", "pathogenesis" "traditional Chinese medicine" and "mechanism" as well as combination application. RESULTS Phytochemical and pharmacological studies revealed that about 86 natural flavonoids extracted from Chinese herbal medicines possess significantly anti-fibrosis effect and the mechanisms maybe through anti-inflammatory, antioxidant, inhibiting hepatic stellate cells activation and clearing activated hepatic stellate cells. CONCLUSIONS This review summarizes the flavonoids which are effective in HF and the mechanisms in vivo and in vitro. However, fewer studies are focused on the pharmacokinetics of flavonoids in HF model and most studies are limited to preclinical studies, therefore there is no reliable data from clinical trials for the development of new drugs. Further in-depth research related it can be conducted to improve the bioavailability of flavonoids and serve the development of new drugs.
Collapse
Affiliation(s)
- Zhu Wenbo
- Faculty of Chinese Medicine, Jiangsu College of Nursing, Huaian 223001, China.
| | - Han Jianwei
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150000, China
| | - Liu Hua
- NHC Key Laboratory of Birth Defect for Research and Prevention (Hunan Provincial Maternal and Child Health Care Hospital), Changsha, Hunan 410008, China
| | - Tang Lei
- Faculty of Chinese Medicine, Jiangsu College of Nursing, Huaian 223001, China
| | - Chen Guijuan
- Faculty of Chinese Medicine, Jiangsu College of Nursing, Huaian 223001, China
| | - Tian Mengfei
- Faculty of Chinese Medicine, Jiangsu College of Nursing, Huaian 223001, China
| |
Collapse
|
3
|
Behera BP, Mishra SR, Mahapatra KK, Patil S, Efferth T, Bhutia SK. SIRT1-activating butein inhibits arecoline-induced mitochondrial dysfunction through PGC1α and MTP18 in oral cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155511. [PMID: 38723523 DOI: 10.1016/j.phymed.2024.155511] [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: 01/12/2024] [Revised: 02/20/2024] [Accepted: 03/01/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Mitochondrial dysfunction associated with mitochondrial DNA mutations, enzyme defects, generation of ROS, and altered oxidative homeostasis is known to induce oral carcinogenesis during exposure to arecoline. Butein, a natural small molecule from Butea monosperma, possesses anti-inflammatory, anti-diabetic, and anti-cancer effects. However, the role of butein in the mitochondrial quality control mechanism has not been illuminated clearly. PURPOSE This study aimed to explore the role of butein in preserving mitochondrial quality control during arecoline-induced mitochondrial dysfunction in oral cancer to curtail the early onset of carcinogenesis. METHODS Cell viability was evaluated by MTT assay. The relative protein expressions were determined by western blotting. Immunofluorescence and confocal imaging were used to analyze the relative fluorescence and co-localization of proteins. Respective siRNAs were used to examine the knockdown-based studies. RESULTS Butein, in the presence of arecoline, significantly caused a decrease in mitochondrial hyperpolarization and ROS levels in oral cancer cells. Mechanistically, we found an increase in COXIV, TOM20, and PGC1α expression during butein treatment, and inhibition of PGC1α blunted mitochondrial biogenesis and decreased the mitochondrial pool. Moreover, the fission protein MTP18, and its molecular partners DRP1 and MFF were dose-dependently increased during butein treatment to maintain mitochondria mass. In addition, we also found increased expression of various mitophagy proteins, including PINK1, Parkin, and LC3 during butein treatment, suggesting the clearance of damaged mitochondria to maintain a healthy mitochondrial pool. Interestingly, butein increased the activity of SIRT1 to enhance the functional mitochondrial pool, and inhibition of SIRT1 found to reduce the mitochondrial levels, as evident from the decrease in the expression of PGC1α and MTP18 in oral cancer cells. CONCLUSION Our study proved that SIRT1 maintains a functional mitochondrial pool through PGC1α and MTP18 for biogenesis and fission of mitochondria during arecoline exposure and could decrease the risk of mitochondria dysfunctionality associated with the onset of oral carcinogenesis.
Collapse
Affiliation(s)
- Bishnu Prasad Behera
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, 769008, Odisha, India
| | - Soumya Ranjan Mishra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, 769008, Odisha, India
| | - Kewal Kumar Mahapatra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, 769008, Odisha, India; Current affiliation: Department of Agriculture and Allied Sciences (Zoology), C. V. Raman Global University, Bhubaneswar, 752054, Odisha, India
| | - Shankargouda Patil
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, 84095, UT, USA
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, 769008, Odisha, India.
| |
Collapse
|
4
|
Liao W, Li Y, Liu J, Mou Y, Zhao M, Liu J, Zhang T, Sun Q, Tang J, Wang Z. Homotherapy for heteropathy: therapeutic effect of Butein in NLRP3-driven diseases. Cell Commun Signal 2024; 22:315. [PMID: 38849890 PMCID: PMC11158000 DOI: 10.1186/s12964-024-01695-7] [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: 11/30/2023] [Accepted: 06/02/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Aberrant inflammatory responses drive the initiation and progression of various diseases, and hyperactivation of NLRP3 inflammasome is a key pathogenetic mechanism. Pharmacological inhibitors of NLRP3 represent a potential therapy for treating these diseases but are not yet clinically available. The natural product butein has excellent anti-inflammatory activity, but its potential mechanisms remain to be investigated. In this study, we aimed to evaluate the ability of butein to block NLRP3 inflammasome activation and the ameliorative effects of butein on NLRP3-driven diseases. METHODS Lipopolysaccharide (LPS)-primed bone-marrow-derived macrophages were pretreated with butein and various inflammasome stimuli. Intracellular potassium levels, ASC oligomerization and reactive oxygen species production were also detected to evaluate the regulatory mechanisms of butein. Moreover, mouse models of LPS-induced peritonitis, dextran sodium sulfate-induced colitis, and high-fat diet-induced non-alcoholic steatohepatitis were used to test whether butein has protective effects on these NLRP3-driven diseases. RESULTS Butein blocks NLRP3 inflammasome activation in mouse macrophages by inhibiting ASC oligomerization, suppressing reactive oxygen species production, and upregulating the expression of the antioxidant pathway nuclear factor erythroid 2-related factor 2 (Nrf2). Importantly, in vivo experiments demonstrated that butein administration has a significant protective effect on the mouse models of LPS-induced peritonitis, dextran sodium sulfate-induced colitis, and high-fat diet-induced non-alcoholic steatohepatitis. CONCLUSION Our study illustrates the connotation of homotherapy for heteropathy, i.e., the application of butein to broaden therapeutic approaches and treat multiple inflammatory diseases driven by NLRP3.
Collapse
Affiliation(s)
- Wenhao Liao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yuchen Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jingwen Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yu Mou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Mei Zhao
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Juan Liu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Tianxin Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Qin Sun
- National Traditional Chinese Medicine Clinical Research Base of the Affiliated Traditional Chinese Medicine, Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jianyuan Tang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Zhilei Wang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| |
Collapse
|
5
|
Kim S, Yoon H, Park SK. Butein Increases Resistance to Oxidative Stress and Lifespan with Positive Effects on the Risk of Age-Related Diseases in Caenorhabditis elegans. Antioxidants (Basel) 2024; 13:155. [PMID: 38397753 PMCID: PMC10886231 DOI: 10.3390/antiox13020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Butein is a flavonoid found in many plants, including dahlia, butea, and coreopsis, and has both antioxidant and sirtuin-activating activities. In light of the postulated role of free radicals in aging, we examined the effects of butein on aging and on genetic or nutritional models of age-related diseases in Caenorhabditis elegans. Butein showed radical scavenging activity and increased resistance to oxidative stress in Caenorhabditis elegans. The mean lifespan of Caenorhabditis elegans was significantly increased by butein, from 22.7 days in the untreated control to 25.0 days in the butein-treated group. However, the lifespan-extending effect of butein was accompanied by reduced production of progeny as a trade-off. Moreover, the age-related decline in motility was delayed by butein supplementation. Genetic analysis showed that the lifespan-extending effect of butein required the autophagic protein BEC-1 and the transcription factor DAF-16 to regulate stress response and aging. At the genetic level, expression of the DAF-16 downstream target genes hsp-16.2 and sod-3 was induced in butein-treated worms. Butein additionally exhibited a preventive effect in models of age-related diseases. In an Alzheimer's disease model, butein treatment significantly delayed the paralysis caused by accumulation of amyloid-beta in muscle, which requires SKN-1, not DAF-16. In a high-glucose-diet model of diabetes mellitus, butein markedly improved survival, requiring both SKN-1 and DAF-16. In a Parkinson's disease model, dopaminergic neurodegeneration was completely inhibited by butein supplementation and the accumulation of α-synuclein was significantly reduced. These findings suggest the use of butein as a novel nutraceutical compound for aging and age-related diseases.
Collapse
Affiliation(s)
- Seona Kim
- Department of Medical Sciences, General Graduate School, Soonchunhyang University, 22 Soonchunhyang-ro, Asan 31538, Republic of Korea
| | - Hyemin Yoon
- Department of Medical Biotechnology, Soonchunhyang University, 22 Soonchunhyang-ro, Asan 31538, Republic of Korea
| | - Sang-Kyu Park
- Department of Medical Sciences, General Graduate School, Soonchunhyang University, 22 Soonchunhyang-ro, Asan 31538, Republic of Korea
- Department of Medical Biotechnology, Soonchunhyang University, 22 Soonchunhyang-ro, Asan 31538, Republic of Korea
| |
Collapse
|
6
|
Chen M, Zhong W, Xu W. Alcohol and the mechanisms of liver disease. J Gastroenterol Hepatol 2023; 38:1233-1240. [PMID: 37423758 DOI: 10.1111/jgh.16282] [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: 05/23/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023]
Abstract
Alcoholic liver disease (ALD), which is a leading cause of morbidity and mortality worldwide, covers a large spectrum of liver injuries ranging from simple steatosis to steatohepatitis, advanced fibrosis, cirrhosis, and hepatocellular carcinoma. The pathogenesis of ALD includes genetic and epigenetic alterations, oxidative stress, acetaldehyde-mediated toxicity and cytokine and chemokine-induced inflammation, metabolic reprogramming, immune damage, and dysbiosis of the gut microbiota. This review discusses the progress in the pathogenesis and molecular mechanism of ALD, which could provide evidence for further research on the potential therapeutic strategies targeting these pathways.
Collapse
Affiliation(s)
- Mo Chen
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wanglei Zhong
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Weiqi Xu
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Rehman M, Chaudhary R, Rajput S, Agarwal V, Kaushik AS, Srivastava S, Srivastava S, Singh R, Aziz I, Singh S, Mishra V. Butein Ameliorates Chronic Stress Induced Atherosclerosis via Targeting Anti-inflammatory, Anti-fibrotic and BDNF Pathways. Physiol Behav 2023; 267:114207. [PMID: 37100219 DOI: 10.1016/j.physbeh.2023.114207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/31/2023] [Accepted: 04/23/2023] [Indexed: 04/28/2023]
Abstract
Chronic stress is a major risk factor for various diseases, including cardiovascular diseases (CVDs). Chronic stress enhances the release of pro-inflammatory cytokines like IL-1β, IL-6, and TNF-α, making individuals susceptible to atherosclerosis which is dominant cause for CVDs. In present study, we validated a mouse model of chronic unpredictable stress (CUS), and assessed the characteristic features of atherosclerosis in thoracic aortas of CUS mice. The CUS procedure consisted of exposing groups of mice to random stressors daily for 10-weeks. The stress response was verified by presence of depressive-like behaviors and increased serum corticosterone in mice which was determined by battery of behavioural tests (SPT, EPMT, NSFT) and ELISA, respectively. Atherosclerosis parameters in CUS mice were evaluated by lipid indices estimation followed by histological assessment of plaque deposition and fibrosis in thoracic aorta. Further, we assessed the efficacy of a polyphenol, i.e. Butein in conferring protection against chronic stress-induced atherosclerosis and the possible mechanism of action. Butein (20mg/kg x 28 days, alternatively, i.p.) was administered to CUS mice after 6-weeks of CUS exposure till the end of the protocol. Butein treatment decreased peripheral IL-1β and enhanced peripheral as well as central BDNF levels. Histological assessment revealed decreased macrophage expression and reduced fibrosis in thoracic aorta of Butein treated mice. Further, treatment with Butein lowered lipid indices in CUS mice. Our findings thus, suggest that 10-weeks of CUS induce characteristic features of atherosclerosis in mice and Butein can offer protection in CUS-induced atherosclerosis through multiple mechanisms including anti-inflammatory, antifibrotic and anti-adipogenic actions.
Collapse
Affiliation(s)
- Mujeeba Rehman
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Sonu Rajput
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Vipul Agarwal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Arjun Singh Kaushik
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Siddhi Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Sukriti Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Rohit Singh
- Centre for Animal Disease Research and Diagnosis, ICAR-Indian Veterinary Research Institute, Izzatnagar, Bareilly 243122, Uttar Pradesh, India
| | - Irfan Aziz
- Integral University, Kursi road, Lucknow, Uttar Pradesh 226026, India
| | - Sanjay Singh
- Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025, India.
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India.
| |
Collapse
|
8
|
Wang F, Zhan Y, Li M, Wang L, Zheng A, Liu C, Wang H, Wang T. Cell-Permeable PROTAC Degraders against KEAP1 Efficiently Suppress Hepatic Stellate Cell Activation through the Antioxidant and Anti-Inflammatory Pathway. ACS Pharmacol Transl Sci 2022; 6:76-87. [PMID: 36654751 PMCID: PMC9841780 DOI: 10.1021/acsptsci.2c00165] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Indexed: 12/12/2022]
Abstract
Accumulating evidence indicates that oxidative stress and inflammation are involved in the physiopathology of liver fibrogenesis. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a key transcription factor, which regulates the expression of redox regulators to establish cellular redox homeostasis. The Nrf2 modulator can serve as a primary cellular defense against the cytotoxic effects of oxidative stress. We designed a chimeric Keap1-Keap1 peptide (KKP1) based on the proteolysis-targeting chimera technology. The KKP1 peptide not only can efficiently penetrate into the rat hepatic stellate cell line (HSC-T6) cells but also can induce Keap1 protein degradation by the ubiquitination-proteasome degradation pathway, which releases Nrf2 and promotes the transcriptional activity of the Nrf2/antioxidant response element pathway. It then activates the protein expression of the downstream antioxidant factors, the glutamate-cysteine ligase catalytic subunit and heme oxygenase-1 (HO-1). Finally, Keap1 protein degradation inhibits the nuclear factor-kappaB inflammatory signal pathway, the downstream inflammatory factor tumor necrosis factor alpha, and the interleukin-1beta protein expression and further inhibits the expression of the fibrosis biomarker gene. The current research suggests that our designed KKP1 may provide a new avenue for the future treatment of liver fibrosis.
Collapse
Affiliation(s)
- Fengqin Wang
- Hubei
Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Ying Zhan
- Hubei
Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Manman Li
- Hubei
Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Lidan Wang
- Hubei
Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China,Department
of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China
| | - Austin Zheng
- Institute
of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21215, United States
| | - Changbai Liu
- Hubei
Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Hu Wang
- Hubei
Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China,Institute
of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21215, United States,
| | - Tao Wang
- The
First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443003, China,
| |
Collapse
|
9
|
Mahdinloo S, Hemmati S, Valizadeh H, Mahmoudian M, Mahmoudi J, Roshangar L, Sarfraz M, Zakeri-Milani P. Synthesis and preparation of vitamin A coupled butein-loaded solid lipid nanoparticles for liver fibrosis therapy in rats. Int J Pharm 2022; 625:122063. [PMID: 35964827 DOI: 10.1016/j.ijpharm.2022.122063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/15/2022]
Abstract
The development of a therapeutic system for hepatic fibrosis has become a research hotspot to date. Butein, a simple chalcone derivative, displays anti-fibrotic effects through different pathways. However, impurities, low solubility, and low concentration in the target tissue hinder therapy with herbal ingredients. Hepatic stellate cells (HSCs), the vitamin A (VA) storage cells, as the main contributors to liver fibrogenesis, are not readily accessible to drugs owing to their anatomical location. Targeted delivery of therapeutics to the activated HSCs is therefore critical for successful treatment. For these reasons, the current study aimed at increasing butein delivery to the liver. Hence, high purity butein was synthesized in three steps. A novel VA-Myrj52 ester conjugate was also synthesized using all-trans retinoic acid and a hydrophilic emulsifier (Myrj52) as a targeting agent. Next, butein was encapsulated inside the novel VA-modified solid lipid nanoparticles (VA-SLNs) and studied in vitro and in vivo. According to our evaluations, negatively charged SLNs with a mean diameter of 150 nm and entrapment efficacy of 75 % were successful in liver fibrosis amelioration. Intraperitoneal (i.p.) injection of VA-SLNs in fibrotic rats, for four weeks long, reduced serum AST and ALT by 58% (P, 0.001) and 72% (P, 0.05), respectively, concerning the CCl4 group. Additionally, histologic damage score decline and normalization of tissue oxidative stress markers collectively confirmed the efficacy of formulations in hepatic fibrosis and kidney damage amelioration.
Collapse
Affiliation(s)
- Somayeh Mahdinloo
- Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz 5166616471, Iran
| | - Salar Hemmati
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran
| | - Hadi Valizadeh
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran.
| | - Mohammad Mahmoudian
- Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz 5166616471, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical sciences, Tabriz 5166614756, Iran
| | - Leyla Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran
| | - Muhammad Sarfraz
- College of Pharmacy, Al Ain University, Al Ain 64141, United Arab Emirates.
| | - Parvin Zakeri-Milani
- Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran.
| |
Collapse
|
10
|
Ding Q, Wang H, Wang Y, Lu Y. A thioredoxin reductase 1 inhibitor pyrano [3,2-a] phenazine inhibits A549 cells proliferation and migration through the induction of reactive oxygen species production. Mol Biol Rep 2022; 49:8835-8845. [PMID: 35780225 DOI: 10.1007/s11033-022-07733-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/20/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Thioredoxin reductase 1 (TrxR1) inhibitor, pyrano [3,2-a] phenazine, named CPUL-1, was synthesized with potential anticancer activity. The aim of the present work was to explore the potential anti-proliferative and anti-metastatic ability of CPUL-1 against A549 cancer cell lines in vitro. METHODS AND RESULTS First, Cell Counting Kit-8 (CCK8) assay was used to assess cell proliferation. The A549 cell migration was evaluated by wound healing assay and transwell assay. Second, the epithelial-mesenchymal transition (EMT)-related proteins in A549 cells treated with CPUL-1 were analyzed by western blot methods. Then, TrxR1 enzyme activity assay and reactive oxygen species (ROS) assay were conducted to evaluate the effect of CPUL-1 on TrxR1 inhibition and ROS levels. Finally, western blotting was used to explore the mechanism of CPUL-1. The study results revealed that the ability of cell proliferation and migration was decreased under CPUL-1 treatment. CPUL-1 could distinctly restrain the migration and invasion of A549 cells through inhibiting EMT process. The results of TrxR1 enzyme activity assay, ROS assay and western blotting showed that CPUL-1 influenced EMT via inducing ROS-mediated ERK/JNK signaling by inhibiting TrxR1 enzyme activity. CONCLUSIONS Together, proliferation suppression and anti-metastasis activity of CPUL-1 in A549 cells were demonstrated by all the evidence. Our findings highlight the great potential of phenazine compound CPUL-1 to suppress A549 cells proliferation and metastasis.
Collapse
Affiliation(s)
- Qifan Ding
- Department of Marine Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hengyu Wang
- Department of Marine Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ying Wang
- Department of Marine Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Yuanyuan Lu
- Department of Marine Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
11
|
Butein Inhibits Oxidative Stress Injury in Rats with Chronic Heart Failure via ERK/Nrf2 Signaling. Cardiovasc Ther 2022; 2022:8684014. [PMID: 35069800 PMCID: PMC8752302 DOI: 10.1155/2022/8684014] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 12/09/2021] [Indexed: 12/25/2022] Open
Abstract
Background Chronic heart failure (CHF) is a serious heart disease resulting from cardiac dysfunction. Oxidative stress is an important factor in aging and disease. Butein, however, has antioxidant properties. To determine the effect of butein on oxidative stress injury in rats, a CHF rat model was established. Methods The CHF rat model was induced by abdominal aortic coarctation (AAC). Rats in CHF+butein and sham+butein group were given 100 mg/kg butein via gavage every day to detect the effect of butein on oxidative stress injury and myocardial dysfunction. The cardiac structural and functional parameters, including the left ventricular end-systolic dimension (LVESD), the left ventricular end-diastolic dimension (LVEDD), the left ventricular ejection fraction (LVEF), and the left ventricular fractional shortening (LVFS), were measured. Oxidative stress was measured through the production of reactive oxygen species (ROS), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), catalase (CAT), and malondialdehyde (MDA). Cardiac injury markers like creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH), and aspartate aminotransferase (AST) were evaluated. Hematoxylin and eosin (H&E) staining was used to observe the myocardial cell morphology. The effect of butein on the extracellular signal-regulated kinase (ERK)/nuclear factor-E2 p45-related factor (Nrf2) signaling was confirmed by Western blot analysis. Results Butein had a significant effect on CHF in animal models. In detail, butein inhibited oxidative stress, relieved cardiac injury, and alleviated myocardial dysfunction. Importantly, butein activated the ERK1/2 pathway, which contributed to Nrf2 activation and subsequent heme oxygenase-1 (HO-1) and glutathione cysteine ligase regulatory subunit (GCLC) induction. Conclusions In this study, butein inhibits oxidative stress injury in CHF rat model via ERK/Nrf2 signaling pathway.
Collapse
|
12
|
Hsu YK, Chen HY, Wu CC, Huang YC, Hsieh CP, Su PF, Huang YF. Butein induces cellular senescence through reactive oxygen species-mediated p53 activation in osteosarcoma U-2 OS cells. ENVIRONMENTAL TOXICOLOGY 2021; 36:773-781. [PMID: 33325610 DOI: 10.1002/tox.23079] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 12/06/2020] [Indexed: 06/12/2023]
Abstract
Butein is a flavonoid isolated from various medicinal plants. It is known to have different biological activities including anti-inflammation, anti-adipogenesis, and anti-angiogenesis. In the study, we demonstrated the anti-proliferative effect of butein in human osteosarcoma U-2 OS cells. Our data showed that butein significantly suppressed the viability and colony formation ability of U-2 OS cells. Further experiments revealed butein exposure resulted in a cell cycle arrest at S and G2/M phase in U-2 OS cells. Importantly, we found that butein activated the tumor suppressor p53, and trigged a p53-dependent senescence in U-2 OS cells. Knockdown of p53 suppressed the senescence and rescued the viability in butein-treated U-2 OS cells. Furthermore, we observed that butein exposure significantly enhanced reactive oxygen species (ROS) levels in U-2 OS cells. Co-administration of the ROS inhibitor NAC largely abolished the up-regulated p53 protein level, and rescued the suppressed viability and colony formation ability in butein-exposed U-2 OS cells. Taken together, our data proposed the increased ROS by butein exposure activated p53, and the activated p53 was involved in the anti-proliferative effect of butein via inducing senescence in U-2 OS cells. This report suggests that butein is a promising candidate for cancer therapy against osteosarcoma.
Collapse
Affiliation(s)
- Yung-Ken Hsu
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Hsuan-Ying Chen
- Orthopedics and Sports Medicine Laboratory, Changhua Christian Hospital, Changhua, Taiwan
| | - Chia-Chieh Wu
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua, Taiwan
- School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ying-Chih Huang
- Department of Research, Changhua Christian Hospital, Changhua, Taiwan
| | - Cheng-Pu Hsieh
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua, Taiwan
- Orthopedics and Sports Medicine Laboratory, Changhua Christian Hospital, Changhua, Taiwan
| | - Po-Feng Su
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Yi-Fu Huang
- Orthopedics and Sports Medicine Laboratory, Changhua Christian Hospital, Changhua, Taiwan
| |
Collapse
|
13
|
Ramalingam V, Rajaram R. A paradoxical role of reactive oxygen species in cancer signaling pathway: Physiology and pathology. Process Biochem 2021. [DOI: 10.1016/j.procbio.2020.09.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
14
|
Liu YM, Cong S, Cheng Z, Hu YX, Lei Y, Zhu LL, Zhao XK, Mu M, Zhang BF, Fan LD, Yu L, Cheng ML. Platycodin D alleviates liver fibrosis and activation of hepatic stellate cells by regulating JNK/c-JUN signal pathway. Eur J Pharmacol 2020; 876:172946. [PMID: 31996320 DOI: 10.1016/j.ejphar.2020.172946] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 01/13/2020] [Accepted: 01/24/2020] [Indexed: 12/12/2022]
Abstract
Liver fibrosis is involved in the progression of most chronic liver diseases. Even though we have made a huge progress in order to understand the pathogenesis of liver fibrosis, however, there is still a lack of productive treatments. Being a traditional Chinese medicine, Platycodin D (PD), an oleanane kind of triterpenoid saponin has been put to extensive use for treating different kinds of illnesses that include not just anti-nociceptive, but also antiviral, anti-inflammatory, and anti-cancer for thousands of years. Nonetheless, there has been no clarification made for its effects on the progression of liver fibrosis. In this manner, we carried out in vitro studies for the purpose of investigating the anti-fibrosis impact of PD. Activation of hepatic stellate cells was evaluated by means of the detection of the proliferation of HSCs and the expression of specific proteins. We discovered the fact that PD had the potential of activating HSCs. Thereafter, we detected the apoptosis and autophagy of the HSCs; as the results suggested, PD induced apoptosis and autophagy of the HSCs. It augmented the expression level of apoptotic proteins that included Bax, Cytochrome C (cyto-c), cleaved caspase3 and cleaved caspase9, in addition to the autophagy relevant proteins, for instance, LC3II, beclin1, Atg5 and Atg9. Further research was carried out for the investigation of the underlying molecular mechanism, and discovered that PD promoted the phosphorylation of JNK and c-Jun. Treating the JNK inhibitor P600125 inhibited the effect of PD, confirming the impact of PD on the regulation of JNK/c-Jun pathway. Thus, we speculated that PD alleviates liver fibrosis and activation of hepatic stellate via promoting phosphorylation of JNK and c-Jun and further altering the autophagy along with apoptosis of HSCs.
Collapse
Affiliation(s)
- Yong-Mei Liu
- Department of Infectious Disease, The Affiliated Hospital, Guizhou Medical University, Beijing Road 9, Guiyang, 550004, Guizhou, China
| | - Shuo Cong
- Department of Blood Transfusion, The Affiliated Tumor Hospital, Guizhou Medical University, Beijing west Road1, Guiyang, 550000, Guizhou, China
| | - Zhuo Cheng
- Peking University Health Science Center School of Foundational Education, Beijing, 100191, Beijing, China
| | - Ya-Xin Hu
- Prenatal Diagnosis Center,The Affiliated Hospital, Guizhou Medical University, Beijing Road 9, Guiyang, 550004, Guizhou, China
| | - Yu Lei
- Department of Infectious Disease, The Affiliated Hospital, Guizhou Medical University, Beijing Road 9, Guiyang, 550004, Guizhou, China
| | - Li-Li Zhu
- Department of Infectious Disease, The Affiliated Hospital, Guizhou Medical University, Beijing Road 9, Guiyang, 550004, Guizhou, China
| | - Xue-Ke Zhao
- Department of Infectious Disease, The Affiliated Hospital, Guizhou Medical University, Beijing Road 9, Guiyang, 550004, Guizhou, China
| | - Mao Mu
- Department of Infectious Disease, The Affiliated Hospital, Guizhou Medical University, Beijing Road 9, Guiyang, 550004, Guizhou, China
| | - Bao-Fang Zhang
- Department of Infectious Disease, The Affiliated Hospital, Guizhou Medical University, Beijing Road 9, Guiyang, 550004, Guizhou, China
| | - Lin-da Fan
- Department of Infectious Disease, The Affiliated Hospital, Guizhou Medical University, Beijing Road 9, Guiyang, 550004, Guizhou, China
| | - Lei Yu
- Prenatal Diagnosis Center,The Affiliated Hospital, Guizhou Medical University, Beijing Road 9, Guiyang, 550004, Guizhou, China.
| | - Ming-Liang Cheng
- Department of Infectious Disease, The Affiliated Hospital, Guizhou Medical University, Beijing Road 9, Guiyang, 550004, Guizhou, China.
| |
Collapse
|
15
|
Salehi B, Butnariu M, Corneanu M, Sarac I, Vlaisavljevic S, Kitic D, Rahavian A, Abedi A, Karkan MF, Bhatt ID, Jantwal A, Sharifi-Rad J, Rodrigues CF, Martorell M, Martins N. Chronic pelvic pain syndrome: Highlighting medicinal plants toward biomolecules discovery for upcoming drugs formulation. Phytother Res 2019; 34:769-787. [PMID: 31799719 DOI: 10.1002/ptr.6576] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/18/2019] [Accepted: 11/14/2019] [Indexed: 12/27/2022]
Abstract
Chronic pelvic pain syndrome (CPPS) can be triggered by a various types of gynecological, gastrointestinal, urological, and musculoskeletal disorders. Recently, the role of the central nervous system has proven to be an integral part on the development of any chronic pain syndrome, including CPPS. However, owing to the complex and heterogeneous etiology and pathophysiology of CPPS, the establishment of effective therapeutic interventions remains challenging for both physicians and patients. Nonetheless, recent studies have pointed that medicinal plants and their secondary metabolites can be effectively used in CPPS therapy, besides contributing to restore the patients' quality of life and potentiate the conventional CPPS management. In this sense, this review aims to provide a careful overview on the biomedical data for the use of medicinal plants use and their secondary metabolites on CPPS management.
Collapse
Affiliation(s)
- Bahare Salehi
- Student Research Committee, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | - Monica Butnariu
- Banat's University of Agricultural Sciences and Veterinary Medicine "King Michael I of Romania" from Timisoara, University of Timişoara, Timişoara, Romania
| | - Mihaela Corneanu
- Banat's University of Agricultural Sciences and Veterinary Medicine "King Michael I of Romania" from Timisoara, University of Timişoara, Timişoara, Romania
| | - Ioan Sarac
- Banat's University of Agricultural Sciences and Veterinary Medicine "King Michael I of Romania" from Timisoara, University of Timişoara, Timişoara, Romania
| | - Sanja Vlaisavljevic
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - Dusanka Kitic
- Faculty of Medicine, Department of Pharmacy, University of Niš, Bul. Zorana Djindjica 81, Serbia
| | - Amirhossein Rahavian
- Department of Urology, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirreza Abedi
- Department of Urology, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morteza F Karkan
- Department of Urology, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Indra D Bhatt
- G.B. Pant National Institute of Himalayan Environment and Sustainable Development, Kosi Katarmal, Almora, India
| | - Arvind Jantwal
- Department of Pharmaceutical Sciences, Bhimtal Campus, Kumaun University, Nainital, India
| | - Javad Sharifi-Rad
- Zabol Medicinal Plants Research Center, Zabol University of Medical Sciences, Zabol, Iran
| | - Célia F Rodrigues
- LEPABE, Department of Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, University of Concepcion, Concepcion, Chile.,Universidad de Concepción, Unidad de Desarrollo Tecnológico, Concepcion, Chile
| | - Natália Martins
- Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, Porto, Portugal.,Institute for Research and Innovation in Health (i3S), University of Porto, Portugal
| |
Collapse
|
16
|
Pandey G, Marimuthu M, Kanagavalli P, Ravichandiran V, Balamurugan K, Veerapandian M. Chitosanylated MoO3–Ruthenium(II) Nanocomposite as Biocompatible Probe for Bioimaging and Herbaceutical Detection. ACS Biomater Sci Eng 2019; 5:3606-3617. [DOI: 10.1021/acsbiomaterials.9b00575] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Gaurav Pandey
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700 054, India
| | - Mohana Marimuthu
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu 630 003, India
| | | | - Velayutham Ravichandiran
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700 054, India
| | - Krishnaswamy Balamurugan
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu 630 003, India
| | | |
Collapse
|
17
|
Yeh Y, Liang C, Chen M, Tsai F, Lin Y, Lee M, Wu J, Kuo C. Apoptotic effects of hsian-tsao ( Mesona procumbens Hemsley) on hepatic stellate cells mediated by reactive oxygen species and ERK, JNK, and caspase-3 pathways. Food Sci Nutr 2019; 7:1891-1898. [PMID: 31139404 PMCID: PMC6526671 DOI: 10.1002/fsn3.1046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/31/2019] [Accepted: 04/07/2019] [Indexed: 12/20/2022] Open
Abstract
The activation of hepatic stellate cells (HSCs) is an important step in the progress of liver fibrosis. Fibrosis can be impeded by HSC reversion to a quiescent state or HSC clearance through apoptosis. To investigate the apoptotic effects of hsian-tsao (Mesona procumbens Hemsl) on human HSCs, the expression levels of cleaved caspase-3, p38, and c-Jun N-terminal kinase (JNK) were assessed using Western blotting, and the caspase-3 activity was measured using caspase-3/CPP32 colorimetric assay kit. Hsian-tsao extract (HTE) increased the activity of caspase-3 and the level of activated caspase-3, indicating the activation of apoptosis. The intracellular reactive oxygen species (ROS) level increased in a dose-dependent manner. This increase was prevented by an antioxidant, suggesting that HTE induces ROS accumulation. In addition, we found that HTE induced the phosphorylation of the mitogen-activated protein kinases JNK and p38. These collective data indicate that HTE induces apoptosis via ROS production through the p38, JNK, and caspase-3-dependent pathways. HTE may decrease HSC activation in liver fibrosis and may have a therapeutic potential.
Collapse
Affiliation(s)
- Yung‐Hsiang Yeh
- Division of GastroenterologyChang Bing Show Chwan Memorial HospitalChanghuaTaiwan
| | - Chun‐Ya Liang
- Department of Medical Research and DevelopmentChang Bing Show Chwan Memorial HospitalChanghuaTaiwan
| | - Mao‐Liang Chen
- Department of Research, Taipei Tzu Chi HospitalBuddhist Tzu Chi Medical FoundationNew Taipei CityTaiwan
| | - Fu‐Ming Tsai
- Department of Research, Taipei Tzu Chi HospitalBuddhist Tzu Chi Medical FoundationNew Taipei CityTaiwan
| | - Yi‐Ying Lin
- Department of Research, Taipei Tzu Chi HospitalBuddhist Tzu Chi Medical FoundationNew Taipei CityTaiwan
| | - Ming‐Cheng Lee
- Department of Research, Taipei Tzu Chi HospitalBuddhist Tzu Chi Medical FoundationNew Taipei CityTaiwan
| | - Jiunn‐Sheng Wu
- Division of Infectious DiseasesTaipei Tzu Chi HospitalBuddhist Tzu Chi Medical FoundationNew Taipei CityTaiwan
| | - Chan‐Yen Kuo
- Department of Research, Taipei Tzu Chi HospitalBuddhist Tzu Chi Medical FoundationNew Taipei CityTaiwan
| |
Collapse
|
18
|
Mafuru M, Wu S, He S, Lu X, Huang J, Jiang H. The Influence of Proinflammatory Cytokines on Voriconazole Trough Concentration in Patients With Different Forms of Hematologic Disorders. J Clin Pharmacol 2019; 59:1340-1350. [PMID: 30997931 DOI: 10.1002/jcph.1422] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/28/2019] [Indexed: 12/13/2022]
Abstract
Even though multiple factors are involved in the high fluctuation of voriconazole (VCZ) plasma concentration, little is known regarding the influence of proinflammatory cytokines on VCZ concentration. The aim of this study was to investigate the influence of proinflammatory cytokines, namely, interleukin (IL)-1β, IL-6, IL-18, interferon-γ, tumor necrosis factor-α, and transforming growth factor (TGF)-β1 on VCZ trough concentration (VCZ-Cmin ) in Chinese patients with different forms of hematologic disorders. A total of 250 plasma samples from 113 patients were analyzed for VCZ-Cmin and proinflammatory cytokines using a validated liquid chromatography-tandem mass spectrometry and enzyme-linked immunosorbent assay methods, respectively. Patient demographics and clinical characteristics were obtained from hospital records. VCZ-Cmin was significantly correlated with IL-18 in acute myeloid leukemia (r = 0.456; P ˂ .0001), acute lymphoblastic leukemia (r = 0.317; P = .019), and chronic myeloid leukemia (r = 0.737; P = .004) while VCZ-Cmin and TGF-β1 were correlated (r = 0.436; P ˂ .001) in acute myeloid leukemia patients only. VCZ-Cmin at different concentration range showed significant inhibitory effect of IL-6. A backward multiple linear regression model revealed patient age (coefficient [β] = 0.025; P = .04), gamma-glutamyl transferase (β = 0.003; P = .023), IL-6 (β = -0.001; P = .024), proton pump inhibitor coadministration (β = 1.518; P = .002), and cytochrome P450 (CYP) 2C19 polymorphism as predictors of VCZ-Cmin ; however, these factors explained only 29% of VCZ-Cmin variation. In conclusion, IL-18 and TGF-β1 have correlation with VCZ-Cmin in Chinese patients with leukemia. Apparently, VCZ may have an inhibitory effect on IL-6 levels. Furthermore, patient age, gamma-glutamyl transferase, IL-6, PPI coadministration, and cytochrome P450 2C19 polymormorphism partially predicted the VCZ-Cmin . Therapeutic drug monitoring of VCZ in Chinese patients is highly encouraged.
Collapse
Affiliation(s)
- Magesa Mafuru
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sanlan Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sijie He
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Lu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiangeng Huang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongliang Jiang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Suzuki M, Kon K, Ikejima K, Arai K, Uchiyama A, Aoyama T, Yamashina S, Ueno T, Watanabe S. The Chemical Chaperone 4-Phenylbutyric Acid Prevents Alcohol-Induced Liver Injury in Obese KK-A y Mice. Alcohol Clin Exp Res 2019; 43:617-627. [PMID: 30748014 DOI: 10.1111/acer.13982] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 02/08/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Co-occurrence of metabolic syndrome and chronic alcohol consumption is increasing worldwide. The present study investigated the effect of the chemical chaperone 4-phenylbutyric acid (PBA)-which has been shown to alleviate dietary steatohepatitis caused by endoplasmic reticulum (ER) stress-on chronic-plus-binge ethanol (EtOH)-induced liver injury in a mouse model of obesity. METHODS Male KK-Ay mice (8 weeks old) were fed a Lieber-DeCarli diet (5% EtOH) for 10 days. Some mice were given PBA intraperitoneally (120 mg/kg body weight, daily) during the experimental period. On day 11, mice were gavaged with a single dose of EtOH (4 g/kg body weight). Control mice were given a dextrin gavage after being pair-fed a control diet. All mice were then serially euthanized before or at 9 hours after gavage. RESULTS Chronic-plus-binge EtOH intake induced massive hepatic steatosis along with hepatocyte apoptosis and inflammation, which was reversed by PBA treatment. Administration of PBA also suppressed chronic-plus-binge EtOH-induced up-regulation of ER stress-related genes including binding immunoglobulin protein (Bip), unspliced and spliced forms of X-box-binding protein-1 (uXBP1 and sXBP1, respectively), inositol trisphosphate receptor (IP3R), and C/EBP homologous protein (CHOP). Further, it blocked chronic-plus-binge EtOH-induced expression of the oxidative stress marker heme oxygenase-1 (HO-1) and 4-hydroxynonenal. Chronic EtOH alone (without binge) increased Bip and uXBP1, but it did not affect those of sXBP1, IP3R, CHOP, or HO-1. PBA reversed the prebinge expression of these genes to control levels, but it did not affect chronic EtOH-induced hepatic activity of cytochrome P450 2E1. CONCLUSIONS Binge EtOH intake after chronic consumption induces massive ER stress-related oxidative stress and liver injury in a mouse model of obesity through dysregulation of the unfolded protein response. PBA ameliorated chronic-plus-binge EtOH-induced liver injury by reducing ER and oxidative stress after an EtOH binge.
Collapse
Affiliation(s)
- Maiko Suzuki
- Department of Gastroenterology , Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuyoshi Kon
- Department of Gastroenterology , Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kenichi Ikejima
- Department of Gastroenterology , Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kumiko Arai
- Department of Gastroenterology , Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akira Uchiyama
- Department of Gastroenterology , Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomonori Aoyama
- Department of Gastroenterology , Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shunhei Yamashina
- Department of Gastroenterology , Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takashi Ueno
- Laboratory of Proteomics and Biomolecular Science, Laboratory of Proteomics and Medical Science, Research Support Center, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Sumio Watanabe
- Department of Gastroenterology , Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
20
|
SIRT1 activation by butein attenuates sepsis-induced brain injury in mice subjected to cecal ligation and puncture via alleviating inflammatory and oxidative stress. Toxicol Appl Pharmacol 2018; 363:34-46. [PMID: 30336174 DOI: 10.1016/j.taap.2018.10.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 10/10/2018] [Accepted: 10/12/2018] [Indexed: 12/13/2022]
Abstract
Sepsis-induced brain injury is frequently encountered in critically ill patients with severe systemic infection. Butein (3,4,2',4'-tetrahydroxychalcone) has been demonstrated as the neuro-protective agent via reducing inflammation and oxidative stress on neurons. Moreover, activation of silent information regulator 1 (SIRT1) inhibits apoptosis, oxidation and inflammation thus alleviating sepsis-induced multiorgan injuries. In present study, we show that butein administrated intraperitoneally (10 mg/kg) saved mice from sepsis-induced lethality by increasing 7-day survival rate after cecal ligation and puncture (CLP) surgery. Additionally, butein treatment enhanced SIRT1 signaling thus decreasing the Ac-NF-κB, Ac-FOXO1 and Ac-p53 levels, thus attenuating the brain injury of mice after CLP surgery by decreasing cerebral edema, maintaining the blood-brain barrier integrity, inhibiting neuronal apoptosis, and decreasing pro-inflammatory cytokines production (IL-6, TNF-α and IL-1β) and oxidative stress (downregulation of MDA, and upregulation of SOD and CAT) in both serum and cerebral cortex tissues. Moreover, butein treatment attenuated LPS induced neurological function loss. However, all above mentioned neuro-protective actions of butein were partially inhibited by EX527 co-treatment, one standard SIRT1 inhibitor. Collectively, butein attenuates sepsis-induced brain injury through alleviation of cerebral inflammation, oxidative stress and apoptosis by SIRT1 signaling activation.
Collapse
|
21
|
Wang Y, Zhao L, Jiao FZ, Zhang WB, Chen Q, Gong ZJ. Histone deacetylase inhibitor suberoylanilide hydroxamic acid alleviates liver fibrosis by suppressing the transforming growth factor-β1 signal pathway. Hepatobiliary Pancreat Dis Int 2018; 17:423-429. [PMID: 30249543 DOI: 10.1016/j.hbpd.2018.09.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Histone deacetylases (HDACs) inhibitors are new anti-fibrotic drugs that inhibit the activity of hepatic stellate cells. The present study focused on the anti-fibrotic function of HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) by suppressing transforming growth factor-β1 (TGF-β1) signaling. METHODS Male Sprague-Dawley rats were used to induce liver fibrosis with carbon tetrachloride (CCl4) and LX2 cell (human hepatic stellate cell line) was stimulated by TGF-β1. Both animals and cells were treated with SAHA. The Smad7 and connective tissue growth factor (CTGF) mRNA levels were detected by real-time polymerase chain reaction (PCR). Western blotting was used to examine the protein levels of CTGF, Histone H3 (H3), Smad7, Smad2/3, Acetyl-Histone H3 (AH3), HDAC2, α-smooth muscle actin (α-SMA), HDAC6, p-Smad2/3 and HDAC8. In addition, the TGF-β1 and liver enzyme levels from rat serum were detected. Histopathological changes were examined by hematoxylin and eosin (HE), Sirius red and Masson trichrome staining. The α-SMA expression was detected by immumohistochemical staining. RESULTS Compared with control group, the TGF-β1 and liver enzyme levels from rat serum, together with the mRNA levels of CTGF and protein levels of CTGF, HDAC2, α-SMA, HDAC6, p-Smad2/3 and HDAC8 were elevated in fibrotic rats (P < 0.01). But the Smad7 mRNA and AH3 protein levels were notably suppressed in the fibrotic rats (P < 0.01). Pathological examination showed the typical changes of liver fibrosis in the fibrotic rats. After the treatment with SAHA, the levels of liver enzymes, TGF-β1, CTGF, HDAC2, α-SMA, HDAC6, p-Smad2/3 and HDAC8 were reduced (P < 0.01) and Smad7 and AH3 protein contents were elevated in liver fibrotic rats (P < 0.01). Moreover, immumohistochemistry showed that SAHA significantly suppressed the α-SMA protein content in fibrotic liver (P < 0.01). CONCLUSION The HDAC inhibitor SAHA alleviated liver fibrosis by suppressing the TGF-β1 signaling.
Collapse
Affiliation(s)
- Yao Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fang-Zhou Jiao
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wen-Bin Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qian Chen
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zuo-Jiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
22
|
Salama SM, Ibrahim IAA, Shahzad N, Al-Ghamdi S, Ayoub N, AlRashdi AS, Abdulla MA, Salehen N, Bilgen M. Hepatoprotectivity of Panduratin A against liver damage: In vivo demonstration with a rat model of cirrhosis induced by thioacetamide. APMIS 2018; 126:710-721. [PMID: 30058214 DOI: 10.1111/apm.12878] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 06/22/2018] [Indexed: 12/16/2022]
Abstract
This experiment evaluated Panduratin A (PA), a chalcone isolated from Boesenbergia rotunda rhizomes, for its hepatoprotectivity. Rats were subjected to liver damage induced by intra-peritoneal injection of thioacetamide (TAA). PA was tested first for its acute toxicity and then administered by oral gavage at doses 5, 10, and 50 mg/kg to rats. At the end of the 8th week, livers from all rats were excised and evaluated ex vivo. Measurements included alkaline phosphatase (AP), alanine transaminase (ALT), aspartate transaminase (AST) and gamma-glutamyl transferase (GGT), serum platelet-derived growth factor (PDGF) and transforming growth factor (TGF-β1), and hepatic metalloproteinase enzyme (MMP-2) and its inhibitor extracellular matrix protein (TIMP-1). Oxidative stress was measured by liver malondialdehyde (MDA) and nitrotyrosine levels, urinary 8-hydroxy 2- deoxyguanosine (8-OH-dG), and hepatic antioxidant enzyme activities. The immunohistochemistry of TGF-β1 was additionally performed. PA revealed safe dose of 250 mg/kg on experimental rats and positive effect on the liver. The results suggested reduced hepatic stellate cells (HSCs) activity as verified from the attenuation of serum PDGF and TGF-β1, hepatic MMP-2 and TIMP-1, and oxidative stress. The extensive data altogether conclude that PA treatment could protect the liver from the progression of cirrhosis through a possible mechanism inhibiting HSCs activity.
Collapse
Affiliation(s)
- Suzy M Salama
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Ibrahim Abdel Aziz Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Naiyer Shahzad
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Saeed Al-Ghamdi
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Nahla Ayoub
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ahmed Salim AlRashdi
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mahmood Ameen Abdulla
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Nur'Ain Salehen
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mehmet Bilgen
- Biophysics Department, Faculty of Medicine, Adnan Menderes University, Aydin, Turkey
| |
Collapse
|
23
|
Karimi-Sales E, Mohaddes G, Alipour MR. Chalcones as putative hepatoprotective agents: Preclinical evidence and molecular mechanisms. Pharmacol Res 2018; 129:177-187. [DOI: 10.1016/j.phrs.2017.11.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/16/2017] [Accepted: 11/17/2017] [Indexed: 02/08/2023]
|
24
|
The Bioactive Extract of Pinnigorgia sp. Induces Apoptosis of Hepatic Stellate Cells via ROS-ERK/JNK-Caspase-3 Signaling. Mar Drugs 2018; 16:md16010019. [PMID: 29315209 PMCID: PMC5793067 DOI: 10.3390/md16010019] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/30/2017] [Accepted: 01/06/2018] [Indexed: 12/12/2022] Open
Abstract
The activation of hepatic stellate cells (HSCs) is a significant phenomenon during the pathogenesis of liver disorders, including liver cirrhosis and fibrosis. Here, we identified that the extract from a gorgonian coral Pinnigorgia sp. (Pin) induced apoptosis of HSC-T6 cells. Pin inhibited the viability of HSC-T6 cells and increased their subG1 population, DNA fragmentation, caspase-3 activation, and reactive oxygen species (ROS) production in a concentration-dependent manner. The Pin-induced ROS generation and apoptotic effects were significantly reversed by a thiol antioxidant, N-acetylcysteine (NAC). Additionally, Pin induced ERK/JNK phosphorylation and pharmacological inhibition of ERK/JNK rescued the Pin-induced cell death. Pin-activated ERK/JNK were significantly reduced after the administration of NAC; however, the inhibition of ERK/JNK failed to change the Pin-induced ROS production. Similarly, pinnigorgiol A, a pure compound isolated from Pin, elicited ROS production and apoptosis in HSC-T6 cells. The pinnigorgiol A-induced apoptosis was retrained by NAC. Together, it appears that Pin leads to apoptosis in HSC-T6 cells through ROS-mediated ERK/JNK signaling and caspase-3 activation. Pinnigorgiol A serves as a bioactive compound of Pin and may exhibit therapeutic potential by clearance of HSCs.
Collapse
|
25
|
Islam MS, Akhtar MM, Segars JH, Castellucci M, Ciarmela P. Molecular targets of dietary phytochemicals for possible prevention and therapy of uterine fibroids: Focus on fibrosis. Crit Rev Food Sci Nutr 2018; 57:3583-3600. [PMID: 28609115 DOI: 10.1080/10408398.2016.1245649] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Uterine fibroids (myomas or leiomyomas) are common benign tumors of reproductive aged women. Fibroids are clinically apparent in 20-50% of women, and cause abnormal uterine bleeding, abdominal pain and discomfort, pregnancy complications and infertility. Unfortunately, limited numbers of medical treatment are available but no effective preventive strategies exist. Moreover, the benefits of medical treatments are tempered by lack of efficacy or serious adverse side effects. Fibrosis has recently been recognized as a key pathological event in leiomyoma development and growth. It is defined by the excessive deposition of extracellular matrix (ECM). ECM plays important role in making bulk structure of leiomyoma, and ECM-rich rigid structure is believed to be a cause of abnormal bleeding and pelvic pain/pressure. Dietary phytochemicals are known to regulate fibrotic process in different biological systems, and being considered as potential tool to manage human health. At present, very few dietary phytochemicals have been studied in uterine leiomyoma, and they are mostly known for their antiproliferative effects. Therefore, in this review, our aim was to introduce some dietary phytochemicals that could target fibrotic processes in leiomyoma. Thus, this review could serve as useful resource to develop antifibrotic drugs for possible prevention and treatment of uterine fibroids.
Collapse
Affiliation(s)
- Md Soriful Islam
- a Department of Experimental and Clinical Medicine , Faculty of Medicine, Università Politecnica delle Marche , Ancona , Italy.,b Biotechnology and Microbiology Laboratory, Department of Botany , University of Rajshahi , Rajshahi , Bangladesh
| | - Most Mauluda Akhtar
- a Department of Experimental and Clinical Medicine , Faculty of Medicine, Università Politecnica delle Marche , Ancona , Italy.,c Department of Clinical and Molecular Sciences , Faculty of Medicine, Università Politecnica delle Marche , Ancona , Italy
| | - James H Segars
- d Howard W. and Georgeanna Seegar Jones Division of Reproductive Sciences, Department of Gynecology and Obstetrics , Johns Hopkins School of Medicine , Baltimore , Maryland , USA
| | - Mario Castellucci
- a Department of Experimental and Clinical Medicine , Faculty of Medicine, Università Politecnica delle Marche , Ancona , Italy
| | - Pasquapina Ciarmela
- a Department of Experimental and Clinical Medicine , Faculty of Medicine, Università Politecnica delle Marche , Ancona , Italy.,e Department of Information Engineering , Università Politecnica delle Marche , Ancona , Italy
| |
Collapse
|
26
|
Xie ZY, Xiao ZH, Wang FF. Inhibition of autophagy reverses alcohol-induced hepatic stellate cells activation through activation of Nrf2-Keap1-ARE signaling pathway. Biochimie 2018; 147:55-62. [PMID: 29305174 DOI: 10.1016/j.biochi.2017.12.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 12/28/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Numerous documents have indicated a critical role of autophagy in alcoholic liver fibrosis (ALF), but few papers have reported its function in hepatic stellate cells (HSCs) activation. The current study aimed to investigate the regulation effect of autophagy in HSCs activation, in further to explore the underlying mechanism involved. METHODS HSC-T6 cells were treated with ethanol, 3-MA (autophagy inhibitor) or rapamycin (autophagy inducer), and cells were also transfected with si-Nrf2 or si-Keap1. Moreover, ALF animal model was established and Nrf-2(-/-), Keap1 (-/-) mice were purchased. The level of autophagy, the expression of α-SMA and CoL1A1, and Nrf2 antioxidant response were evaluated in stellate cells and livers. RESULTS Ethanol treatment in cultured cells increased autophagy, oxidative stress level and promoted HSCs activation. Inhibition of autophagy reversed alcohol-induced HSCs activation and suppressed HSCs oxidative stress. Nrf2-Keap1-ARE pathway was involved in HSCs activation and oxidative stress regulated by autophagy. In addition, through in vivo study, we found that inhibition of autophagy could alleviate alcoholic fatty liver injury in ALF model mice and Nrf2 signaling was involved in autophagy regulated HSCs activation. CONCLUSION These data implicated mechanisms underlying autophagy in regulating the fibrogenic response in HSCs activation.
Collapse
Affiliation(s)
- Zheng-Yuan Xie
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Zhi-Hua Xiao
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Fen-Fen Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| |
Collapse
|
27
|
Zhou J, Zhang J, Wang C, Qu S, Zhu Y, Yang Z, Wang L. Açaí ( Euterpe oleracea Mart.) attenuates alcohol-induced liver injury in rats by alleviating oxidative stress and inflammatory response. Exp Ther Med 2017; 15:166-172. [PMID: 29399060 PMCID: PMC5769297 DOI: 10.3892/etm.2017.5427] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 04/28/2017] [Indexed: 01/03/2023] Open
Abstract
The present study aimed to investigate the therapeutic effects of Euterpe oleracea Mart. (EO) on alcoholic liver diseases (ALD). A total of 30 Wistar rats were randomly divided into three groups (10 rats per group), including alcohol group (alcohol intake), EO group (alcohol + EO puree intake) and control group (distilled water intake). The activity of superoxide dismutase (SOD) and alkaline phosphatase (ALP), alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and the levels of cholesterol (CHO), triglyceride (TG), malondialdehyde (MDA) and glutathione (GSH) in the serum as well as the liver tissue levels of interleukin 8 (IL-8), tumor necrosis factor-α (TNF-α) and transforming growth factor-β (TGF-β) were measured. Histopathological changes in liver tissues were observed by hematoxylin and eosin staining. Reverse-transcription quantitative PCR analysis was performed for detecting the expression of nuclear factor (NF)-κB and CD68. The results indicated that EO intake significantly decreased ALT, AST, ALP, TG and CHO as well as the hepatic index in alcohol-treated rats. In addition, EO treatment relieved alcohol-induced oxidative stress by decreasing the levels of MDA and TG, and increasing the activity of SOD and GSH levels. In addition, the expression of TNF-α, TGF-β, IL-8, NF-κB and CD-68 in the liver were decreased by EO treatment. Furthermore, EO intake alleviated the histopathological liver damage, including severe steatosis and abundant infiltrated inflammatory cells. In conclusion, EO alleviated alcohol-induced liver injury in rats by alleviating oxidative stress and inflammatory response.
Collapse
Affiliation(s)
- Jianyu Zhou
- Department of Pharmaceutics of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, P.R. China.,Department of Traditional Chinese Clinical Pharmacology, School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing 100029, P.R. China.,Department of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Jianjun Zhang
- Department of Traditional Chinese Clinical Pharmacology, School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Chun Wang
- Department of Traditional Chinese Clinical Pharmacology, School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Shengsheng Qu
- Department of Traditional Chinese Clinical Pharmacology, School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Yingli Zhu
- Department of Traditional Chinese Clinical Pharmacology, School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Zhihui Yang
- Department of Psychiatry, University of Florida, Gainesville, FL 32608, USA
| | - Linyuan Wang
- Department of Pharmaceutics of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| |
Collapse
|
28
|
Betulinic acid, natural pentacyclic triterpenoid prevents arsenic-induced nephrotoxicity in male Wistar rats. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s00580-017-2548-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
29
|
Luo L, Xi C, Xu T, Zhang G, Qun E, Zhang W. Muscarinic receptor mediated signaling pathways in hepatocytes from CCL4 - induced liver fibrotic rat. Eur J Pharmacol 2017; 807:109-116. [DOI: 10.1016/j.ejphar.2017.03.047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 03/17/2017] [Accepted: 03/21/2017] [Indexed: 12/21/2022]
|
30
|
The antioxidative and hepatoprotective effects comparison of Chinese angelica polysaccharide(CAP)and selenizing CAP (sCAP) in CCl4 induced hepatic injury mice. Int J Biol Macromol 2017; 97:46-54. [DOI: 10.1016/j.ijbiomac.2017.01.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 12/07/2016] [Accepted: 01/02/2017] [Indexed: 12/17/2022]
|
31
|
Lu C, Zou Y, Liu Y, Niu Y. Rosmarinic acid counteracts activation of hepatic stellate cells via inhibiting the ROS-dependent MMP-2 activity: Involvement of Nrf2 antioxidant system. Toxicol Appl Pharmacol 2017; 318:69-78. [PMID: 28115189 DOI: 10.1016/j.taap.2017.01.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 01/08/2017] [Accepted: 01/16/2017] [Indexed: 01/18/2023]
Abstract
Recently, oxidative stress is involved in hepatofibrogenesis. Matrix metalloproteinase-2 (MMP-2) is required for activation of hepatic stellate cells (HSCs) in response to reactive oxygen species (ROS). This study was designed to explore the hypothesis that the inhibitory effect of rosmarinic acid (RA) on HSCs activation might mainly result from its antioxidant capability by increasing the synthesis of glutathione (GSH) involved in nuclear factor kappa B (NF-κB)-dependent inhibition of MMP-2 activity. Here, we demonstrate that RA reverses activated HSCs to quiescent cells. Concomitantly, RA inhibits MMP-2 activity. RNA interference-imposed knockdown of NF-κB abolished down-regulation of MMP-2 by RA. RA-mediated inactivation of NF-κB could be blocked by the diphenyleneiodonium chloride (DPI; a ROS inhibitor). Conversely, transfection of dominant-negative (DN) mutant of extracellular signal-regulated kinases 2 (ERK2), c-Jun N-terminal kinase 1 (JNK1), or p38α kinase had no such effect. Simultaneously, RA suppresses ROS generation and lipid peroxidation (LPO) whereas increases cellular GSH in HSC-T6 cells. Furthermore, RA significantly increased antioxidant response element (ARE)-mediated luciferase activity, nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and catalytic subunits from glutamate cysteine ligase (GCLc) expression, but not modulatory subunits from GCL (GCLm). RA-mediated up-regulation of GClc is inhibited by the shRNA-induced Nrf2 knockdown. The knocking down of Nrf2 or buthionine sulfoximine (a GCL inhibitor) abolished RA-mediated inhibition of ROS. Collectively, these results provide novel insights into the mechanisms of RA as an antifibrogenic candidate in the prevention and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Changfang Lu
- The Institute of Medicine, Qiqihar Medical University, Qiqihar 161006, China
| | - Yu Zou
- The Institute of Medicine, Qiqihar Medical University, Qiqihar 161006, China
| | - Yuzhang Liu
- The Institute of Medicine, Qiqihar Medical University, Qiqihar 161006, China
| | - Yingcai Niu
- The Institute of Medicine, Qiqihar Medical University, Qiqihar 161006, China.
| |
Collapse
|
32
|
Dong H, Guo H, Liang Y, Wang X, Niu Y. Astragaloside IV synergizes with ferulic acid to suppress hepatic stellate cells activation in vitro. Free Radic Res 2017; 51:167-178. [PMID: 28147890 DOI: 10.1080/10715762.2017.1290233] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Because hepatic fibrosis usually involves more than one pathological process, combination therapy with modalities that target aberrant signaling cascade in activated hepatic stellate cells (HSCs) represents an alternative strategy. This study evaluates the hypothesis that astragaloside IV (AS-IV) and ferulic acid (FA) synergize to inhibit HSCs activation via simultaneous activating nuclear factor erythroid-2-related factor-2 (Nrf2) and blocking transforming growth factor-β (TGF-β) pathways. The combination of FA and AS-IV, hereafter referred to as the AS-IV/FA, at suboptimal concentrations synergistically inhibited HSCs activation, as measured by expressions of α-smooth muscle actin (α-SMA), collagen α type I (Col I) and fibronectin. Nrf2 nuclear accumulation, glutathione (GSH) increase, and reactive oxygen species (ROS) reduction by AS-IV were not potentiated by co-treatment with FA. Similarly, inhibition of TGF-β1 secretion and Smad activity by FA also was not enhanced by combined treatment with AS-IV. AS-IV/FA synergistically suppresses the p38 mitogen-activated protein kinase (MAPK) activity. Inhibition of HSCs activation by AS-IV/FA could be completely blocked by TGF-βs-neutralizing antibody plus shRNA-mediated knockdown of Nrf2. Dual blockade of the TGF-β1/Smad pathway by FA and activation of Nrf2/ARE pathway by AS-IV contributed to the synergistic effects of this combination treatment. These results suggest that combinatorial treatments that target different pathway may afford a more effective strategy to inhibit HSC activation.
Collapse
Affiliation(s)
- Haiying Dong
- a The Institute of Medicine, Qiqihar Medical University , Qiqihar , China
| | - Hongyan Guo
- a The Institute of Medicine, Qiqihar Medical University , Qiqihar , China
| | - Yini Liang
- a The Institute of Medicine, Qiqihar Medical University , Qiqihar , China
| | - Xing Wang
- a The Institute of Medicine, Qiqihar Medical University , Qiqihar , China
| | - Yingcai Niu
- a The Institute of Medicine, Qiqihar Medical University , Qiqihar , China
| |
Collapse
|
33
|
Padmavathi G, Roy NK, Bordoloi D, Arfuso F, Mishra S, Sethi G, Bishayee A, Kunnumakkara AB. Butein in health and disease: A comprehensive review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 25:118-127. [PMID: 28190465 DOI: 10.1016/j.phymed.2016.12.002] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 12/03/2016] [Accepted: 12/11/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND The risk of suffering from many chronic diseases seems to have made no improvement despite the advancement in medications available in the modern world. Moreover, the use of synthetic chemicals as medications has proved to worsen the scenario due to the various adverse side effects associated with them. PURPOSE Extensive research on natural medicines provides ample evidence on the safety and efficacy of phytochemicals and nutraceuticals against diverse chronic ailments. Therefore, it is advisable to use natural products in the management of such diseases. This article aims to present a comprehensive and critical review of known pharmacological and biological effects of butein, an important chalcone polyphenol first isolated from Rhus verniciflua Stokes, implicated in the prevention and treatment of various chronic disease conditions. METHODS An extensive literature search was conducted using PubMed, ScienceDirect, Scopus and Web of ScienceTM core collections using key words followed by evaluation of the bibliographies of relevant articles. RESULTS Butein has been preclinically proven to be effective against several chronic diseases because it possesses a wide range of biological properties, including antioxidant, anti-inflammatory, anticancer, antidiabetic, hypotensive and neuroprotective effects. Furthermore, it has been shown to affect multiple molecular targets, including the master transcription factor nuclear factor-κB and its downstream molecules. Moreover, since it acts on multiple pathways, the chances of non-responsiveness and resistance development is reduced, supporting the use of butein as a preferred treatment option. CONCLUSION Based on numerous preclinical studies, butein shows significant therapeutic potential against various diseases. Nevertheless, well-designed clinical studies are urgently needed to validate the preclinical findings.
Collapse
Affiliation(s)
- Ganesan Padmavathi
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, Assam 781 039, India
| | - Nand Kishor Roy
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, Assam 781 039, India
| | - Devivasha Bordoloi
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, Assam 781 039, India
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, West Australia 6009, Australia
| | - Srishti Mishra
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Biosciences Research Precinct, Curtin University, Western Australia 6009, Australia.
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, Miami, FL 33169, USA.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, Assam 781 039, India.
| |
Collapse
|
34
|
Islam MS, Segars JH, Castellucci M, Ciarmela P. Dietary phytochemicals for possible preventive and therapeutic option of uterine fibroids: Signaling pathways as target. Pharmacol Rep 2017; 69:57-70. [DOI: 10.1016/j.pharep.2016.10.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/03/2016] [Accepted: 10/19/2016] [Indexed: 02/07/2023]
|
35
|
Butein inhibits IL-1β-induced inflammatory response in human osteoarthritis chondrocytes and slows the progression of osteoarthritis in mice. Int Immunopharmacol 2016; 42:1-10. [PMID: 27863298 DOI: 10.1016/j.intimp.2016.11.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 10/22/2016] [Accepted: 11/11/2016] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is a progressive degenerative disease characterized by irreversible articular cartilage destruction. Butein, a polyphenolic compound isolated from the stem bark of cashews and Rhus verniciflua Stokes, has been reported to have anti-inflammatory effects. This study aimed to assess the effect of butein on human OA chondrocytes and mice OA models induced by destabilization of the medial meniscus (DMM). In vitro, human OA chondrocytes were pretreated with butein at 10, 50μM and subsequently stimulated with IL-1β (10ng/ml) for 24h. Production of NO, PGE2, TNF-α and IL-6 was evaluated by the Griess reaction and ELISAs. The mRNA expression of COX-2, iNOS, TNF-α, IL-6, MMP-1, MMP-3, MMP-13, ADAMTS-4, ADAMTS-5, COL-2 and SOX-9 were measured by real-time PCR. The protein expression of COX-2, iNOS, MMP-13, COL-2, SOX-9, p65 and IκB-α were detected by Western blot. P65 nuclear translocation was detected by immunofluorescence. In vivo, the severity of OA was determined by histological analysis. We found that butein significantly inhibited the IL-1β-induced production of NO and PGE2, expression of COX-2, iNOS, TNF-α, IL-6 and MMP-13, degradation of COL-2 and SOX-9 at mRNA and protein levels as well as MMP-1, MMP-3, ADAMTS-4 and ADAMTS-5 gene expression. Furthermore, butein dramatically suppressed IL-1β-stimulated IκB-α degradation and NF-kB p65 activation. In vivo, the cartilage in butein-treated mice exhibited less Safranin O loss, cartilage erosion and lower OARSI scores. Butein also reduced subchondral bone plate thickness and alleviated synovitis. Taken together, these findings indicate that butein may be a potential agent in the treatment of OA.
Collapse
|
36
|
Yan SL, Wang ZH, Yen HF, Lee YJ, Yin MC. Reversal of ethanol-induced hepatotoxicity by cinnamic and syringic acids in mice. Food Chem Toxicol 2016; 98:119-126. [PMID: 27793734 DOI: 10.1016/j.fct.2016.10.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/21/2016] [Accepted: 10/23/2016] [Indexed: 02/07/2023]
Abstract
Ethanol was used to induce acute hepatotoxicity in mice. Effects of cinnamic acid (CA) and syringic acid (SA) post-intake for hepatic recovery from alcoholic injury was investigated. Ethanol treated mice were supplied by CA or SA at 40 or 80 mg/kg BW/day for 5 days. Results showed that ethanol stimulated protein expression of CYP2E1, p47phox, gp91phox, cyclooxygenase-2 and nuclear factor kappa B in liver. CA or SA post-intake restricted hepatic expression of these molecules. Ethanol suppressed nuclear factor erythroid 2-related factor (Nrf2) expression, and CA or SA enhanced Nrf2 expression in cytosolic and nuclear fractions. Ethanol increased the release of reactive oxygen species, oxidized glutathione, interleukin-6, tumor necrosis factor-alpha, nitric acid and prostaglandin E2. CA or SA lowered hepatic production of these oxidative and inflammatory factors. Histological data revealed that ethanol administration caused obvious foci of inflammatory cell infiltration, and CA or SA post-intake improved hepatic inflammatory infiltration. These findings support that cinnamic acid and syringic acid are potent nutraceutical agents for acute alcoholic liver disease therapy. However, potential additive or synergistic benefits of cinnamic and syringic acids against ethanol-induced hepatotoxicity need to be investigated.
Collapse
Affiliation(s)
- Sheng-Lei Yan
- Division of Gastroenterology, Department of Internal Medicine, Chang Bing Show-Chwan Memorial Hospital, Changhua County, Taiwan
| | - Zhi-Hong Wang
- Department of Nutrition, China Medical University, Taichung City, Taiwan
| | - Hsiu-Fang Yen
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan City, Taiwan
| | - Yi-Ju Lee
- Department of Pathology, Chung Shan Medical University Hospital, Taichung City, Taiwan
| | - Mei-Chin Yin
- Department of Nutrition, China Medical University, Taichung City, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung City, Taiwan.
| |
Collapse
|
37
|
Gil MN, Choi DR, Yu KS, Jeong JH, Bak DH, Kim DK, Lee NS, Lee JH, Jeong YG, Na CS, Na DS, Ryu KH, Han SY. Rhus verniciflua Stokes attenuates cholestatic liver cirrhosis-induced interstitial fibrosis via Smad3 down-regulation and Smad7 up-regulation. Anat Cell Biol 2016; 49:189-198. [PMID: 27722012 PMCID: PMC5052228 DOI: 10.5115/acb.2016.49.3.189] [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: 08/18/2016] [Revised: 09/02/2016] [Accepted: 09/06/2016] [Indexed: 12/21/2022] Open
Abstract
Cholestatic liver cirrhosis (CLC) eventually proceeds to end-stage liver failure by mediating overwhelming deposition of collagen, which is produced by activated interstitial myofibroblasts. Although the beneficial effects of Rhus verniciflua Stokes (RVS) on various diseases are well-known, its therapeutic effect and possible underlying mechanism on interstitial fibrosis associated with CLC are not elucidated. This study was designed to assess the protective effects of RVS and its possible underlying mechanisms in rat models of CLC established by bile duct ligation (BDL). We demonstrated that BDL markedly elevated the serological parameters such as aspartate aminotransferase, alanine transaminase, total bilirubin, and direct bilirubin, all of which were significantly attenuated by the daily uptake of RVS (2 mg/kg/day) for 28 days (14 days before and after operation) via intragastric route. We observed that BDL drastically induced the deterioration of liver histoarchitecture and excessive deposition of extracellular matrix (ECM), both of which were significantly attenuated by RVS. In addition, we revealed that RVS inhibited BDL-induced proliferation and activation of interstitial myofibroblasts, a highly suggestive cell type for ECM production, as shown by immunohistochemical and semi-quantitative detection of α-smooth muscle actin and vimentin. Finally, we demonstrated that the anti-fibrotic effect of RVS was associated with the inactivation of Smad3, the key downstream target of a major fibrogenic cytokine, i.e., transforming growth factor β (TGF-β). Simultaneously, we also found that RVS reciprocally increased the expression of Smad7, a negative regulatory protein of the TGF-β/Smad3 pathway. Taken together, these results suggested that RVS has a therapeutic effect on CLC, and these effects are, at least partly, due to the inhibition of liver fibrosis by the downregulation of Smad3 and upregulation of Smad7.
Collapse
Affiliation(s)
- Mi Na Gil
- Department of Anatomy, Konyang University College of Medicine, Daejeon, Korea
| | - Du Ri Choi
- Department of Anatomy, Konyang University College of Medicine, Daejeon, Korea
| | - Kwang Sik Yu
- Department of Anatomy, Konyang University College of Medicine, Daejeon, Korea
| | - Ji Heun Jeong
- Department of Anatomy, Konyang University College of Medicine, Daejeon, Korea
| | - Dong-Ho Bak
- Department of Dermatology, Chung-Ang University R&D Center, Seoul, Korea
| | - Do-Kyung Kim
- Department of Anatomy, Konyang University College of Medicine, Daejeon, Korea
| | - Nam-Seob Lee
- Department of Anatomy, Konyang University College of Medicine, Daejeon, Korea
| | - Je-Hun Lee
- Department of Anatomy, Konyang University College of Medicine, Daejeon, Korea
| | - Young-Gil Jeong
- Department of Anatomy, Konyang University College of Medicine, Daejeon, Korea
| | | | | | - Ki-Hyun Ryu
- Department of Gastroenterology and Hepatology, Konyang University Hospital, Daejeon, Korea
| | - Seung Yun Han
- Department of Anatomy, Konyang University College of Medicine, Daejeon, Korea.; Myunggok Research Institute, Konyang University College of Medicine, Daejeon, Korea
| |
Collapse
|
38
|
Antioxidant and anti-inflammatory role of zingerone in ethanol-induced hepatotoxicity. Mol Cell Biochem 2016; 421:169-81. [DOI: 10.1007/s11010-016-2798-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/12/2016] [Indexed: 01/01/2023]
|
39
|
Pan JH, Lee KY, Kim JH, Shin H, Lee JH, Kim YJ. Prunus mume Sieb. et Zucc. fruit ameliorates alcoholic liver injury in mice by inhibiting apoptosis and inflammation through oxidative stress. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.04.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
40
|
Bian K, Zhang F, Wang T, Zou X, Duan X, Chen G, Zhuge Y. S-Adenosylmethionine suppresses the expression of Smad3/4 in activated human hepatic stellate cells via Rac1 promoter methylation. Mol Med Rep 2016; 13:3867-73. [PMID: 26986629 PMCID: PMC4838126 DOI: 10.3892/mmr.2016.4997] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 01/20/2016] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate whether S-adenosylmethionine (SAM) was able to suppress activated human hepatic stellate cells (HSCs). Human LX-2 HSCs were cultured with SAM or NSC23766, and were transfected with plasmids encoding ras-related C3 botulinum toxin substrate 1 (Rac1) protein or an empty expression vector. Cell proliferation was detected by Cell Counting Kit-8. Cell migration and invasion were determined using the Transwell assay. The expression levels of Rac1 and Smad3/4 were detected by reverse transcription‑quantitative polymerase chain reaction (PCR) or western blotting. The methylation status of Rac1 promoters was measured by methylation‑specific PCR. The results demonstrated that SAM and NSC23766 suppressed the expression of Smad3/4 in LX‑2 cells. The overexpression of Rac1 enhanced the proliferation, migration and invasion of LX‑2 cells. In addition, compared with the control groups, a marked increase was observed in the protein expression levels of Smad3/4 in the LX‑2 cells transfected with Rac1 plasmids. The methylation-specific PCR findings showed that SAM increased the methylation of Rac1 promoters. The results of the present study suggested that Rac1 enhanced the expression of Smad3/4 in activated HSCs; however, this increase may be suppressed by SAM-induced methylation of Rac1 promoters.
Collapse
Affiliation(s)
- Kangqi Bian
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Feng Zhang
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Tingting Wang
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Xiaoping Zou
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Xuhong Duan
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Guangxia Chen
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Yuzheng Zhuge
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
41
|
Gu L, Tao X, Xu Y, Han X, Qi Y, Xu L, Yin L, Peng J. Dioscin alleviates BDL- and DMN-induced hepatic fibrosis via Sirt1/Nrf2-mediated inhibition of p38 MAPK pathway. Toxicol Appl Pharmacol 2016; 292:19-29. [DOI: 10.1016/j.taap.2015.12.024] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/29/2015] [Accepted: 12/29/2015] [Indexed: 01/08/2023]
|
42
|
Boye A, Zou YH, Yang Y. Metabolic derivatives of alcohol and the molecular culprits of fibro-hepatocarcinogenesis: Allies or enemies? World J Gastroenterol 2016; 22:50-71. [PMID: 26755860 PMCID: PMC4698508 DOI: 10.3748/wjg.v22.i1.50] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 10/12/2015] [Accepted: 11/19/2015] [Indexed: 02/06/2023] Open
Abstract
Chronic intake of alcohol undoubtedly overwhelms the structural and functional capacity of the liver by initiating complex pathological events characterized by steatosis, steatohepatitis, hepatic fibrosis and cirrhosis. Subsequently, these initial pathological events are sustained and ushered into a more complex and progressive liver disease, increasing the risk of fibro-hepatocarcinogenesis. These coordinated pathological events mainly result from buildup of toxic metabolic derivatives of alcohol including but not limited to acetaldehyde (AA), malondialdehyde (MDA), CYP2E1-generated reactive oxygen species, alcohol-induced gut-derived lipopolysaccharide, AA/MDA protein and DNA adducts. The metabolic derivatives of alcohol together with other comorbidity factors, including hepatitis B and C viral infections, dysregulated iron metabolism, abuse of antibiotics, schistosomiasis, toxic drug metabolites, autoimmune disease and other non-specific factors, have been shown to underlie liver diseases. In view of the multiple etiology of liver diseases, attempts to delineate the mechanism by which each etiological factor causes liver disease has always proved cumbersome if not impossible. In the case of alcoholic liver disease (ALD), it is even more cumbersome and complicated as a result of the many toxic metabolic derivatives of alcohol with their varying liver-specific toxicities. In spite of all these hurdles, researchers and experts in hepatology have strived to expand knowledge and scientific discourse, particularly on ALD and its associated complications through the medium of scientific research, reviews and commentaries. Nonetheless, the molecular mechanisms underpinning ALD, particularly those underlying toxic effects of metabolic derivatives of alcohol on parenchymal and non-parenchymal hepatic cells leading to increased risk of alcohol-induced fibro-hepatocarcinogenesis, are still incompletely elucidated. In this review, we examined published scientific findings on how alcohol and its metabolic derivatives mount cellular attack on each hepatic cell and the underlying molecular mechanisms leading to disruption of core hepatic homeostatic functions which probably set the stage for the initiation and progression of ALD to fibro-hepatocarcinogenesis. We also brought to sharp focus, the complex and integrative role of transforming growth factor beta/small mothers against decapentaplegic/plasminogen activator inhibitor-1 and the mitogen activated protein kinase signaling nexus as well as their cross-signaling with toll-like receptor-mediated gut-dependent signaling pathways implicated in ALD and fibro-hepatocarcinogenesis. Looking into the future, it is hoped that these deliberations may stimulate new research directions on this topic and shape not only therapeutic approaches but also models for studying ALD and fibro-hepatocarcinogenesis.
Collapse
|
43
|
Abstract
Natural compounds isolated from various plant sources have been used for therapeutic purpose for centuries. These compounds have been routinely used for the management of various chronic ailments and have gained considerable attention because of their significant efficacy and comparatively low side effects. Butein, a chacolnoid compound that has been isolated from various medicinal plants has exhibited a wide range of beneficial pharmacological effects, such as anti-inflammatory, anticancer, antioxidant, and anti-angiogenic in diverse disease models. This article briefly summarizes the past published literature related to the therapeutic and protective effects of butein, as demonstrated in various models of human chronic diseases. Further analysis of its important cellular targets, toxicity, and pharmacokinetic profile may further significantly expand its therapeutic application.
Collapse
|
44
|
Padmavathi G, Rathnakaram SR, Monisha J, Bordoloi D, Roy NK, Kunnumakkara AB. Potential of butein, a tetrahydroxychalcone to obliterate cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:1163-1171. [PMID: 26598915 DOI: 10.1016/j.phymed.2015.08.015] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 08/15/2015] [Accepted: 08/23/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Despite the major advances made in the field of cancer biology, it still remains one of the most fatal diseases in the world. It is now well established that natural products are safe and efficacious and have high potential in the prevention and treatment of different diseases including cancer. Butein is one such compound which is now found to have anti-cancer properties against various malignancies. PURPOSE To thoroughly review the literature available on the anti-cancer properties of butein against different cancers and its molecular targets. METHODS A thorough literature search has been done in PubMed for butein, its biological activities especially cancer and its molecular targets. RESULTS Our search retrieved several reports on the various biological activities of butein in which around 43 articles reported that butein shows potential anti-proliferative effect against a wide range of neoplasms and the molecular target varies with cancer types. Most often it targets NF-κB and its downstream pathways. In addition, butein induces the expression of genes which mediate the cell death and apoptosis in cancer cells. It also inhibits tumor angiogenesis, invasion and metastasis in prostate, liver and bladder cancers through the inhibition of MMPs, VEGF etc. Moreover, it inhibits the overexpression of several proteins and enzymes such as STAT3, ERK, CXCR4, COX-2, Akt, EGFR, Ras etc. involved in tumorigenesis. CONCLUSION Collectively, all these findings suggest the enormous potential and efficacy of butein as a multitargeted chemotherapeutic, chemopreventive and chemosensitizing agent against a wide range of cancers with minimal or no adverse side effects.
Collapse
Affiliation(s)
- Ganesan Padmavathi
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Sivakumar Raju Rathnakaram
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Javadi Monisha
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Devivasha Bordoloi
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Nand Kishor Roy
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India .
| |
Collapse
|
45
|
Xu P, Zhang Y, Liu Y, Yuan Q, Song L, Liu M, Liu Z, Yang Y, Li J, Li D, Ren G. Fibroblast growth factor 21 attenuates hepatic fibrogenesis through TGF-β/smad2/3 and NF-κB signaling pathways. Toxicol Appl Pharmacol 2015; 290:43-53. [PMID: 26592322 DOI: 10.1016/j.taap.2015.11.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/06/2015] [Accepted: 11/20/2015] [Indexed: 02/06/2023]
Abstract
Fibroblast growth factor 21 (FGF-21) is a secreted protein, which has anti-diabetic and lipocaic effects, but its ability to protect against hepatic fibrosis has not been studied. In this study, we investigated the ability of FGF-21 to attenuate dimethylnitrosamine (DMN)-induced hepatic fibrogenesis in mice and the mechanism of its action. Hepatic fibrosis was induced by injection of DMN, FGF-21 was administered to the mice once daily in association with DMN injection till the end of the experiment. Histopathological examination, tissue 4-hydroxyproline content and expressions of smooth muscle α-actin (α-SMA) and collagen I were measured to assess hepatic fibrosis. Ethanol/PDGF-BB-activated hepatic stellate cells (HSCs) were used to understand the mechanisms of FGF-21 inhibited hepatic fibrogenesis. Results showed that FGF-21 treatment attenuated hepatic fibrogenesis and was associated with a significant decrease in intrahepatic fibrogenesis, 4-hydroxyproline accumulation, α-SMA expression and collagen I deposition. FGF-21 treatment inhibited the activation of HSCs via down-regulating the expression of TGF-β, NF-κB nuclear translocation, phosphorylation levels of smad2/3 and IκBα. Besides, FGF-21 treatment caused activated HSC apoptosis with increasing expression of Caspase-3, and decreased the ratio of Bcl-2 to Bax. In conclusion, FGF-21 attenuates hepatic fibrogenesis and inhibits the activation of HSC warranting the use of FGF-21 as a potential therapeutic agent in the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Pengfei Xu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, PR China
| | - Yingjie Zhang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, PR China
| | - Yunye Liu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, PR China
| | - Qingyan Yuan
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, PR China
| | - Liying Song
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, PR China
| | - Mingyao Liu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, PR China
| | - Zhihang Liu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, PR China
| | - Yongbi Yang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, PR China
| | - Junyan Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, PR China
| | - Deshan Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, PR China.
| | - Guiping Ren
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, PR China.
| |
Collapse
|
46
|
Roy S, Benz F, Vargas Cardenas D, Vucur M, Gautheron J, Schneider A, Hellerbrand C, Pottier N, Alder J, Tacke F, Trautwein C, Roderburg C, Luedde T. miR-30c and miR-193 are a part of the TGF-β-dependent regulatory network controlling extracellular matrix genes in liver fibrosis. J Dig Dis 2015; 16:513-24. [PMID: 26120970 DOI: 10.1111/1751-2980.12266] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE MicroRNAs (miRNAs) have recently emerged as novel regulators in liver fibrosis. miR-30c and miR-193 are involved in fibrotic remodeling processes and cancer development, respectively. This study aimed to explore the role of miR-30c and miR-193 in liver fibrosis. METHODS The regulation of miRNAs in carbon tetrachloride-induced liver fibrosis was analyzed by microarray. Expression patterns of miR-193 and miR-30c were further confirmed in fibrotic liver samples obtained from two murine models of hepatic fibrosis and human tissues. On a functional level, miRNA levels were analyzed in the context of transforming growth factor (TGF-β) mediated activation of hepatic stellate cells (HSCs). Finally, predicted targets were assessed for their roles in fibrosis by transfecting murine HSCs with miRNA mimics. RESULTS Microarray analysis in murine fibrotic livers revealed a panel of 44 dysregulated miRNAs. In addition to previously established miRNAs known to be regulated in liver fibrosis in a TGF-β-dependent manner (e.g., miR-29, miR-133), miR-193 and miR-30c were observed to be specifically downregulated not only in experimental hepatofibrogenesis but also in human liver fibrosis, while they showed a reciprocal expression pattern after recovery from liver fibrosis. Functional experiments confirmed the TGF-β-dependent downregulation of these respective new miRNAs in HSCs. Finally, we identified TGF-β2 and SNAIL1, important regulators of extracellular matrix, as potential target genes of miR-193 and miR-30 in liver fibrosis. CONCLUSION These results suggest that miR-30 and miR-193 are members of a network of miRNAs modifying the TGF-β-dependent regulation of extracellular matrix-related genes in HSCs in the manifestation and resolution of liver fibrosis.
Collapse
Affiliation(s)
- Sanchari Roy
- Department of Medicine III, University of Aachen (RWTH), Aachen, Germany
| | - Fabian Benz
- Department of Medicine III, University of Aachen (RWTH), Aachen, Germany
| | | | - Mihael Vucur
- Department of Medicine III, University of Aachen (RWTH), Aachen, Germany
| | - Jeremie Gautheron
- Department of Medicine III, University of Aachen (RWTH), Aachen, Germany
| | - Anne Schneider
- Department of Medicine III, University of Aachen (RWTH), Aachen, Germany
| | - Claus Hellerbrand
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Nicolas Pottier
- EA4483, Faculté de Médecine de Lille, Pole Recherche, Lille, France
| | - Jan Alder
- Department of Medicine III, University of Aachen (RWTH), Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, University of Aachen (RWTH), Aachen, Germany
| | | | | | - Tom Luedde
- Department of Medicine III, University of Aachen (RWTH), Aachen, Germany
| |
Collapse
|
47
|
Modulation of IKKβ/NF-κB and TGF-β1/Smad via Fuzheng Huayu recipe involves in prevention of nutritional steatohepatitis and fibrosis in mice. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2015; 18:404-11. [PMID: 26019805 PMCID: PMC4439457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 11/24/2014] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Fuzheng Huayu recipe (FZHY) exerts significant protective effects against liver fibrosis by strengthening the body's resistance and removing blood stasis. However, the molecular mechanisms through which FZHY affects liver fibrosis are still unclear. In this study, we examined the expression levels of factors involved in the inhibitor κB kinase-β (IKK-β)/nuclear factor-κB (NF-κB) and transforming growth factor beta 1 (TGF-β1)/Smad signaling pathways to elucidate whether FZHY could attenuate nutritional steatohepatitis and fibrosis in mice. MATERIALS AND METHODS C57BL/6J mice were fed with methionine-choline deficient (MCD) diet for 8 weeks to induce fibrotic steatohepatitis. FZHY and/or heme oxygenase-1 (HO-1) chemical inducer (hemin) were administered to mice. The effects of FZHY alone and in combination with hemin were assessed by comparing the severity of hepatic injury, activation of hepatic stellate cells (HSCs), and the expression of oxidative stress, inflammation and fibrogenesis related genes. RESULTS Administration of FZHY, hemin and FZHY plus hemin significantly ameliorated liver injury. Additionally, our analysis indicated that administration of these agents significantly attenuated oxidative stress, downregulated the expression of pro-inflammatory and pro-fibrotic genes, including IKK-β, NF-κB, monocyte chemoattractant protein-1 (MCP-1), α-smooth muscle actin (α-SMA), TGF-β1, Smad3 and Smad4, and upregulated the expression of the antifibrogenic gene Smad7 (P< 0.001). CONCLUSION FZHY-containing therapies prevented nutritional steatohepatitis and fibrosis through modulating the expression of factors associated with the IKKβ/NF-κB and TGF-β1/Smad signaling pathways and oxidative stress related genes.
Collapse
|
48
|
Yan SL, Yang HT, Lee HL, Yin MC. Protective effects of maslinic acid against alcohol-induced acute liver injury in mice. Food Chem Toxicol 2014; 74:149-55. [PMID: 25301236 DOI: 10.1016/j.fct.2014.09.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 08/21/2014] [Accepted: 09/25/2014] [Indexed: 12/11/2022]
Abstract
Protective effects of maslinic acid (MA) at 10, 15 or 20 mg/kg body weight/day against alcohol-induced acute hepatotoxicity in mice were examined. Mice were administrated by MA for 3 weeks, and followed by alcohol treatment. Results showed that MA pre-intake at three doses resulted in its accumulation in the liver; and dose-dependently lowered cytochrome P450 2E1 activity and protein expression at 23.5-51.2% and 21.4-62.3%, respectively (P <0.05). MA pre-intake decreased subsequent alcohol-induced reactive oxygen species, interleukin-6, tumor necrosis factor-alpha, monocyte chemoattractant protein-1, nitric oxide and prostaglandin E2 production; retained glutathione content; maintained catalase and glutathione peroxidase activities; and declined cyclooxygenase-2 and total nitric oxide synthase activities in the liver (P <0.05). Furthermore, MA pre-intake suppressed 17.3-51.7% nuclear factor kappa (NF-κ)B p50, 23.5-58.8% NF-κB p65, 25.6-62.4% p-p38 and 24.1-63.0% p-JNK expression in the liver (P <0.05). Histological data indicated that MA intake at test doses attenuated hepatic inflammatory infiltrate. These findings support that maslinic acid is a potent preventive agent against acute alcoholic liver disease.
Collapse
Affiliation(s)
- Sheng-lei Yan
- Division of Gastroenterology, Department of Internal Medicine, Chang Bing Show-Chwan Memorial Hospital, Changhua County, Taiwan
| | - Hui-ting Yang
- Department of Nutrition, China Medical University, Taichung City, Taiwan
| | - Hsiang-lin Lee
- Department of Surgery, Chung Shan Medical University Hospital, Taichung City, Taiwan
| | - Mei-chin Yin
- Department of Nutrition, China Medical University, Taichung City, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung City, Taiwan.
| |
Collapse
|
49
|
TGF-β1-elevated TRPM7 channel regulates collagen expression in hepatic stellate cells via TGF-β1/Smad pathway. Toxicol Appl Pharmacol 2014; 280:335-44. [PMID: 25150141 DOI: 10.1016/j.taap.2014.08.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Revised: 08/06/2014] [Accepted: 08/08/2014] [Indexed: 12/13/2022]
Abstract
Transdifferentiation of hepatic stellate cells (HSCs) into myofibroblasts plays a critical role in the development of liver fibrosis, since myofibroblasts are the key cells responsible for excessive deposition of ECM proteins. Transient receptor potential melastatin 7 (TRPM7), a non-selective cation channel with protein serine/threonine kinase activity, has been demonstrated to function in the proliferation of activated HSCs. Here, we investigated the functional role of TRPM7 in collagen deposition in activated HSC-T6 cells (a rat hepatic stellate cell line). TRPM7 mRNA and protein were measured by Real-time PCR and Western blot in TGF-β1-activated HSC-T6 cells in vitro. Results demonstrated that TRPM7 protein was dramatically increased in fibrotic human livers. Stimulation of HSC-T6 cells with TGF-β1 increased TRPM7 mRNA and protein level in a time-dependent manner. Nevertheless, TGF-β1-elicited upregulation of TRPM7 in HSC-T6 cells was abrogated by SB431542 (TGF-β1 receptor blocker) or SIS3 (inhibitor of Smad3 phosphorylation). Additionally, blockade of TRPM7 channels with non-specific TRPM7 blocker 2-APB or synthetic siRNA targeting TRPM7 attenuated TGF-β1-induced expression of myofibroblast markers, as measured by the induction of α-SMA and Col1α1. Silencing TRPM7 also increased the ratio of MMPs/TIMPs by increasing MMP-13 expression and decreasing TIMP-1 and TIMP-2 levels. Strikingly, phosphorylation of p-Smad2 and p-Smad3, associated with collagen production, was decreased in TRPM7 deficient HSC-T6 cells. These observations suggested that TGF-β1 elevates TRPM7 expression in HSCs via Smad3-dependant mechanisms, which in turn contributes Smad protein phosphorylation, and subsequently increases fibrous collagen expression. Therefore, TRPM7 may constitute a useful target for the treatment of liver fibrosis.
Collapse
|
50
|
Yu SS, Zhu X. Role of NADPH oxidase family members in promoting liver fibrosis. Shijie Huaren Xiaohua Zazhi 2014; 22:2710-2715. [DOI: 10.11569/wcjd.v22.i19.2710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is one of hepatic wound-repair responses to a variety of chronic liver injuries, which is characterized by excessive deposition of extracellular matrix. Increasing evidence indicates that nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and oxidative stress caused by reactive oxygen species play a key role in liver fibrosis. NADPH oxidase is a multi-subunit complex. In the liver, both phagocytic and non-phagocytic NADPH oxidases are functionally expressed. They have a significant fibrogenic effect on the hepatic stellate cells, the main cell type causing liver fibrosis. In this paper, we review the recent advances in understanding the role of the NADPH oxidase family in the occurrence and development of liver fibrosis.
Collapse
|