1
|
Wu Z, Shi J, Zhang Y, Shi R, Guo Q, Zhang J, Lu B, Huang Z, Ji L. Uncovering the pharmacological mechanism and the main herbal medicine contributing to the efficacy of Xiaoyanlidan Tablet (XYLDT) in treating cholestatic liver injury. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119163. [PMID: 39613007 DOI: 10.1016/j.jep.2024.119163] [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: 09/26/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiaoyanlidan Tablet (XYLDT) is a Chinese patent medicine consisted of three traditional Chinese medicines (TCMs) including Andrographis Herba (AH), Linearstripe Rabdosia Herba (LRH) and Picrasmae Ramulus et Folium (PRF). In Chinese traditional medicine theory, XYLDT has the "heat-clearing, dampness-dispelling and gallbladder function promoting" properties, and was widely used in the clinic for decades to treat pain in the subcostal region or bitter taste in the mouth, which were induced by liver-gallbladder dampness-heat. Meanwhile, it was also used for the therapy of acute cholecystitis and cholangitis. AIM OF THE STUDY To explore the mechanism of XYLDT in alleviating the alpha-naphthylisothiocyanate (ANIT)-induced cholestatic liver injury (CLI), and to find out which TCM consisted in XYLDT contributed the most to the therapeutic efficacy of XYLDT. METHODS ANIT was orally given to mice to induce CLI in vivo. Each TCM in XYLDT alone, XYLDT-without one TCM or XYLDT was orally given to mice before or after ANIT administration. Serum biochemical indicators were measured by using commercial kits. Liver histopathology was observed. Clinical data analysis was used to predict molecules and signal pathways involved in the XYLDT-provided improvement on CLI, which was further verified by using RT-PCR and Western-blot assay. RESULTS The alleviation of XYLDT on ANIT-induced CLI was proved by the data of serum biochemical indicators and liver histological observation. Results from clinical data analysis indicated that XYLDT improved CLI via improving mitochondrial function, oxidative phosphorylation, oxidative stress. XYLDT reduced the ROS level, MDA content, and increased GSH content. Meanwhile XYLDT improved the level of Nrf2 into the nucleus and mRNA expression of Nqo1, Gclc, Gclm. Andrographis Herba was proved to be the most crucial for the XYLDT-provided therapeutic efficacy on CLI. Moreover, andrographolide and neoandrographolide, two main active compounds in Andrographis Herba, had the apparent anti-inflammatory ability in LPS-stimulated RAW264.7 cells. Andrographolide also promoted nuclear translocation activation of Nrf2 in antioxidant response elements (ARE)-luciferin transfected L-02 cells. CONCLUSION XYLDT alleviated the ANIT-induced CLI via improving oxidative stress and activated Nrf2-related signaling pathways. Andrographis Herba was important for the XYLDT-provided alleviation on CLI.
Collapse
Affiliation(s)
- Zeqi Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jionghua Shi
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yue Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ruijia Shi
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qian Guo
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jingnan Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bin Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhenlin Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Lili Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
2
|
Tran QA, Tran GV, Velic S, Xiong HM, Kaur J, Moosavi Z, Nguyen P, Duong N, Luu VT, Singh G, Bui T, Rose M, Ho L. Effects of Astragaloside IV and Formononetin on Oxidative Stress and Mitochondrial Biogenesis in Hepatocytes. Int J Mol Sci 2025; 26:774. [PMID: 39859490 PMCID: PMC11765978 DOI: 10.3390/ijms26020774] [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: 12/06/2024] [Revised: 01/07/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Over-accumulation of reactive oxygen species (ROS) causes hepatocyte dysfunction and apoptosis that might lead to the progression of liver damage. Sirtuin-3 (SIRT3), the main NAD+-dependent deacetylase located in mitochondria, has a critical role in regulation of mitochondrial function and ROS production as well as in the mitochondrial antioxidant mechanism. This study explores the roles of astragaloside IV (AST-IV) and formononetin (FMR) in connection with SIRT3 for potential antioxidative effects. It was shown that the condition of combined pre- and post-treatment with AST-IV or FMR at all concentrations statistically increased and rescued cell proliferation. ROS levels were not affected by pre-or post-treatment individually with AST-IV or pre-treatment with FMR; however, post-treatment with FMR resulted in significant increases in ROS in all groups. Significant decreases in ROS levels were seen when pre- and post-treatment with AST-IV were combined at 5 and 10 μM, or FMR at 5 and 20 μM. In the condition of combined pre- and post-treatment with 10 μM AST-IV, there was a significant increase in SOD activity, and the transcriptional levels of Sod2, Cat, and GPX1 in all treatment groups, which is indicative of reactive oxygen species detoxification. Furthermore, AST-IV and FMR activated PGC-1α and AMPK as well as SIRT3 expression in AML12 hepatocytes exposed to t-BHP-induced oxidative stress, especially at high concentrations of FMR. This study presents a novel mechanism whereby AST-IV and FMR yield an antioxidant effect through induction of SIRT3 protein expression and activation of an antioxidant mechanism as well as mitochondrial biogenesis and mitochondrial content and potential. The findings suggest these agents can be used as SIRT3 modulators in treating oxidative-injury hepatocytes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Linh Ho
- College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA
| |
Collapse
|
3
|
Mohamed Azar KAH, Ezhilarasan D, Karthick M, Shree Harini K, Kumar V. Coleus vettiveroides Root Extract Protects Against Thioacetamide-Induced Chronic Liver Injury by Inhibiting NF-κB Signaling Pathway. ENVIRONMENTAL TOXICOLOGY 2024. [PMID: 39705085 DOI: 10.1002/tox.24465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 08/05/2024] [Accepted: 12/08/2024] [Indexed: 12/22/2024]
Abstract
The roots of Coleus vettiveroides (CV) have been traditionally used in Indian medicinal systems such as Ayurveda and Siddha for its antioxidant, anti-inflammatory, and antidiabetic effects. This study examines the antifibrotic potential of CV ethanolic root extract (CVERE) against thioacetamide (TAA)-induced liver fibrosis in Wistar rats. TAA was administered via i.p., thrice weekly for 11 weeks to induce liver fibrosis in rats. In separate groups, rats were administered with TAA and were concurrently treated with CVERE 125 mg/kg, CVERE 250 mg/kg, and silymarin (SIL) 100 mg/kg. Liver marker enzymes of hepatotoxicity, oxidative stress markers, proinflammatory marker gene expression (TNF-α, NF-κB, COX, and ILs), fibrotic marker gene expression (collagen I and III), immune histochemical expression of fibrosis marker proteins, and histopathologic changes were analyzed. TAA administration led to a significant (p < 0.001) increase in the serum level of hepatotoxic marker enzymes. The TAA-treated group showed higher levels (p < 0.001) of MDA and reduced activities of SOD and CAT in the liver. TAA administration increased CYP2E1 expression, proinflammatory, and fibrotic marker gene expressions in rat liver. The histopathology of the liver confirms TAA-induced architectural distortion and fibrotic changes. CVERE and SIL simultaneous treatments significantly protected against TAA-induced oxidative stress, inflammation, and liver fibrosis. In conclusion, CVERE inhibited TAA-induced liver fibrosis through downregulation of TAA metabolic activation, redox imbalance, and inflammation through repression of the NF-κB pathway.
Collapse
Affiliation(s)
- Kadmad Abdul Hameed Mohamed Azar
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, India
| | - Devaraj Ezhilarasan
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, India
| | - Munusamy Karthick
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, India
| | - Karthik Shree Harini
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, India
| | - Venkatesan Kumar
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
4
|
Ezhilarasan D, Karthikeyan S, Najimi M, Vijayalakshmi P, Bhavani G, Jansi Rani M. Preclinical liver toxicity models: Advantages, limitations and recommendations. Toxicology 2024; 511:154020. [PMID: 39637935 DOI: 10.1016/j.tox.2024.154020] [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: 10/21/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Experimental animal models are crucial for elucidating the pathophysiology of liver injuries and for assessing new hepatoprotective agents. Drugs and chemicals such as acetaminophen, isoniazid, valproic acid, ethanol, carbon tetrachloride (CCl4), dimethylnitrosamine (DMN), and thioacetamide (TAA) are metabolized by the CYP2E1 enzyme, producing hepatotoxic metabolites that lead to both acute and chronic liver injuries. In experimental settings, acetaminophen (centrilobular necrosis), carbamazepine (centrilobular necrosis and inflammation), sodium valproate (necrosis, hydropic degeneration and mild inflammation), methotrexate (sinusoidal congestion and inflammation), and TAA (centrilobular necrosis and inflammation) are commonly used to induce various types of acute liver injuries. Repeated and intermittent low-dose administration of CCl4, TAA, and DMN activates quiescent hepatic stellate cells, transdifferentiating them into myofibroblasts, which results in abnormal extracellular matrix production and fibrosis induction, more rapidly with DMN and CCL4 than TAA (DMN > CCl4 > TAA). Regarding toxicity and mortality, CCl4 is more toxic than DMN and TAA (CCl4 > DMN > TAA). Models used to induce metabolic dysfunction-associated liver disease (MAFLD) vary, but MAFLD's multifactorial nature driven by factors like obesity, fatty liver, dyslipidaemia, type II diabetes, hypertension, and cardiovascular disease makes it challenging to replicate human metabolic dysfunction-associated steatohepatitis accurately. From an experimental point of view, the degree and pattern of liver injury are influenced by various factors, including the type of hepatotoxic agent, exposure duration, route of exposure, dosage, frequency of administration, and the animal model utilized. Therefore, there is a pressing need for standardized protocols and regulatory guidelines to streamline the selection of animal models in preclinical studies.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India.
| | - Sivanesan Karthikeyan
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Paramasivan Vijayalakshmi
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India; Department of Pharmacology, Asan Memorial Dental College and Hospital, Chengalpattu, Tamil Nadu, India
| | - Ganapathy Bhavani
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India; Department of Pharmacology, Meenakshi Ammal Dental College and Hospital, Meenakshi Academy of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Muthukrishnan Jansi Rani
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| |
Collapse
|
5
|
Ezhilarasan D, Langeswaran K. Hepatocellular Interactions of Potential Nutraceuticals in the Management of Inflammatory NAFLD. Cell Biochem Funct 2024; 42:e4112. [PMID: 39238138 DOI: 10.1002/cbf.4112] [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: 04/06/2024] [Revised: 07/17/2024] [Accepted: 08/09/2024] [Indexed: 09/07/2024]
Abstract
Numerous studies highlight the potential of natural antioxidants, such as those found in foods and plants, to prevent or treat nonalcoholic fatty liver disease (NAFLD). Inflammation is a key factor in the progression from high-fat diet-induced NAFLD to nonalcoholic steatohepatitis (NASH). Injured liver cells and immune cells release inflammatory cytokines, activating hepatic stellate cells. These cells acquire a profibrogenic phenotype, leading to extracellular matrix accumulation and fibrosis. Persistent fibrosis can progress to cirrhosis. Fatty infiltration, oxidative stress, and inflammation exacerbate fatty liver diseases. Thus, many plant-derived antioxidants, like silymarin, silibinin, curcumin, resveratrol, berberine, and quercetin, have been extensively studied in experimental models and clinical patients with NAFLD. Experimentally, these compounds have shown beneficial effects in reducing lipid accumulation, oxidative stress, and inflammatory markers by modulating the ERK, NF-κB, AMPKα, and PPARγ pathways. They also help decrease metabolic endotoxemia, intestinal permeability, and gut inflammation. Clinically, silymarin and silibinin have been found to reduce transaminase levels, while resveratrol and curcumin help alleviate inflammation in NAFLD patients. However, these phytocompounds exhibit poor water solubility, leading to low oral bioavailability and hindering their biological efficacy. Additionally, inconclusive clinical results highlight the need for further trials with larger populations, longer durations, and standardized protocols.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Hepatology and Molecular Medicine Lab, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, India
| | - Kulanthaivel Langeswaran
- Department of Biomedical Science, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, India
| |
Collapse
|
6
|
Yan X, Chen X, Zhang X, Qureshi A, Wang Y, Tang X, Hu T, Zhuang H, Ran X, Ma G, Luo P, Shen L. Proteomic analysis of the effects of Dictyophora polysaccharide on arsenic-induced hepatotoxicity in rats. Exp Mol Pathol 2024; 138:104910. [PMID: 38876078 DOI: 10.1016/j.yexmp.2024.104910] [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: 09/20/2023] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 06/16/2024]
Abstract
Arsenic (As) is a highly toxic environmental toxicant and a known human carcinogen. Long-term exposure to As can cause liver injury. Dictyophora polysaccharide (DIP) is a biologically active natural compound found in the Dictyophora with excellent antioxidation, anti-inflammation, and immune protection properties. In this study, the Sprague-Dawley (SD) rat model of As toxicity was established using a feeding method, followed by DIP treatment in rats with As-induced liver injury. The molecular mechanisms of As toxicity to the rat liver and the protective effect of DIP were investigated by proteomic studies. The results showed that 172, 328 and 191 differentially expressed proteins (DEPs) were identified between the As-exposed rats versus control rats (As/Ctrl), DIP treated rats versus As-exposed rats (DIP+As/As), and DIP treated rats versus control rats (DIP+As /Ctrl), respectively. Among them, the expression of 90 DEPs in the As/Ctrl groups was reversed by DIP treatment. As exposure caused dysregulation of metabolic pathways, mitochondria, oxidative stress, and apoptosis-related proteins in the rat liver. However, DIP treatment changed or restored the levels of these proteins, which attenuated the damage to the livers of rats caused by As exposure. The results provide new insights into the mechanisms of liver injury induced by As exposure and the treatment of DIP in As poisoning.
Collapse
Affiliation(s)
- Xi Yan
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China
| | - Xiaolu Chen
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China
| | - Xinglai Zhang
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China
| | - Ayesha Qureshi
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, PR China
| | - Yi Wang
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China
| | - Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, PR China
| | - Ting Hu
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China
| | - Hongbin Zhuang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, PR China
| | - Xiaoqian Ran
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China
| | - Guanwei Ma
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China
| | - Peng Luo
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China.
| | - Liming Shen
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China; College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, PR China.
| |
Collapse
|
7
|
Ezhilarasan D. Deciphering the molecular pathways of saroglitazar: A dual PPAR α/γ agonist for managing metabolic NAFLD. Metabolism 2024; 155:155912. [PMID: 38609038 DOI: 10.1016/j.metabol.2024.155912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
Saroglitazar (SARO), a dual peroxisome proliferator activated receptor (PPAR)-α/γ agonist, has been used to treat metabolic diseases such as insulin resistance and diabetic dyslipidemia in patients with non-alcoholic fatty liver disease (NAFLD). SARO, administered at a dose of 4 mg/day, has been consistently studied in clinical trials with different time points ranging from 4 to 24 weeks with NAFLD patients. Due to its PPAR-γ agonistic action, SARO prevents adipose tissue-mediated fatty acid delivery to the liver by increasing insulin sensitivity and regulating adiponectin and leptin levels in adipose tissue. In hepatocytes, SARO induces fatty acid β-oxidation in mitochondria and transcriptionally activates lipid metabolizing genes in peroxisomes. SARO inhibits insulin resistance, thereby preventing the activation of sterol regulatory element-binding proteins -1c and carbohydrate response element binding protein in hepatocytes through its PPAR-α agonistic action. SARO treatment reduces lipotoxicity-mediated oxidative stress by activating the nuclear factor erythroid 2-related factor 2 and transcriptionally expressing the antioxidants from the antioxidant response element in the nucleus through its PPAR-γ agonistic action. SARO provides a PPAR-α/γ-mediated anti-inflammatory effect by preventing the phosphorylation of mitogen-activated protein kinases (JNK and ERK) and nuclear factor kappa B in hepatocytes. Additionally, SARO interferes with transforming growth factor-β/Smad downstream signaling, thereby reducing liver fibrosis progression through its PPAR-α/γ agonistic actions. Thus, SARO improves insulin resistance and dyslipidemia in NAFLD, reduces lipid accumulation in the liver, and thereby prevents mitochondrial toxicity, oxidative stress, inflammation, and fibrosis progression. This review summarizes the possible molecular mechanism of SARO in the NAFLD.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu 600 077, India.
| |
Collapse
|
8
|
Panritdum P, Muangnoi C, Tuntipopipat S, Charoenkiatkul S, Sukprasansap M. Cleistocalyx nervosum var. paniala berry extract and cyanidin-3-glucoside inhibit hepatotoxicity and apoptosis. Food Sci Nutr 2024; 12:2947-2962. [PMID: 38628219 PMCID: PMC11016384 DOI: 10.1002/fsn3.3975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/14/2023] [Accepted: 01/06/2024] [Indexed: 04/19/2024] Open
Abstract
Excessive oxidative toxicity in liver cells is a significant risk factor that can cause cellular injury, leading to the development of chronic liver disease (CLD). Natural anthocyanins have been shown to prevent the harmful effects of oxidative toxicity in mammalian cells. Ripe Cleistocalyx nervosum var. paniala berry fruits are rich in anthocyanins, which have been reported to possess many health benefits. Therefore, this study examined the protective effect of ethanolic fruit extract of C. nervosum var. paniala (CNPE) against hydrogen peroxide (H2O2)-induced oxidative damage and cell death in human hepatoma HepG2 cells. Results showed that CNPE had strong antioxidant capabilities and high amounts of total phenolics and anthocyanins. HPLC analysis showed that CNPE consists of cyanidin-3-glucoside (C3G). Our investigations found that HepG2 cells pretreated with CNPE or anthocyanin C3G inhibited H2O2-induced cellular damage and apoptosis by increasing the viability of cells, the expression of antiapoptotic Bcl-2 protein, and the activities of cellular antioxidant enzymes, namely SOD, CAT, and GPx. Moreover, both CNPE and C3G significantly suppressed expression of apoptotic proteins (Bax and cytochrome c) and the activities of cleaved caspase-9 and caspase-3 caused by H2O2. Our results indicate that CNPE and C3G can suppress H2O2-induced hepatotoxicity and cell death through stimulation of endogenous antioxidant enzyme activities and inhibition of apoptosis pathway in HepG2 cells. These findings might support development of CNPE as an alternative natural product for preventing CLD.
Collapse
Affiliation(s)
- Pasitta Panritdum
- Graduate student in Master of Science Program in Nutrition, Faculty of Medicine Ramathibodi Hospital and Institute of NutritionMahidol UniversityBangkokThailand
| | - Chawanphat Muangnoi
- Cell and Animal Model Unit, Institute of NutritionMahidol UniversityNakhon PathomThailand
| | - Siriporn Tuntipopipat
- Cell and Animal Model Unit, Institute of NutritionMahidol UniversityNakhon PathomThailand
| | | | | |
Collapse
|
9
|
Ezhilarasan D, Shree Harini K, Karthick M, Lavanya P. Boldine protects against carbon tetrachloride-induced chronic liver injury by regulating NF-κB signaling pathway. J Biochem Mol Toxicol 2024; 38:e23691. [PMID: 38500399 DOI: 10.1002/jbt.23691] [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: 08/10/2023] [Revised: 02/05/2024] [Accepted: 03/07/2024] [Indexed: 03/20/2024]
Abstract
Sustained liver injuries predominantly promote oxidative stress and inflammation that lead to the progression of chronic liver disease (CLD), including fibrosis, cirrhosis, and hepatocellular carcinoma. Boldine, an alkaloid isolated from Peumus boldus, has been shown to have antioxidant and anti-inflammatory effects. Currently, there is no definitive treatment option available for CLD. Therefore, we investigated the hepatoprotective effect of boldine against carbon tetrachloride (CCl4 )-induced chronic liver injury in rats. CCl4 (2 mL/kg., b.w., i.p.) was administered twice weekly for 5 weeks to induce chronic liver injury in rats. Separate groups of rats were given boldine (20 mg/kg b.w., and 40 mg/kg b.w.) and silymarin (100 mg/kg b.w.) orally, daily. Serum transaminases, lipid peroxidation, and antioxidant levels were measured, and nuclear factor-κB (NF-κB), tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (cox-2), interleukin-1 β (IL-1β), and α-smooth muscle actin (α-SMA) gene and protein expressions were evaluated. CCl4 administration increased liver marker enzymes of hepatotoxicity in serum and oxidative stress markers, inflammatory genes and α-smooth muscle actin expression in liver tissue. Boldine concurrent treatment suppressed CCl4 -induced elevation of transaminase levels in serum, restored enzymic and non-enzymic antioxidants, and downregulated NF-κB, TNF-α, Cox-2 and IL-1β expressions, thereby suppressing hepatic inflammation. Boldine administration also repressed α-SMA expression. The results of this study demonstrate the antioxidant, anti-inflammatory, and antifibrotic properties of boldine, and it can be a potential therapeutic candidate in the treatment of CLD.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Karthik Shree Harini
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Munusamy Karthick
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Prathap Lavanya
- Department of Anatomy, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| |
Collapse
|
10
|
Raghunandhakumar S, Ezhilarasan D, Shree Harini K. Thymoquinone protects thioacetamide-induced chronic liver injury by inhibiting TGF-β1/Smad3 axis in rats. J Biochem Mol Toxicol 2024; 38:e23694. [PMID: 38504479 DOI: 10.1002/jbt.23694] [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: 09/28/2022] [Revised: 01/19/2024] [Accepted: 03/12/2024] [Indexed: 03/21/2024]
Abstract
Chronic liver injury due to various etiological factors results in excess secretion and accumulation of extracellular matrix proteins, leading to scarring of liver tissue and ultimately to hepatic fibrosis. If left untreated, fibrosis might progress to cirrhosis and even hepatocellular carcinoma. Thymoquinone (TQ), an active compound of Nigella sativa, has been reported to exhibit antioxidant, anti-inflammatory and anticancer activities. Therefore, the effect of TQ against thioacetamide (TAA)-induced liver fibrosis was assessed in rats. Fibrosis was induced with intraperitoneal administration of TAA (250 mg/kg b.w.) twice a week for 5 weeks. TQ (20 mg/kg b.w.) and silymarin (50 mg/kg b.w.) were orally administered daily for 5 weeks separately in TAA administered groups. Liver dysfunction was reported by elevated liver enzymes, increased oxidative stress, inflammation and fibrosis upon TAA administration. Our study demonstrated that TQ inhibited the elevation of liver marker enzymes in serum. TQ administration significantly increased antioxidant markers, such as superoxide dismutase, catalase, glutathione, glutathione peroxidase and glutathione reductase in the liver tissue of rats. Further, TQ significantly attenuated liver fibrosis, as illustrated by the downregulation of TAA-induced interleukin-β, tumour necrosis factor-α, inducible nitric oxide synthase and fibrosis markers like transforming growth factor-β (TGF-β), α-smooth muscle actin, collagen-1, Smad3 and 7. Therefore, these findings suggest that TQ has a promising hepatoprotective property, as indicated by its potential to effectively suppress TAA-induced liver fibrosis in rats by inhibiting oxidative stress and inflammation via TGF-β/Smad signaling.
Collapse
Affiliation(s)
- Subramanian Raghunandhakumar
- Department of Pharmacology, Hepatology & Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, 600 077, Tamil Nadu, India
| | - Devaraj Ezhilarasan
- Department of Pharmacology, Hepatology & Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, 600 077, Tamil Nadu, India
| | - Karthik Shree Harini
- Department of Pharmacology, Hepatology & Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, 600 077, Tamil Nadu, India
| |
Collapse
|
11
|
Lashgari NA, Khayatan D, Roudsari NM, Momtaz S, Dehpour AR, Abdolghaffari AH. Therapeutic approaches for cholestatic liver diseases: the role of nitric oxide pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1433-1454. [PMID: 37736835 DOI: 10.1007/s00210-023-02684-2] [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: 04/20/2023] [Accepted: 08/21/2023] [Indexed: 09/23/2023]
Abstract
Cholestasis describes bile secretion or flow impairment, which is clinically manifested with fatigue, pruritus, and jaundice. Neutrophils play a crucial role in many diseases such as cholestasis liver diseases through mediating several oxidative and inflammatory pathways. Data have been collected from clinical, in vitro, and in vivo studies published between 2000 and December 2021 in English and obtained from the PubMed, Google Scholar, Scopus, and Cochrane libraries. Although nitric oxide plays an important role in the pathogenesis of cholestatic liver diseases, excessive levels of NO in serum and affected tissues, mainly synthesized by the inducible nitric oxide synthase (iNOS) enzyme, can exacerbate inflammation. NO induces the inflammatory and oxidative processes, which finally leads to cell damage. We found that natural products such as baicalin, curcumin, resveratrol, and lycopene, as well as chemical likes ursodeoxycholic acid, dexamethasone, rosuvastatin, melatonin, and sildenafil, are able to markedly attenuate the NO production and iNOS expression, mainly through inducing the nuclear factor κB (NF-κB), Janus kinase and signal transducer and activator of transcription (JAK/STAT), and toll like receptor-4 (TLR4) signaling pathways. This study summarizes the latest scientific data about the bile acid signaling pathway, the neutrophil chemotaxis recruitment process during cholestasis, and the role of NO in cholestasis liver diseases. Literature review directed us to propose that suppression of NO and its related pathways could be a therapeutic option for preventing or treating cholestatic liver diseases.
Collapse
Affiliation(s)
- Naser-Aldin Lashgari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., Tehran, Iran, P. O. Box: 19419-33111
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Danial Khayatan
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., Tehran, Iran, P. O. Box: 19419-33111
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nazanin Momeni Roudsari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., Tehran, Iran, P. O. Box: 19419-33111
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., Tehran, Iran, P. O. Box: 19419-33111.
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
12
|
Ouyang H, Miao H, Li Z, Wu D, Gao SC, Dai YY, Gao XD, Chai HS, Hu WY, Zhu JF. Yinhuang granule alleviates carbon tetrachloride-induced liver fibrosis in mice and its mechanism. World J Hepatol 2024; 16:264-278. [PMID: 38495271 PMCID: PMC10941736 DOI: 10.4254/wjh.v16.i2.264] [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] [Received: 10/18/2023] [Revised: 01/09/2024] [Accepted: 02/01/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Liver fibrosis is a formidable global medical challenge, with no effective clinical treatment currently available. Yinhuang granule (YHG) is a proprietary Chinese medicine comprising Scutellariae Radix and Lonicerae Japonicae Flos. It is frequently used for upper respiratory tract infections, pharyngitis, as well as acute and chronic tonsillitis. AIM To investigate the potential of YHG in alleviating carbon tetrachloride (CCl4)-induced liver fibrosis in mice. METHODS To induce a hepatic fibrosis model in mice, this study involved intraperitoneal injections of 2 mL/kg of CCl4 twice a week for 4 wk. Meanwhile, liver fibrosis mice in the low dose of YHG (0.4 g/kg) and high dose of YHG (0.8 g/kg) groups were orally administered YHG once a day for 4 wk. Serum alanine/aspartate aminotransferase (ALT/AST) activity and liver hydroxyproline content were detected. Sirius red and Masson's trichrome staining assay were conducted. Real-time polymerase chain reaction, western-blot and enzyme-linked immunosorbent assay were conducted. Liver glutathione content, superoxide dismutase activity level, reactive oxygen species and protein carbonylation amount were detected. RESULTS The administration of YHG ameliorated hepatocellular injury in CCl4-treated mice, as reflected by decreased serum ALT/AST activity and improved liver histological evaluation. YHG also attenuated liver fibrosis, evident through reduced liver hydroxyproline content, improvements in Sirius red and Masson's trichrome staining, and lowered serum hyaluronic acid levels. Furthermore, YHG hindered the activation of hepatic stellate cells (HSCs) and ameliorated oxidative stress injury and inflammation in liver from CCl4-treated mice. YHG prompted the nuclear accumulation of nuclear factor erythroid 2-related factor 2 (Nrf2) and upregulated the expression of Nrf2-dependent downstream antioxidant genes. In addition, YHG promoted mitochondrial biogenesis in liver from CCl4-treated mice, as demonstrated by increased liver adenosine triphosphate content, mitochondrial DNA levels, and the expression of peroxisome proliferator-activated receptor gamma coactivator 1 alpha and nuclear respiratory factor 1. CONCLUSION YHG effectively attenuates CCl4-induced liver fibrosis in mice by inhibiting the activation of HSCs, reducing inflammation, alleviating liver oxidative stress damage through Nrf2 activation, and promoting liver mitochondrial biogenesis.
Collapse
Affiliation(s)
- Hao Ouyang
- Department of Liver, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hui Miao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 201203, China
| | - Zhen Li
- Department of Liver, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Duan Wu
- Department of Liver, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Si-Cheng Gao
- Department of Liver, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yao-Yao Dai
- Department of Liver, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao-Di Gao
- Department of Liver, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hai-Sheng Chai
- Department of Liver, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wei-Ye Hu
- Department of Liver, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jun-Feng Zhu
- Department of Liver, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
13
|
Mao J, Tan L, Tian C, Wang W, Zhang H, Zhu Z, Li Y. Research progress on rodent models and its mechanisms of liver injury. Life Sci 2024; 337:122343. [PMID: 38104860 DOI: 10.1016/j.lfs.2023.122343] [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: 09/21/2023] [Revised: 11/22/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
The liver is the most important organ for biological transformation in the body and is crucial for maintaining the body's vital activities. Liver injury is a serious pathological condition that is commonly found in many liver diseases. It has a high incidence rate, is difficult to cure, and is prone to recurrence. Liver injury can cause serious harm to the body, ranging from mild to severe fatty liver disease. If the condition continues to worsen, it can lead to liver fibrosis and cirrhosis, ultimately resulting in liver failure or liver cancer, which can seriously endanger human life and health. Therefore, establishing an rodent model that mimics the pathogenesis and severity of clinical liver injury is of great significance for better understanding the pathogenesis of liver injury patients and developing more effective clinical treatment methods. The author of this article summarizes common chemical liver injury models, immune liver injury models, alcoholic liver injury models, drug-induced liver injury models, and systematically elaborates on the modeling methods, mechanisms of action, pathways of action, and advantages or disadvantages of each type of model. The aim of this study is to establish reliable rodent models for researchers to use in exploring anti-liver injury and hepatoprotective drugs. By creating more accurate theoretical frameworks, we hope to provide new insights into the treatment of clinical liver injury diseases.
Collapse
Affiliation(s)
- Jingxin Mao
- Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Lihong Tan
- Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing 400030, China
| | - Cheng Tian
- Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing 400030, China
| | - Wenxiang Wang
- Chongqing Three Gorges Medical College, Chongqing 404120, China
| | - Hao Zhang
- Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing 400030, China
| | - Zhaojing Zhu
- Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing 400030, China
| | - Yan Li
- Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing 400030, China.
| |
Collapse
|
14
|
Zhao Y, Zhao M, Wang Z, Zhao C, Zhang Y, Wang M. Danggui Shaoyao San: Chemical characterization and inhibition of oxidative stress and inflammation to treat CCl 4-induced hepatic fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116870. [PMID: 37423517 DOI: 10.1016/j.jep.2023.116870] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/24/2023] [Accepted: 06/28/2023] [Indexed: 07/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Danggui Shaoyao San (DSS) has effective in treating hepatic ascites and liver disease. AIM OF THE STUDY To explore the chemical characterization of DSS and protective effect on CCl4-induced hepatic fibrosis and its mechanism, especially its anti-oxidative stress and anti-inflammation. MATERIALS AND METHODS The chemical characterization of DSS was determined by HPLC-Q-Exactive Orbitrap MS. And the antioxidant activity of DSS in vitro was determined. The hepatic fibrosis model was established using intragastric administration of 40% CCl4/soybean oil (v/v) twice weekly for 13 weeks. From 6th week, the DSS group and the positive control group were given DSS (2, 4, 8 g/kg/d) and silymarin (50 mg/kg/d), respectively. The livers of rats were examined histologically by H&E. The ALT, AST, ALB and TBIL were determined, and hepatic fibrosis markers (HA, LN, CIV, PIIINP), oxidative stress (SOD, MDA, GST, GSH) and inflammatory factor (IL-6, TNF-α) were tested using ELISA kits. In addition, the levels of TAC, TOS, LOOH and AOPP in the liver were also determined. RESULTS The chemical characterization of DSS was determined by HPLC-Q-Exactive Orbitrap MS. The results show that DSS mainly includes triterpenoids, monoterpenes, phenols, sesquiterpenes, butyl phthalide, etc., and DSS has good antioxidant activity in vitro. In addition, the ALT, AST and TBIL of rats were remarkably reduced after treatment with DSS at three doses. Liver histopathological analysis showed that DSS alleviated the inflammatory infiltration, hepatocyte swelling, necrosis and hepatic fibrosis induced by CCl4. DSS significantly decreased HA, IV-C, PIIINP and LN. Further determination showed that DSS significantly increased TAC, OSI and decreased TOC, LOOH and MDA, indicating that DSS could regulate redox balance and reduce lipid peroxidation in vivo. DSS also increased the activity of GST, SOD and GSH concentration. In addition, DSS also reduced IL-6 and TNF-α. CONCLUSIONS In this study, we described the chemical characterization of DSS and found that it has good antioxidant activity. We proved that DSS has the functions of reducing oxidative stress, anti-inflammatory, protecting liver cells and reducing hepatic fibrosis.
Collapse
Affiliation(s)
- Yanhui Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Min Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Zheyong Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Chunjie Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China.
| | - Yumeng Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China.
| | - Miao Wang
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China.
| |
Collapse
|
15
|
Ezhilarasan D, Shree Harini K, Karthick M, Selvaraj C. Ethyl gallate concurrent administration protects against acetaminophen-induced acute liver injury in mice: An in vivo and in silico approach. Chem Biol Drug Des 2024; 103:e14369. [PMID: 37817304 DOI: 10.1111/cbdd.14369] [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: 07/31/2023] [Revised: 09/13/2023] [Accepted: 09/25/2023] [Indexed: 10/12/2023]
Abstract
Acetaminophen (APAP) in high doses causes acute liver injury and acute liver failure. Ethyl gallate (EG) is a natural polyphenol, possessing antioxidant, anti-inflammatory, and anti-microbial properties. Therefore, in this study, we evaluated the protective role of EG against APAP-induced acute liver injury in mice. Acute liver injury was induced by a single dose of APAP (400 mg/kg., i.p.). In separate groups, EG (10 mg/kg), EG (20 mg/kg), and N-acetylcysteine (NAC; 1200 mg/kg., i.p.) were administered concurrently with APAP. The mice were sacrificed after 24 h of treatment. Liver marker enzymes of hepatotoxicity, antioxidant markers, inflammatory markers, and histopathological studies were done. APAP administration caused a significant elevation of marker enzymes of hepatotoxicity and lipid peroxidation. APAP administration also decreased enzymic and nonenzymic antioxidants. Acute APAP intoxication induced nuclear factor κ B, tumor necrosis factor-α, interleukin-1, p65, and p52 and downregulated IκB gene expressions. Our histopathological studies have confirmed the presence of centrilobular necrosis, 24 h after APAP intoxication. All the above abnormalities were significantly inhibited in groups of mice that were concurrently administered with APAP + EG and APAP + NAC. Our in silico analysis further confirms that hydroxyl groups of EG interact with the above inflammatory proteins at the 3,4,5-trihydroxybenzoic acid region. These effects of EG against APAP-induced acute liver injury could be attributed to its antioxidative, free radical scavenging, and anti-inflammatory potentials. Therefore, this study suggests that EG can be an efficient therapeutic approach to protect the liver from APAP intoxication.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Karthik Shree Harini
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Munusamy Karthick
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Chandrabose Selvaraj
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
- Laboratory for Artificial Intelligence and Molecular Modelling, Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| |
Collapse
|
16
|
Rohit Singh T, Ezhilarasan D, Karthick M, Shree Harini K. Lagerstroemia speciosa Pers. (Lythraceae) Ethanolic Extract Attenuates Isoniazid-Induced Oxidative Stress and Hepatic Inflammation in Rats. Cureus 2024; 16:e51609. [PMID: 38313882 PMCID: PMC10837052 DOI: 10.7759/cureus.51609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/03/2024] [Indexed: 02/06/2024] Open
Abstract
Background Drug-induced liver injury is a common cause of acute liver failure. Isoniazid (INH) is used as a first-line treatment for tuberculosis. Clinical and experimental studies have reported abnormal liver function after INH therapy. Lagerstroemia speciosa Pers., commonly known as banaba, has been traditionally used to treat various ailments including diabetes and obesity due to its antioxidant and anti-inflammatory properties. Aim To investigate the hepatoprotective effect of ethanolic banaba leaf extract (EBLE) against INH-induced hepatotoxicity in rats. Materials and methods A total of 30 male Wistar albino rats (150 - 200 g) were divided into five groups (n = 6). Group I rats were served as a control and were administered dimethyl sulfoxide for the first 30 days and water for the next 30 consecutive days. Group II rats were administered INH (50 mg/kg, p.o.) once in the first 30 consecutive days and sacrificed at Day 30. Group III rats were administered INH for 30 consecutive days and left without treatment for the next 30 days. In Groups IV and V, rats were post-treated orally with EBLE 250 and 500 mg/kg, p.o. (0.3 ml/rat) for 30 days after INH administration. At the end of Day 60, the remaining group of animals were sacrificed. The blood and liver tissues were collected. The marker enzymes of hepatotoxicity, oxidative stress markers, inflammatory markers, and histopathology were analyzed. Results INH administration induced significant elevation of marker enzymes (aspartate transaminase, alanine transaminase, alkaline phosphatase, lactate dehydrogenase, bilirubin, gamma-glutamyl transpeptidase) of hepatotoxicity in the serum. This treatment also increased lipid peroxidation and proinflammatory marker expression (tumor necrosis factor-alpha, transforming growth factor-beta, and nuclear factor kappa B (NF-κB) except inhibitor of NF-κB) and decreased antioxidants such superoxide dismutase, catalase, and glutathione in the liver tissue. All these abnormalities were significantly mitigated after treatment with EBLE. Conclusion The results of this study suggest that EBLE can be used for INH-induced hepatotoxicity.
Collapse
Affiliation(s)
- T Rohit Singh
- Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Devaraj Ezhilarasan
- Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Munusamy Karthick
- Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Karthik Shree Harini
- Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
17
|
Sharma N, Sistla R, Andugulapati SB. Yohimbine ameliorates liver inflammation and fibrosis by regulating oxidative stress and Wnt/β-catenin pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155182. [PMID: 37952411 DOI: 10.1016/j.phymed.2023.155182] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/17/2023] [Accepted: 11/01/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND AND PURPOSE Chronic liver injury, caused by various aetiologies, causes recurrent tissue damage, culminating in decreased liver regenerative ability and resulting in fibrosis followed by cirrhosis. In this study, the anti-fibrotic activity of Yohimbine hydrochloride (YHC) was investigated using various in vitro models and in vivo models. METHODS To assess the anti-inflammatory, antioxidant, and anti-fibrotic effects of YHC, lipopolysaccharide or TGF-β induced differentiation or lipid-induced oxidative-stress models were employed using HLECs, HSC-LX2, and HepG2 cells. Further, thioacetamide (TAA) induced hepatic inflammation/fibrosis models were utilized to validate the YHC's anti-fibrotic activity in rats. RESULTS Inflammation/differentiation experiments in HLECs and HSC-LX2 revealed that YHC treatment significantly (p < 0.001) mitigated the lipopolysaccharide or TGF-β induced upregulation of inflammatory and fibrotic markers expression respectively. In addition, YHC dose-dependently reduced the TGF-β induced migration and palmitic acid-induced oxidative stress in HepG2 cells. Further, TAA administration (5 weeks) in vivo rat model showed increased inflammatory marker levels/expression, oxidative stress, and pathological abnormalities. Additionally, TAA administration (9 weeks) elevated the fibrotic marker expression, collagen deposition in liver tissues, and shortened longevity in rats. Treatment with YHC dose-dependently mitigated the TAA-induced abnormalities in both inflammation and fibrosis models and improved the survival of the rats. Further mechanistic approaches revealed that TAA administration elevated the JNK, Wnt components and β-catenin expression in hepatic stellate cells and animal tissues. Further treatment with YHC significantly modulated the JNK/Wnt/β-catenin signaling. Moreover, the β-catenin nuclear translocation results showed that β-catenin levels were significantly elevated in the nuclear fraction of TAA control samples and reduced in YHC-treated samples. CONCLUSION Yohimbine treatment significantly improved inflammation and fibrosis by inhibiting differentiation, oxidative stress, and collagen deposition by partly modulating the JNK/Wnt/β-catenin pathway. These results might serve as a foundation for proposing yohimbine as a potential lead compound for liver fibrosis.
Collapse
Affiliation(s)
- Nidhi Sharma
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad-500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh-201 002, India
| | - Ramakrishna Sistla
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad-500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh-201 002, India
| | - Sai Balaji Andugulapati
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad-500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh-201 002, India.
| |
Collapse
|
18
|
Aldaba-Muruato LR, Sánchez-Barbosa S, Rodríguez-Purata VH, Cabrera-Cruz G, Rosales-Domínguez E, Martínez-Valentín D, Alarcón-López YA, Aguirre-Vidal P, Hernández-Serda MA, Cárdenas-Granados LA, Vázquez-Valadez VH, Angeles E, Macías-Pérez JR. In Vivo and In Silico Studies of the Hepatoprotective Activity of Tert-Butylhydroquinone. Int J Mol Sci 2023; 25:475. [PMID: 38203648 PMCID: PMC10779046 DOI: 10.3390/ijms25010475] [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: 11/16/2023] [Revised: 12/14/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Tert-butylhydroquinone (TBHQ) is a synthetic food antioxidant with biological activities, but little is known about its pharmacological benefits in liver disease. Therefore, this work aimed to evaluate TBHQ during acute liver damage induced by CCl4 (24 h) or BDL (48 h) in Wistar rats. It was found that pretreatment with TBHQ prevents 50% of mortality induced by a lethal dose of CCl4 (4 g/kg, i.p.), and 80% of BDL+TBHQ rats survived, while only 50% of the BDL group survived. Serum markers of liver damage and macroscopic and microscopic (H&E staining) observations suggest that TBHQ protects from both hepatocellular necrosis caused by the sublethal dose of CCl4 (1.6 g/kg, i.p.), as well as necrosis/ductal proliferation caused by BDL. Additionally, online databases identified 49 potential protein targets for TBHQ. Finally, a biological target candidate (Keap1) was evaluated in a proof-of-concept in silico molecular docking assay, resulting in an interaction energy of -5.5491 kcal/mol, which was higher than RA839 and lower than monoethyl fumarate (compounds known to bind to Keap1). These findings suggest that TBHQ increases the survival of animals subjected to CCl4 intoxication or BDL, presumably by reducing hepatocellular damage, probably due to the interaction of TBHQ with Keap1.
Collapse
Affiliation(s)
- Liseth Rubi Aldaba-Muruato
- Biomedical Science Laboratory, Clinical Chemistry, Faculty of Professional Studies Huasteca Zone, Autonomous University of San Luis Potosi, Ciudad Valles 79060, San Luis Potosi, Mexico; (L.R.A.-M.); (S.S.-B.); (G.C.-C.); (E.R.-D.); (D.M.-V.)
| | - Sandra Sánchez-Barbosa
- Biomedical Science Laboratory, Clinical Chemistry, Faculty of Professional Studies Huasteca Zone, Autonomous University of San Luis Potosi, Ciudad Valles 79060, San Luis Potosi, Mexico; (L.R.A.-M.); (S.S.-B.); (G.C.-C.); (E.R.-D.); (D.M.-V.)
| | - Víctor Hugo Rodríguez-Purata
- Pharmacobiological Sciences, Faculty of Chemical Sciences, Autonomous University of San Luis Potosi, San Luis Potosi 78210, Mexico;
| | - Georgina Cabrera-Cruz
- Biomedical Science Laboratory, Clinical Chemistry, Faculty of Professional Studies Huasteca Zone, Autonomous University of San Luis Potosi, Ciudad Valles 79060, San Luis Potosi, Mexico; (L.R.A.-M.); (S.S.-B.); (G.C.-C.); (E.R.-D.); (D.M.-V.)
| | - Estefany Rosales-Domínguez
- Biomedical Science Laboratory, Clinical Chemistry, Faculty of Professional Studies Huasteca Zone, Autonomous University of San Luis Potosi, Ciudad Valles 79060, San Luis Potosi, Mexico; (L.R.A.-M.); (S.S.-B.); (G.C.-C.); (E.R.-D.); (D.M.-V.)
| | - Daniela Martínez-Valentín
- Biomedical Science Laboratory, Clinical Chemistry, Faculty of Professional Studies Huasteca Zone, Autonomous University of San Luis Potosi, Ciudad Valles 79060, San Luis Potosi, Mexico; (L.R.A.-M.); (S.S.-B.); (G.C.-C.); (E.R.-D.); (D.M.-V.)
| | - Yoshio Aldo Alarcón-López
- Laboratorio de Química Teórica y Medicinal, FESC, Universidad Nacional Autónoma de México, Avenida 1 de Mayo S/N, Santa María las Torre, Cuautitlán Izcalli 54750, Estado de México, Mexico; (Y.A.A.-L.); (P.A.-V.); (M.A.H.-S.); (L.A.C.-G.); (V.H.V.-V.); (E.A.)
| | - Pablo Aguirre-Vidal
- Laboratorio de Química Teórica y Medicinal, FESC, Universidad Nacional Autónoma de México, Avenida 1 de Mayo S/N, Santa María las Torre, Cuautitlán Izcalli 54750, Estado de México, Mexico; (Y.A.A.-L.); (P.A.-V.); (M.A.H.-S.); (L.A.C.-G.); (V.H.V.-V.); (E.A.)
| | - Manuel Alejandro Hernández-Serda
- Laboratorio de Química Teórica y Medicinal, FESC, Universidad Nacional Autónoma de México, Avenida 1 de Mayo S/N, Santa María las Torre, Cuautitlán Izcalli 54750, Estado de México, Mexico; (Y.A.A.-L.); (P.A.-V.); (M.A.H.-S.); (L.A.C.-G.); (V.H.V.-V.); (E.A.)
| | - Luis Alfonso Cárdenas-Granados
- Laboratorio de Química Teórica y Medicinal, FESC, Universidad Nacional Autónoma de México, Avenida 1 de Mayo S/N, Santa María las Torre, Cuautitlán Izcalli 54750, Estado de México, Mexico; (Y.A.A.-L.); (P.A.-V.); (M.A.H.-S.); (L.A.C.-G.); (V.H.V.-V.); (E.A.)
| | - Víctor Hugo Vázquez-Valadez
- Laboratorio de Química Teórica y Medicinal, FESC, Universidad Nacional Autónoma de México, Avenida 1 de Mayo S/N, Santa María las Torre, Cuautitlán Izcalli 54750, Estado de México, Mexico; (Y.A.A.-L.); (P.A.-V.); (M.A.H.-S.); (L.A.C.-G.); (V.H.V.-V.); (E.A.)
| | - Enrique Angeles
- Laboratorio de Química Teórica y Medicinal, FESC, Universidad Nacional Autónoma de México, Avenida 1 de Mayo S/N, Santa María las Torre, Cuautitlán Izcalli 54750, Estado de México, Mexico; (Y.A.A.-L.); (P.A.-V.); (M.A.H.-S.); (L.A.C.-G.); (V.H.V.-V.); (E.A.)
| | - José Roberto Macías-Pérez
- Biomedical Science Laboratory, Clinical Chemistry, Faculty of Professional Studies Huasteca Zone, Autonomous University of San Luis Potosi, Ciudad Valles 79060, San Luis Potosi, Mexico; (L.R.A.-M.); (S.S.-B.); (G.C.-C.); (E.R.-D.); (D.M.-V.)
| |
Collapse
|
19
|
Fu K, Dai S, Li Y, Ma C, Xue X, Zhang S, Wang C, Zhou H, Zhang Y, Li Y. The protective effect of forsythiaside A on 3,5-diethoxycarbonyl-1,4-dihydrocollidine-induced cholestatic liver injury in mice: Based on targeted metabolomics and molecular biology technology. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166822. [PMID: 37523877 DOI: 10.1016/j.bbadis.2023.166822] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023]
Abstract
Cholestasis is a disorder of bile secretion and excretion caused by a variety of etiologies. At present, there is a lack of functional foods or drugs that can be used for intervention. Forsythiaside A (FTA) is a natural phytochemical component isolated from the medicinal plant Forsythia suspensa (Thunb.) Vahl, which has a significant hepatoprotective effect. In this study, we investigated whether FTA could alleviate liver injury induced by cholestasis. In vitro, FTA reversed the decrease in viability of human intrahepatic bile duct epithelial cells, the decrease in antioxidant enzymes (SOD1, CAT and GSH-Px), and cell apoptosis induced by lithocholic acid. In vivo, FTA protected mice from 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-induced liver injury, abnormal serum biochemical indexes, abnormal bile duct hyperplasia, and inflammatory infiltration. Furthermore, FTA treatment alleviated liver fibrosis by inhibiting collagen deposition and HSC activation. The metabonomic results showed that DDC-induced bile acid disorders in the liver and serum were reversed after FTA treatment, which may benefit from the activation of the FXR/BSEP axis. In addition, FTA treatment increased the levels of antioxidant enzymes in the serum and liver. Meanwhile, FTA treatment inhibited ROS and MDA levels and cleaved caspase 3 protein expression, thereby reducing DDC-induced hepatic oxidative stress and apoptosis. Further studies showed that the antioxidant effects of FTA were dependent on the activation of the BRG1/NRF2/HO-1 axis. In a word, FTA has a significant hepatoprotective effect on cholestatic liver injury, and can be further developed as a functional food or drug to prevent and treat cholestatic liver injury.
Collapse
Affiliation(s)
- Ke Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shu Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yanzhi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xinyan Xue
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shenglin Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Honglin Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yafang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
20
|
Jung JI, Choi YJ, Kim J, Baek KS, Kim EJ. Aqueous extract of Laurus nobilis leaf accelerates the alcohol metabolism and prevents liver damage in single-ethanol binge rats. Nutr Res Pract 2023; 17:1113-1127. [PMID: 38053830 PMCID: PMC10694424 DOI: 10.4162/nrp.2023.17.6.1113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/02/2023] [Accepted: 09/08/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND/OBJECTIVES Excessive alcohol consumption has harmful health effects, including alcohol hangovers and alcohol-related liver disease. Therefore, methods to accelerate the alcohol metabolism are needed. Laurus nobilis is a spice, flavoring agent, and traditional herbal medicine against various diseases. This study examined whether the standardized aqueous extract of L. nobilis leaves (LN) accelerates the alcohol metabolism and protects against liver damage in single-ethanol binge Sprague-Dawley (SD) rats. MATERIALS/METHODS LN was administered orally to SD rats 1 h before ethanol administration (3 g/kg body weight [BW]) at 100 and 300 mg/kg BW. Blood samples were collected 0.5, 1, 2, and 4 h after ethanol administration. The livers were excised 1 h after ethanol administration to determine the hepatic enzyme activity. The alcohol dehydrogenase (ADH), aldehyde dehydrogenase (ALDH), superoxide dismutase (SOD), and glutathione peroxidase (GPx) activities in the liver tissue were measured. RESULTS LN decreased the serum ethanol and acetaldehyde levels in ethanol-administered rats. LN increased the hepatic ADH and ALDH activities but decreased the alanine aminotransferase, aspartate aminotransferase, and gamma-glutamyl transferase activities in the ethanol-administered rats. In addition, LN inhibited lipid peroxidation and increased the activities of SOD and GPx. CONCLUSIONS LN modulates the mediators of various etiological effects of excessive alcohol consumption and enhances the alcohol metabolism and antioxidant activity, making it a potential candidate for hangover treatments.
Collapse
Affiliation(s)
- Jae In Jung
- Industry coupled Cooperation Center for Bio Healthcare Materials, Hallym University, Chuncheon 24252, Korea
| | - Yean-Jung Choi
- Department of Food and Nutrition, Sahmyook University, Seoul 01795, Korea
| | - Jinhak Kim
- R&D Division, Daehan Chemtech Co. Ltd., Gwacheon 13840, Korea
| | - Kwang-Soo Baek
- R&D Division, Daehan Chemtech Co. Ltd., Gwacheon 13840, Korea
| | - Eun Ji Kim
- Industry coupled Cooperation Center for Bio Healthcare Materials, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
21
|
Gayatri Devi R, Ezhilarasan D. Concurrent administration of farnesol protects acetaminophen-induced acute hepatic necrosis in mice. J Biochem Mol Toxicol 2023; 37:e23478. [PMID: 37458150 DOI: 10.1002/jbt.23478] [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: 10/25/2022] [Revised: 06/09/2023] [Accepted: 07/08/2023] [Indexed: 07/18/2023]
Abstract
Acetaminophen (APAP) is known to cause acute liver injury and acute liver failure in Western countries. This study investigates the protective role of farnesol (FAR) (C15 H26 O), a natural sesquiterpene alcohol in essential oils, against APAP-induced acute liver necrosis in mice. Mice were injected with a single dose of APAP (300 mg/kg) via an intraperitoneal route. Different groups of mice were concurrently treated with a single dose of FAR 25 mg/kg, FAR 50 mg/kg, and N-acetylcysteine. APAP administration caused a significant increase in transaminase activities and malondialdehyde (MDA) levels in the serum and liver tissue, respectively, with a concomitant decrease in intracellular antioxidants, including reduced glutathione (GSH) in the liver tissue. APAP intoxication upregulated proinflammatory cytokines such as tumor necrosis factor-α, interleukin-1β (IL-1β), IL-6, nuclear factor-κB (NF-κB), and IκB kinase β in the liver tissue. FAR and N-acetylcysteine (NAC) administrations concurrently with APAP prevented serum transaminase increase in serum and MDA levels in the liver tissue. A high dose of FAR and NAC treatments significantly inhibited GSH and other antioxidant depletion. FAR and NAC treatments also downregulated the expression of proinflammatory markers. FAR treatments protects against APAP-induced acute liver injury and offers antioxidant and anti-inflammatory effects by inhibiting the NF-κB pathway involved in the transcription of genes responsible for inflammatory cytokine synthesis.
Collapse
Affiliation(s)
- Ramalingam Gayatri Devi
- Department of Physiology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Devaraj Ezhilarasan
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| |
Collapse
|
22
|
Liu S, Zhao Y, Feng X, Xu H. SARS-CoV-2 infection threatening intestinal health: A review of potential mechanisms and treatment strategies. Crit Rev Food Sci Nutr 2023; 63:12578-12596. [PMID: 35894645 DOI: 10.1080/10408398.2022.2103090] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The outbreak of the COVID-19 pandemic has brought great problems to mankind, including economic recession and poor health. COVID-19 patients are frequently reported with gastrointestinal symptoms such as diarrhea and vomiting in clinical diagnosis. Maintaining intestinal health is the key guarantee to maintain the normal function of multiple organs, otherwise it will be a disaster. Therefore, the purpose of this review was deeply understanded the potential mechanism of SARS-CoV-2 infection threatening intestinal health and put forward reasonable treatment strategies. Combined with the existing researches, we summarized the mechanism of SARS-CoV-2 infection threatening intestinal health, including intestinal microbiome disruption, intestinal barrier dysfunction, intestinal oxidative stress and intestinal cytokine storm. These adverse intestinal events may affect other organs through the circulatory system or aggravate the course of the disease. Typically, intestinal disadvantage may promote the progression of SARS-CoV-2 through the gut-lung axis and increase the disease degree of COVID-19 patients. In view of the lack of specific drugs to inhibit SARS-CoV-2 replication, the current review described new strategies of probiotics, prebiotics, postbiotics and nutrients to combat SARS-CoV-2 infection and maintain intestinal health. To provide new insights for the prevention and treatment of gastrointestinal symptoms and pneumonia in patients with COVID-19.
Collapse
Affiliation(s)
- Shanji Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Yu Zhao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Xiaoyan Feng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
23
|
Ben Ammar R, Zahra HA, Abu Zahra AM, Alfwuaires M, Abdulaziz Alamer S, Metwally AM, Althnaian TA, Al-Ramadan SY. Protective Effect of Fucoxanthin on Zearalenone-Induced Hepatic Damage through Nrf2 Mediated by PI3K/AKT Signaling. Mar Drugs 2023; 21:391. [PMID: 37504922 PMCID: PMC10381773 DOI: 10.3390/md21070391] [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: 05/31/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023] Open
Abstract
Hepatotoxic contaminants such as zearalenone (ZEA) are widely present in foods. Marine algae have a wide range of potential applications in pharmaceuticals, cosmetics, and food products. Research is ongoing to develop treatments and products based on the compounds found in algae. Fucoxanthin (FXN) is a brown-algae-derived dietary compound that is reported to prevent hepatotoxicity caused by ZEA. This compound has multiple biological functions, including anti-diabetic, anti-obesity, anti-microbial, and anti-cancer properties. Furthermore, FXN is a powerful antioxidant. In this study, we examined the effects of FXN on ZEA-induced stress and inflammation in HepG2 cells. MTT assays, ROS generation assays, Western blots, and apoptosis analysis were used to evaluate the effects of FXN on ZEA-induced HepG2 cell inflammation. Pre-incubation with FXN reduced the cytotoxicity of ZEA toward HepG2 cells. FXN inhibited the ZEA-induced production of pro-inflammatory cytokines, including IL-1 β, IL-6, and TNF-α. Moreover, FXN increased HO-1 expression in HepG2 by activating the PI3K/AKT/NRF2 signaling pathway. In conclusion, FXN inhibits ZEA-induced inflammation and oxidative stress in hepatocytes by targeting Nrf2 via activating PI3K/AKT signaling.
Collapse
Affiliation(s)
- Rebai Ben Ammar
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Center of Biotechnology of Borj-Cedria, Laboratory of Aromatic and Medicinal Plants, Technopole of Borj-Cedria, Hammam-Lif 2050, Tunisia
| | - Hamad Abu Zahra
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | | | - Manal Alfwuaires
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Sarah Abdulaziz Alamer
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Ashraf M Metwally
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Botany and Microbiology Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - Thnaian A Althnaian
- Department of Anatomy, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Saeed Y Al-Ramadan
- Department of Anatomy, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| |
Collapse
|
24
|
Afolabi OB, Olasehinde OR, Olanipon DG, Mabayoje SO, Familua OM, Jaiyesimi KF, Agboola EK, Idowu TO, Obafemi OT, Olaoye OA, Oloyede OI. Antioxidant evaluation and computational prediction of prospective drug-like compounds from polyphenolic-rich extract of Hibiscus cannabinus L. seed as antidiabetic and neuroprotective targets: assessment through in vitro and in silico studies. BMC Complement Med Ther 2023; 23:203. [PMID: 37337198 DOI: 10.1186/s12906-023-04023-7] [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: 01/12/2023] [Accepted: 06/03/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Reports have implicated diabetes mellitus (DM) and Alzheimer's disease (AD) as some of the global persistent health challenges with no lasting solutions, despite of significant inputs of modern-day pharmaceutical firms. This study therefore, aimed to appraise the in vitro antioxidant potential, enzymes inhibitory activities, and as well carry out in silico study on bioactive compounds from polyphenolic-rich extract of Hibiscus cannabinus seed (PEHc). METHODS In vitro antioxidant assays were performed on PEHc using standard methods while the identification of phytoconstituents was carried out with high performance liquid chromatography (HPLC). For the in silico molecular docking using Schrodinger's Grid-based ligand docking with energetics software, seven target proteins were retrieved from the database ( https://www.rcsb.org/ ). RESULTS HPLC technique identified twelve chemical compounds in PEHc, while antioxidant quantification revealed higher total phenolic contents (243.5 ± 0.71 mg GAE/g) than total flavonoid contents (54.06 ± 0.09 mg QE/g) with a significant (p < 0.05) inhibition of ABTS (IC50 = 218.30 ± 0.87 µg/ml) and 1, 1-diphenyl-2-picrylhydrazyl free radicals (IC50 = 227.79 ± 0.74 µg/ml). In a similar manner, the extract demonstrated a significant (p < 0.05) inhibitory activity against α-amylase (IC50 = 256.88 ± 6.15 µg/ml) and α-glucosidase (IC50 = 183.19 ± 0.23 µg/ml) as well as acetylcholinesterase (IC50 = 262.95 ± 1.47 µg/ml) and butyrylcholinesterase (IC50 = 189.97 ± 0.82 µg/ml), respectively. Furthermore, In silico study showed that hibiscetin (a lead) revealed a very strong binding affinity energies for DPP-4, (PDB ID: 1RWQ) and α-amylase (PDB ID: 1SMD), gamma-tocopherol ( for peptide-1 receptor; PDB ID: 3C59, AChE; PDB ID: 4EY7 and BChE; PDB ID: 7B04), cianidanol for α-glucosidase; PDB ID: 7KBJ and kaempferol for Poly [ADP-ribose] polymerase 1 (PARP-1); PDB ID: 6BHV, respectively. More so, ADMET scores revealed drug-like potentials of the lead compounds identified in PEHc. CONCLUSION As a result, the findings of this study point to potential drug-able compounds in PEHc that could be useful for the management of DM and AD.
Collapse
Affiliation(s)
- Olakunle Bamikole Afolabi
- Phytomedicine and Toxicology Unit, Biochemistry Programme, Department of Chemical Sciences, College of Sciences, Afe-Babalola University, P.M.B 5454, Ado-Ekiti, Ekiti State, Nigeria.
| | - Oluwaseun Ruth Olasehinde
- Department of Medical Biochemistry, College of Medicine and Health Sciences, Afe Babalola University, P.M.B 5454, Ado-Ekiti, Ekiti State, Nigeria
| | - Damilola Grace Olanipon
- Department of Biological Sciences, College of Sciences, Afe Babalola University, P.M.B. 5454, Ado-Ekiti, Ekiti State, Nigeria
| | - Samson Olatunde Mabayoje
- Department of Biological Sciences, College of Sciences, Afe Babalola University, P.M.B. 5454, Ado-Ekiti, Ekiti State, Nigeria
| | - Olufemi Michael Familua
- Department of Pharmacology and Toxicology, College of Pharmacy, Afe Babalola University, P.M.B. 5454, Ado-Ekiti, Ekiti State, Nigeria
| | - Kikelomo Folake Jaiyesimi
- Phytomedicine and Toxicology Unit, Biochemistry Programme, Department of Chemical Sciences, College of Sciences, Afe-Babalola University, P.M.B 5454, Ado-Ekiti, Ekiti State, Nigeria
| | - Esther Kemi Agboola
- Phytomedicine and Toxicology Unit, Biochemistry Programme, Department of Chemical Sciences, College of Sciences, Afe-Babalola University, P.M.B 5454, Ado-Ekiti, Ekiti State, Nigeria
| | - Tolulope Olajumoke Idowu
- Medicinal Plant Unit, Chemistry Programme, Department of Chemical Sciences, College of Sciences, Afe-Babalola University, P.M.B 5454, Ado- Ekiti, Ekiti State, Nigeria
| | - Olabisi Tajudeen Obafemi
- Phytomedicine and Toxicology Unit, Biochemistry Programme, Department of Chemical Sciences, College of Sciences, Afe-Babalola University, P.M.B 5454, Ado-Ekiti, Ekiti State, Nigeria
| | - Oyindamola Adeniyi Olaoye
- Phytomedicine and Toxicology Unit, Biochemistry Programme, Department of Chemical Sciences, College of Sciences, Afe-Babalola University, P.M.B 5454, Ado-Ekiti, Ekiti State, Nigeria
| | - Omotade Ibidun Oloyede
- Department of Biochemistry, Ekiti State University, P.M.B 5363, Ado-Ekiti, Ekiti State, Nigeria
| |
Collapse
|
25
|
Kalairajan S, K K K, P G. Red Cell Distribution Width in Chronic Liver Disease: An Observational Study. Cureus 2023; 15:e40158. [PMID: 37431329 PMCID: PMC10329736 DOI: 10.7759/cureus.40158] [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: 04/30/2023] [Accepted: 06/08/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND Chronic liver diseases (CLDs) encompass a group of conditions that are marked by diminished liver function due to ongoing inflammation or damage. This study aimed to establish a relationship between the red cell distribution width (RDW) and two scoring systems, namely the Model for End-Stage Liver Disease (MELD) score and Child-Turcotte-Pugh (CTP) score, in individuals diagnosed with CLDs. METHODS The study was carried out at Aarupadai Veedu Medical College & Hospital, Pondicherry, India, following approval from the Institutional Ethical Committee in the Department of General Medicine and Gastroenterology. It involved 50 patients aged 18 years and above who were diagnosed with CLD. The RDW of all selected patients was measured using a three-part autoanalyzer, and its correlation with the MELD and CTP scores was examined. Data analysis was performed using IBM SPSS (Statistical Package for Social Sciences), version 21.0 (IBM Corp., Armonk, NY), with a significance level set at p < 0.05. RESULTS When comparing the baseline characteristics including age, gender, and encephalopathy, no statistically significant differences were found between RDW-standard deviation (RDW-SD) and RDW-corpuscular value (RDW-CV) (p > 0.05). However, a statistically significant correlation was observed between the presence of ascites and RDW-CV values (p = 0.029). Furthermore, there was a significant association between the CTP score and RDW-SD (p < 0.0001). The association between the MELD score and RDW-SD was also found to be statistically significant (p = 0.006). Similarly, statistically significant results were obtained between the MELD score and RDW-CV (p = 0.034). CONCLUSION The utilization of RDW holds promise as a convenient and effective tool for evaluating the severity of individuals with CLD.
Collapse
Affiliation(s)
- Sankar Kalairajan
- Department of Internal Medicine, Aarupadai Veedu Medical College and Hospital, Vinayaka Mission's Research Foundation (DU), Pondicherry, IND
| | - Kavitha K K
- Department of Microbiology, Swamy Vivekanandha Medical College Hospital and Research Institute, Tiruchengode, IND
| | - Govindaraj P
- Department of Internal Medicine, Aarupadai Veedu Medical College and Hospital, Vinayaka Mission's Research Foundation (DU), Pondicherry, IND
| |
Collapse
|
26
|
Ezhilarasan D. Molecular mechanisms in thioacetamide-induced acute and chronic liver injury models. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 99:104093. [PMID: 36870405 DOI: 10.1016/j.etap.2023.104093] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
Thioacetamide (TAA) undergoes bioactivation in the liver by the CYP450 2E1 enzyme, resulting in the formation of TAA-S-oxide and TAA-S-dioxide. TAA-S-dioxide induces oxidative stress via lipid peroxidation of the hepatocellular membrane. A single TAA dose (50-300 mg/kg) administration initiates hepatocellular necrosis around the pericentral region after its covalent binding to macromolecules in the liver. Intermittent TAA administration (150-300 mg/kg, weekly thrice, for 11-16 weeks) activates transforming growth factor (TGF)-β/smad3 downstream signaling in injured hepatocytes, causing hepatic stellate cells (HSCs) to acquire myofibroblast like phenotype. The activated HSCs synthesize a variety of extracellular matrix, leading to liver fibrosis, cirrhosis, and portal hypertension. The TAA induced liver injury varies depending on the animal model, dosage, frequency, and routes of administration. However, TAA induces hepatotoxicity in a reproducible manner, and it is an ideal model to evaluate the antioxidant, cytoprotective, and antifibrotic compounds in experimental animals.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Molecular Medicine and Toxicology Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600 077, India.
| |
Collapse
|
27
|
Wang W, Zhao X, Ma Y, Zhang J, Xu C, Ma J, Hussain MA, Hou J, Qian S. Alleviating Effect of Lacticaseibacillus rhamnosus 1.0320 Combined with Dihydromyricetin on Acute Alcohol Exposure-Induced Hepatic Impairment: Based on Short-Chain Fatty Acids and Adenosine 5'-Monophosphate-Activated Protein Kinase-Mediated Lipid Metabolism Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:4837-4850. [PMID: 36930948 DOI: 10.1021/acs.jafc.2c08523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Excessive drinking has been listed by the World Health Organization as the fifth major risk factor; especially the liver, as the core organ of alcohol metabolism, is prone to organic lesions. Probiotics have received attention due to their bioactivity for liver protection. The beneficial effects of probiotics on hosts are related to their physiological functions. Therefore, based on the concept of second-generation synbiotes, this study explored the protective effects of four dietary polyphenols on the stress tolerance, hydrophobicity, adhesion, and digestive characteristics of L. rhamnosus 1.0320. L. rhamnosus 1.0320 had the best synergistic effect with dihydromyricetin (DMY). Therefore, this combination was selected as a synbiotic supplement to explore the protective effect on acute alcohol exposure-induced hepatic impairment. The results showed that L. rhamnosus 1.0320 combined with DMY restored the intestinal barrier by upregulating short-chain fatty acid levels and activated the adenosine 5'-monophosphate-activated protein kinase-mediated lipid metabolism pathway to inhibit oxidative stress, inflammation, and lipid accumulation in the liver. Furthermore, 109 CFU/mouse/d L. rhamnosus 1.0320 and 50 mg/kg/d DMY by gavage were identified as the optimal doses for protection against acute alcohol expose-induced hepatic impairment. This study provides new insights into alleviating acute alcoholic hepatic impairment by targeting intestinal metabolites through the gut-liver axis.
Collapse
Affiliation(s)
- Wan Wang
- Key Laboratory of Dairy Science, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xu Zhao
- Key Laboratory of Dairy Science, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yue Ma
- Key Laboratory of Dairy Science, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Jing Zhang
- Key Laboratory of Dairy Science, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Cong Xu
- Key Laboratory of Dairy Science, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Jiage Ma
- Key Laboratory of Dairy Science, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Muhammad Altaf Hussain
- Lasbela University of Agriculture, Water and Marine Science, Uthal 90150, Balochistan, Pakistan
| | - Juncai Hou
- Key Laboratory of Dairy Science, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Shanshan Qian
- Analysis and Testing Sharing Center of the State-Owned Asset Management Office, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
28
|
Pandey B, Baral R, Kaundinnyayana A, Panta S. Promising hepatoprotective agents from the natural sources: a study of scientific evidence. EGYPTIAN LIVER JOURNAL 2023. [DOI: 10.1186/s43066-023-00248-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
Abstract
Background
Natural bioactive components derived from plant secondary metabolites have been pronounced as valuable alternatives for anticipating and subsiding hepatotoxic effects and its chronic complications based on experimental verification. The focus of this review is to elucidate the commonly used modern medicine for the treatment of liver disease and how major phytoconstituents have been tested for hepatoprotective activity, mechanism of action of some promising agents from natural sources, and clinical trial data for treating in patients with different liver diseases by the aid of natural phytoconstituents.
Main text
The review shows fifteen major isolated phytoconstituents, their biological sources, chemical structures, utilized plant parts, type of extracts used, hepatoprotective assay method, and their possible mechanism of action on the hepatoprotection. Nine promising hepatoprotective leads from natural sources with their chemistry and hepatoprotective mechanism are mentioned briefly. The review further includes the recent clinical trial studies of some hepatoprotective leads and their clinical outcome with different liver disease patients. Scientific studies revealed that antioxidant properties are the central mechanism for the phytoconstituents to subside different disease pathways by upsurging antioxidant defense system of cells, scavenging free radicals, down surging lipid peroxidation, improving anti-inflammatory potential, and further protecting the hepatic cell injury. In this review, we summarize recent development of natural product-based hepatoprotective leads and their curative potential for various sort of liver diseases. Furthermore, the usefulness of hit and lead molecules from natural sources for significant clinical benefit to discover new drug molecule and downsizing the problems of medication and chemical-induced hepatotoxic effects is extrapolated.
Conclusion
Further research are encouraged to elucidate the pharmacological principle of these natural-based chemical agents which will stimulate future pharmaceutical development of therapeutically beneficial hepatoprotective regimens.
Collapse
|
29
|
Chen L, Xia S, Wang F, Zhou Y, Wang S, Yang T, Li Y, Xu M, Zhou Y, Kong D, Zhang Z, Shao J, Xu X, Zhang F, Zheng S. m 6A methylation-induced NR1D1 ablation disrupts the HSC circadian clock and promotes hepatic fibrosis. Pharmacol Res 2023; 189:106704. [PMID: 36813093 DOI: 10.1016/j.phrs.2023.106704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/04/2023] [Accepted: 02/19/2023] [Indexed: 02/22/2023]
Abstract
The roles of nuclear receptor subfamily 1 group d member 1 (NR1D1) and the circadian clock in liver fibrosis remain unclear. Here, we showed that liver clock genes, especially NR1D1, were dysregulated in mice with carbon tetrachloride (CCl4)-induced liver fibrosis. In turn, disruption of the circadian clock exacerbated experimental liver fibrosis. NR1D1-deficient mice were more sensitive to CCl4-induced liver fibrosis, supporting a critical role of NR1D1 in liver fibrosis development. Validation at the tissue and cellular levels showed that NR1D1 was primarily degraded by N6-methyladenosine (m6A) methylation in a CCl4-induced liver fibrosis model, and this result was also validated in rhythm-disordered mouse models. In addition, the degradation of NR1D1 further inhibited the phosphorylation of dynein-related protein 1-serine site 616 (DRP1S616), resulting in weakened mitochondrial fission function and increased mitochondrial DNA (mtDNA) release in hepatic stellate cell (HSC), which in turn activated the cGMP-AMP synthase (cGAS) pathway. Activation of the cGAS pathway induced a local inflammatory microenvironment that further stimulated liver fibrosis progression. Interestingly, in the NR1D1 overexpression model, we observed that DRP1S616 phosphorylation was restored, and cGAS pathway was also inhibited in HSCs, resulting in improved liver fibrosis. Taken together, our results suggest that targeting NR1D1 may be an effective approach to liver fibrosis prevention and management.
Collapse
Affiliation(s)
- Li Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Siwei Xia
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Feixia Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Yuanyuan Zhou
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Shuqi Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Ting Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Yang Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Min Xu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Ya Zhou
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Desong Kong
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, 157 Daming Road, Nanjing 210023, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Xuefen Xu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China.
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China.
| |
Collapse
|
30
|
Ezhilarasan D. Unraveling the pathophysiologic role of galectin-3 in chronically injured liver. J Cell Physiol 2023; 238:673-686. [PMID: 36745560 DOI: 10.1002/jcp.30956] [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: 03/22/2022] [Revised: 01/04/2023] [Accepted: 01/12/2023] [Indexed: 02/07/2023]
Abstract
Galectin-3 (Gal-3) previously referred to as S-type lectins, is a soluble protein that specifically binds to β-galactoside carbohydrates with high specificity. Gal-3 plays a pivotal role in a variety of pathophysiological processes such as cell proliferation, inflammation, differentiation, angiogenesis, transformation and apoptosis, pre-mRNA splicing, metabolic syndromes, fibrosis, and host defense. The role of Gal-3 has also been implicated in liver diseases. Gal-3 is activated upon a hepatotoxic insult to the liver and its level has been shown to be upregulated in fatty liver diseases, inflammation, nonalcoholic steatohepatitis, fibrosis, cholangitis, cirrhosis, and hepatocellular carcinoma (HCC). Gal-3 directly interacts with the NOD-like receptor family, pyrin domain containing 3, and activates the inflammasome in macrophages of the liver. In the chronically injured liver, Gal-3 secreted by injured hepatocytes and immune cells, activates hepatic stellate cells (HSCs) in a paracrine fashion to acquire a myofibroblast like collagen-producing phenotype. Activated HSCs in the fibrotic liver secrete Gal-3 which acts via autocrine signaling to exacerbate extracellular matrix synthesis and fibrogenesis. In the stromal microenvironment, Gal-3 activates cancer cell proliferation, migration, invasiveness, and metastasis. Clinically, increased serum levels and Gal-3 expression were observed in the liver tissue of nonalcoholic steatohepatitis, fibrotic/cirrhotic, and HCC patients. The pathological role of Gal-3 has been experimentally and clinically reported in the progression of chronic liver disease. Therefore, this review discusses the pathological role of Gal-3 in the progression of chronic liver diseases.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Molecular Medicine and Toxicology Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| |
Collapse
|
31
|
Ali FEM, Abd El-Aziz MK, Sharab EI, Bakr AG. Therapeutic interventions of acute and chronic liver disorders: A comprehensive review. World J Hepatol 2023; 15:19-40. [PMID: 36744165 PMCID: PMC9896501 DOI: 10.4254/wjh.v15.i1.19] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/17/2022] [Accepted: 12/21/2022] [Indexed: 01/16/2023] Open
Abstract
Liver disorders are one of the most common pathological problems worldwide. It affects more than 1.5 billion worldwide. Many types of hepatic cells have been reported to be involved in the initiation and propagation of both acute and chronic liver diseases, including hepatocytes, Kupffer cells, sinusoidal endothelial cells, and hepatic stellate cells (HSCs). In addition, oxidative stress, cytokines, fibrogenic factors, microRNAs, and autophagy are also involved. Understanding the molecular mechanisms of liver diseases leads to discovering new therapeutic interventions that can be used in clinics. Recently, antioxidant, anti-inflammatory, anti-HSCs therapy, gene therapy, cell therapy, gut microbiota, and nanoparticles have great potential for preventing and treating liver diseases. Here, we explored the recent possible molecular mechanisms involved in the pathogenesis of acute and chronic liver diseases. Besides, we overviewed the recent therapeutic interventions that targeted liver diseases and summarized the recent studies concerning liver disorders therapy.
Collapse
Affiliation(s)
- Fares EM Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | | | - Elham I Sharab
- Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Adel G Bakr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| |
Collapse
|
32
|
Wu S, Wang X, Xing W, Li F, Liang M, Li K, He Y, Wang J. An update on animal models of liver fibrosis. Front Med (Lausanne) 2023; 10:1160053. [PMID: 37035335 PMCID: PMC10076546 DOI: 10.3389/fmed.2023.1160053] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
The development of liver fibrosis primarily determines quality of life as well as prognosis. Animal models are often used to model and understand the underlying mechanisms of human disease. Although organoids can be used to simulate organ development and disease, the technology still faces significant challenges. Therefore animal models are still irreplaceable at this stage. Currently, in vivo models of liver fibrosis can be classified into five categories based on etiology: chemical, dietary, surgical, transgenic, and immune. There is a wide variety of animal models of liver fibrosis with varying efficacy, which have different implications for proper understanding of the disease and effective screening of therapeutic agents. There is no high-quality literature recommending the most appropriate animal models. In this paper, we will describe the progress of commonly used animal models of liver fibrosis in terms of their development mechanisms, applications, advantages and disadvantages, and recommend appropriate animal models for different research purposes.
Collapse
Affiliation(s)
- ShuTing Wu
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - XinXin Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - WenBo Xing
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - FenYao Li
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Ming Liang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - KeShen Li
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- *Correspondence: Yan He,
| | - JianMing Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- Department of Hepatobiliary and Pancreatic Surgery, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- JianMing Wang,
| |
Collapse
|
33
|
Nguyen G, Park SY, Do DV, Choi DH, Cho EH. Gemigliptin Alleviates Succinate-Induced Hepatic Stellate Cell Activation by Ameliorating Mitochondrial Dysfunction. Endocrinol Metab (Seoul) 2022; 37:918-928. [PMID: 36377343 PMCID: PMC9816499 DOI: 10.3803/enm.2022.1530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGRUOUND Dipeptidyl peptidase-4 inhibitors (DPP-4Is) are used clinically as oral antidiabetic agents. Although DPP-4Is are known to ameliorate liver fibrosis, the protective mechanism of DPP-4Is in liver fibrosis remains obscure. In this study, gemigliptin was used to investigate the potential of DPP-4Is to alleviate the progression of liver fibrosis. METHODS To clarify the effects and mechanisms of gemigliptin, we conducted various experiments in LX-2 cells (immortalized human hepatic stellate cells [HSCs], the principal effectors of hepatic fibrogenesis), which were activated by succinate and exhibited elevated expression of α-smooth muscle actin, collagen type 1, and pro-inflammatory cytokines and increased cell proliferation. In vivo, we examined the effects and mechanisms of gemigliptin on a high-fat, high-cholesterol-induced mouse model of nonalcoholic steatohepatitis (NASH). RESULTS Gemigliptin decreased the expression of fibrogenesis markers and reduced the abnormal proliferation of HSCs. In addition, gemigliptin reduced the succinate-induced production of mitochondrial reactive oxygen species (ROS), intracellular ROS, and mitochondrial fission in HSCs. Furthermore, in the mouse model of NASH-induced liver fibrosis, gemigliptin alleviated both liver fibrosis and mitochondrial dysfunction. CONCLUSION Gemigliptin protected against HSC activation and liver fibrosis by alleviating mitochondrial dysfunction and ROS production, indicating its potential as a strategy for preventing the development of liver disease.
Collapse
Affiliation(s)
- Giang Nguyen
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - So Young Park
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Dinh Vinh Do
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Dae-Hee Choi
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Eun-Hee Cho
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
- Corresponding author: Eun-Hee Cho. Department of Internal Medicine, Kangwon National University School of Medicine, 1 Gangwondaehak-gil, Chuncheon 24341, Korea Tel: +82-33-258-9167, Fax: +82-33-258-2455, E-mail:
| |
Collapse
|
34
|
Lv L, Shu H, Mo X, Tian Y, Guo H, Sun HY. Activation of the Nrf2 Antioxidant Pathway by Longjing Green Tea Polyphenols in Mice Livers. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221139409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Previous studies have revealed that green tea polyphenol (GTP) could protect against liver injury due to oxidative stress. However, the mechanism underlying the bioactive actions of GTP in the liver has not been systematically evaluated. This study aimed to investigate the effect of GTP on the activation of the nuclear factor erythroid-2-related factor 2 (Nrf2)-Kelch-like ECH-associated protein 1 (keap1) pathway, using in silico and in vivo methods. Furthermore, the regulation of Nrf2 downstream target antioxidant response element (ARE) was also evaluated. The high-performance liquid chromatography analysis indicated that GTP includes 9 major compounds, and molecule docking analysis demonstrated that most of these polyphenols have a strong binding affinity with the keap1 Kelch domain, where keap1 binds to the Neh2 domain of Nrf2. Remarkably, the predominant compound of GTP, that is, epigallocatechin gallate, displayed the best binding affinity score, which can fully occupy all 3 polar subpockets of the keap1 Kelch domain. The Nrf2, keap1, and Nrf2 downstream target gene expression levels were changed in the livers compared to the control group. It showed that the Nrf2 expression level was significantly upregulated in GTP-induced mice liver across most treatments, while the keap1 expression level remained unchanged. Subsequently, we observed a significant increasing trend in the expression of the downstream ARE, including antioxidative enzymes, liver phase II enzymes, and liver efflux transporters in mice livers. The present study demonstrated that GTP could activate the Nrf2 signaling pathway by interrupting the Nrf2-keap1 protein–protein interaction
Collapse
Affiliation(s)
- Le Lv
- Postdoctoral Innovation Practice Base, Shenzhen Polytechnic, Shenzhen, Guangdong, People's Republic of China
- School of Applied Biology, Shenzhen Institute of Technology, Shenzhen, Guangdong, People's Republic of China
| | - Haoyue Shu
- Postdoctoral Innovation Practice Base, Shenzhen Polytechnic, Shenzhen, Guangdong, People's Republic of China
| | - Xiaoye Mo
- School of Food and Drug, Shenzhen Polytechnic, Shenzhen, Guangdong, People's Republic of China
| | - Yongjing Tian
- School of Food and Drug, Shenzhen Polytechnic, Shenzhen, Guangdong, People's Republic of China
| | - Hui Guo
- School of Food and Drug, Shenzhen Polytechnic, Shenzhen, Guangdong, People's Republic of China
| | - Hai-Yan Sun
- School of Food and Drug, Shenzhen Polytechnic, Shenzhen, Guangdong, People's Republic of China
| |
Collapse
|
35
|
Morsy MA, Abdel-Latif R, Hafez SMNA, Kandeel M, Abdel-Gaber SA. Paeonol Protects against Methotrexate Hepatotoxicity by Repressing Oxidative Stress, Inflammation, and Apoptosis-The Role of Drug Efflux Transporters. Pharmaceuticals (Basel) 2022; 15:ph15101296. [PMID: 36297408 PMCID: PMC9611069 DOI: 10.3390/ph15101296] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Methotrexate (MTX) is an effective chemotherapeutic agent against a wide range of tumors and autoimmune diseases; however, hepatotoxicity limits its clinical use. Oxidative stress and inflammation have been implicated in the pathogenesis of MTX-induced hepatotoxicity. Paeonol is a natural phenolic compound reported for its antioxidant and anti-inflammatory properties. The current study aimed to investigate the protective effect of paeonol against MTX-induced hepatotoxicity in rats and various mechanisms that underlie this postulated effect. Paeonol was administered orally in a dose of 100 mg/kg, alone or along with MTX, for 10 days. Hepatotoxicity was induced via a single intraperitoneal dose of MTX (20 mg/kg) on day 5 of the experiment. Concomitant administration of paeonol with MTX significantly ameliorated distorted hepatic function and histological structure, restored hepatic oxidative stress parameters (MDA, NO, and SOD), and combated inflammatory response (iNOS and TNF-α). Additionally, paeonol enhanced cell proliferation and survival, evidenced by upregulating the proliferating cell nuclear antigen (PCNA) and suppressing apoptosis and the disposition of collagen fibers in rat livers treated with MTX. Importantly, paeonol upregulated the drug efflux transporters, namely P-glycoprotein (P-gp) and the multidrug resistance-associated protein 2 (Mrp-2) in MTX-treated rats. In conclusion, paeonol offered a potent protective effect against MTX-induced hepatotoxicity through suppressing oxidative stress, inflammation, fibrosis, and apoptosis pathways, along with P-gp and Mrp-2 upregulation.
Collapse
Affiliation(s)
- Mohamed A. Morsy
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al Hofuf 31982, Al-Ahsa, Saudi Arabia
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt
- Correspondence: ; Tel.: +966-5496-72245
| | - Rania Abdel-Latif
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Minia University, El-Minia 61511, Egypt
| | | | - Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al Hofuf 31982, Al-Ahsa, Saudi Arabia
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Seham A. Abdel-Gaber
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt
| |
Collapse
|
36
|
Bioinformatics Analysis of Common Genetic and Molecular Traits and Association of Portal Hypertension with Pulmonary Hypertension. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:9237701. [PMID: 36312597 PMCID: PMC9613398 DOI: 10.1155/2022/9237701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/19/2022] [Accepted: 09/28/2022] [Indexed: 12/16/2022]
Abstract
Portal hypertension (PH) is an important cause of pulmonary arterial hypertension(PAH), but its mechanism is still unclear. We used genetic data analysis to explore the shared genes and molecular mechanisms of PH and PAH. We downloaded the PH and PAH data from the GEO database, and used the weighted gene coexpression network analysis method (WGCNA) to analyze the coexpression modules of idiopathic noncirrhotic portal hypertension (INCPH) and cirrhotic portal hypertension (CPH) and pulmonary hypertension, respectively. Enrichment analysis was performed on the common genes, and differential gene expressions (DEGs) were used for verification. The target genes of INCPH and PAH were obtained by string and cytoscape software, and the miRNAs of target genes were predicted by miRwalk, miRDB, and TargetScan and their biological functions were analyzed; finally, we used PanglaoDB to predict the expression of target genes in cells. In WGCNA, gene modules significantly related to PAH, CPH, and INCPH were identified, and enrichment function analysis showed that the common pathway of PAH and CPH were “P53 signaling pathway,” “synthesis of neutral lipids”; PAH and INCPH are “terminal,” “Maintenance Regulation of Granules,” and “Toxin Transport.” DEGs confirmed the results of WGCNA; the common miRNA functions of PAH and cirrhosis were enriched for “P53 signaling pathway,” “TGF-β signaling pathway,” “TNF signaling pathway,” and “fatty acid metabolism,” and the miRNAs-mRNAs network suggested that hsa-miR-22a-3p regulates MDM2 and hsa-miR-34a-5p regulates PRDX4; the target genes of PAH and INCPH are EIF5B, HSPA4, GNL3, RARS, UTP20, HNRNPA2B1, HSP90B1, METAP2, NARS, SACM1L, and their target miRNA function enrichment showed EIF5B, HNRNPA2B1, HSP90B1, METAP2, NARS, SACM1L, and HSPA4 are associated with telomeres and inflammation, panglaoDB showed that target genes are located in endothelial cells, smooth muscle cells, etc. In conclusion, the mechanism of pulmonary hypertension induced by portal hypertension may be related to telomere dysfunction and P53 overactivation, and lipid metabolism and intestinal inflammation are also involved in this process.
Collapse
|
37
|
Ezhilarasan D, Mani U. Valproic acid induced liver injury: An insight into molecular toxicological mechanism. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 95:103967. [PMID: 36058508 DOI: 10.1016/j.etap.2022.103967] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 06/15/2023]
Abstract
Valproic acid (VPA) is an anti-seizure drug that causes idiosyncratic liver injury. 2-propyl-4-pentenoic acid (Δ4VPA), a metabolite of VPA, has been implicated in VPA-induced hepatotoxicity. This review summarizes the pathogenesis involved in VPA-induced liver injury. The VPA induce liver injury mainly by i) liberation of Δ4VPA metabolites; ii) decrease in glutathione stores and antioxidants, resulting in oxidative stress; iii) inhibition of fatty acid β-oxidation, inducing mitochondrial DNA depletion and hypermethylation; a decrease in proton leak; oxidative phosphorylation impairment and ATP synthesis decrease; iv) induction of fatty liver via inhibition of carnitine palmitoyltransferase I, enhancing nuclear receptor peroxisome proliferator-activated receptor-gamma and acyl-CoA thioesterase 1, and inducing long-chain fatty acid uptake and triglyceride synthesis. VPA administration aggravates liver injury in individuals with metabolic syndromes. Therapeutic drug monitoring, routine serum levels of transaminases, ammonia, and lipid parameters during VPA therapy may thus be beneficial in improving the safety profile or preventing the progression of DILI.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600 077, India.
| | - Uthirappan Mani
- Animal House Division, CSIR-Central Leather Research Institute, Adyar, Chennai 600 020, India
| |
Collapse
|
38
|
Gheena S, Ezhilarasan D, Shree Harini K, Rajeshkumar S. Syringic acid and silymarin concurrent administration inhibits sodium valproate-induced liver injury in rats. ENVIRONMENTAL TOXICOLOGY 2022; 37:2143-2152. [PMID: 35543257 DOI: 10.1002/tox.23557] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/08/2022] [Accepted: 04/27/2022] [Indexed: 06/14/2023]
Abstract
Sodium valproate (SV) is a well-known anti-epileptic drug, also used to control convulsions, bipolar disorders and migraines. SV has been shown to induce liver toxicity in clinical subjects. Syringic acid (SA), a natural polyphenolic compound has potential antioxidant, anti-inflammatory and several beneficial effects. Therefore, in this study, we evaluated hepatoprotective effect of SA against SV-induced liver injury in rats. Wistar rats were treated with SV orally at a dose of 500 mg/kg, once daily, for 14 days. Another three groups of rats were administered with SV and concurrently treated with SA (40 and 80 mg/kg) and silymarin (SIL) (100 mg/kg) for 14 days. SV administration for 14 days caused significant (p < .001) elevation of liver transaminases and ALP in serum. Liver MDA level was significantly (p < .001) increased with a concomitant decrease (p < .001) in enzymic antioxidants activities in SV administered rats. SV administration also caused the upregulation of proinflammatory markers such as tumor necrosis factor α, c-Jun N-terminal kinase, nuclear factor kappa B, cyclooxygenase-2 and Interleukin 6 expressions in liver tissue. Histopathological studies also revealed the presence of inflammatory cell infiltration and hepatocellular necrosis upon SV administration. At both doses, concurrent administration of SA and SIL significantly (p < .001) inhibited the liver transaminase activities in serum, oxidative stress, and proinflammatory markers expression in liver tissue. Our current results suggest that SA can be a promising herbal drug that can inhibit SV-induced hepatotoxicity when administered together due its potential anti-inflammatory effects.
Collapse
Affiliation(s)
- Sukumaran Gheena
- Department of Oral Pathology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Devaraj Ezhilarasan
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Karthik Shree Harini
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Shanmugam Rajeshkumar
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| |
Collapse
|
39
|
Mustafa G, Zia-ur-Rehman M, Sumrra SH, Ashfaq M, Zafar W, Ashfaq M. A critical review on recent trends on pharmacological applications of pyrazolone endowed derivatives. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
40
|
Devaraj E, Perumal E, Subramaniyan R, Mustapha N. Liver fibrosis: Extracellular vesicles mediated intercellular communication in perisinusoidal space. Hepatology 2022; 76:275-285. [PMID: 34773651 DOI: 10.1002/hep.32239] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 10/29/2021] [Accepted: 11/10/2021] [Indexed: 12/17/2022]
Affiliation(s)
- Ezhilarasan Devaraj
- Department of Pharmacology, The Blue Lab, Molecular Pharmacology and Toxicology Division, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Elumalai Perumal
- Department of Pharmacology, The Blue Lab, Molecular Pharmacology and Toxicology Division, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Raghunandhakumar Subramaniyan
- Department of Pharmacology, The Blue Lab, Molecular Pharmacology and Toxicology Division, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Najimi Mustapha
- Laboratory of Pediatric Hepatology and Cell Therapy, IREC Institute, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
41
|
Chen X, Shen M, Yang J, Yu Q, Chen Y, Wang X, Lu H, Tao X, Li H, Xie J. RNA-seq based elucidation of mechanism underlying Mesona chinensis Benth polysaccharide protected H2O2-induced oxidative damage in L02 cells. Food Res Int 2022; 157:111383. [DOI: 10.1016/j.foodres.2022.111383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/06/2022] [Accepted: 05/14/2022] [Indexed: 02/08/2023]
|
42
|
Oxidative Stress in Chronic Hepatitis B—An Update. Microorganisms 2022; 10:microorganisms10071265. [PMID: 35888983 PMCID: PMC9318593 DOI: 10.3390/microorganisms10071265] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/19/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
In recent years, the role of oxidative stress has been investigated in an increasing number of infections. There is a close link between the inflammation that accompanies infections and oxidative stress. Excessive reactive oxygen species induce harmful effects on cell components, including lipids, proteins, and nucleic acids. A growing body of evidence attests to the role of oxidative stress in the pathogenesis of viral liver infections, especially in hepatitis C virus (HCV) infection. Regarding hepatitis B virus (HBV) infection, the data are limited, but important progress has been achieved in recent years. This review presents the latest advances pertaining to the role of the oxidative stress byproducts in the pathogenesis of chronic hepatitis B, constituting a source of potential new markers for the evaluation and monitoring of patients with chronic hepatitis B.
Collapse
|
43
|
Intervention Study of Dictyophora Polysaccharides on Arsenic-Induced Liver Fibrosis in SD Rats. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7509620. [PMID: 35402611 PMCID: PMC8986371 DOI: 10.1155/2022/7509620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/13/2022] [Accepted: 03/16/2022] [Indexed: 12/12/2022]
Abstract
Long-term arsenic (As) exposure can cause liver injury, hepatic cirrhosis, and cancer. Meanwhile, Dictyophora polysaccharides (DIP) have excellent antioxidation, anti-inflammation, and immune protection effects. There are currently few reports on the protection effects of DIP on As-induced hepatotoxicity and its pharmacological value. Therefore, this study was aimed at elucidating the protection of DIP on As-induced hepatotoxicity and exploring its preventive role in antifibrosis. In our study, the SD rat As poisoning model was established by the feeding method to explore the influence of As exposure on liver fibrosis. Then, DIP treatment was applied to the rats with As-induced liver fibrosis, and the changes of serum biochemical indexes and liver tissue pathology were observed. And the expression of fibrosis-related proteins TGF-β1, CTGF, and α-SMA levels was then determined to explore the DIP intervention function. The results demonstrated that through reduced pathological changes of hepatic and increased serum AST, ALT, TP, ALB, and A/G levels, DIP ameliorated liver fibrosis induced by As as reflected. And the administration of DIP decreased the concentration of HA, LN, PCIII, CIV, TBIL, and DBIL. In addition, the synthesis of TGF-β1 inhibited by DIP might regulate the expression of CTGF and decrease the proliferation of fibrinogen and fibroblasts, which reduced the synthesis of fibroblasts to transform into myofibroblasts. And a decrease of myofibroblasts downregulated the expression of α-SMA, which affected the synthesis and precipitation of ECM and alleviated the liver fibrosis caused by exposure to As. In conclusion, based on the pathological changes of liver tissue, serum biochemical indexes, and related protein expression, DIP can improve the As-induced liver fibrosis in rats and has strong medicinal value.
Collapse
|
44
|
Pang Z, Xu Y, Zhu Q. Early Growth Response 1 Suppresses Macrophage Phagocytosis by Inhibiting NRF2 Activation Through Upregulation of Autophagy During Pseudomonas aeruginosa Infection. Front Cell Infect Microbiol 2022; 11:773665. [PMID: 35096638 PMCID: PMC8790152 DOI: 10.3389/fcimb.2021.773665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/22/2021] [Indexed: 12/11/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that causes life-threatening infections in cystic fibrosis patients and immunocompromised individuals. A tightly regulated immune response possessed by healthy individuals can effectively control P. aeruginosa infections, whereas the patients with dysregulated immune response are susceptible to this bacterial pathogen. Early growth response 1 (Egr-1) is a zinc-finger transcription factor involved in regulation of various cellular functions, including immune responses. We previously identified that Egr-1 was deleterious to host in a mouse model of acute P. aeruginosa pneumonia by promoting systemic inflammation and impairing bacterial clearance in lung, which associated with reduced phagocytosis and bactericidal ability of leucocytes, including macrophages and neutrophils. However, the molecular mechanisms underlying the Egr-1-suppressed phagocytosis of P. aeruginosa are incompletely understood. Herein, we investigated whether the Egr-1-regulated autophagy play a role in macrophage phagocytosis during P. aeruginosa infection by overexpression or knockdown of Egr-1. We found that overexpression of Egr-1 inhibited the phagocytic activity of macrophages, and the autophagy activator rapamycin and inhibitor chloroquine could reverse the effects of Egr-1 knockdown and Egr-1 overexpression on phagocytosis of P. aeruginosa, respectively. Furthermore, the Egr-1-overexpressing macrophages displayed upregulated expression of autophagy-related proteins LC3A, LC3B and Atg5, and decreased levels of p62 in macrophages. Further studies revealed that the macrophages with Egr-1 knockdown displayed enhanced activation of transcription factor NRF2 and expression of scavenger receptors MACRO and MSR1. Altogether, these findings suggest that Egr-1 suppresses the phagocytosis of P. aeruginosa by macrophages through upregulation of autophagy and inhibition of NRF2 signaling.
Collapse
Affiliation(s)
- Zheng Pang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Xu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingjun Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
45
|
Beneficial Effects of Jujube Juice Fermented by Lactobacillus plantarum NXU19009 on Acute Alcoholic Liver Injury in Mice. FERMENTATION-BASEL 2022. [DOI: 10.3390/fermentation8020054] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Red jujube (Ziziphus jujuba Mill.) is an important fruit that has the concomitant function of both medicine and food. It has been proven to be rich in various bioactive components. In the present study, jujube juice was fermented by Lactobacillus plantarum NXU19009 to enhance the flavor and nutritional benefits. Its potential for the prevention and treatment of acute alcohol induced-liver injury in mice was examined in this study. The results showed that the administration of the fermented jujube juice along with alcohol significantly decreased (p < 0.01) the liver indices, as well as the levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), alcohol dehydrogenase (ADH), total triglyceride (TG), total cholesterol (TC), and liver malondialdehyde (MDA) in the serum. In contrast, the levels of liver superoxide dismutase (SOD) and glutathione (GSH) in mice administered with fermented jujube juice were found to increase significantly (p < 0.01). Furthermore, the administration of fermented jujube juice in mice was found to alter their intestinal microbiota and an improvement was observed based on the results obtained in the histopathology examination. Therefore, Jujube juice fermented by Lactobacillus plantarum NXU19009 protects against liver injury and may prove to be an effective supplement to attenuate acute alcoholic liver injury.
Collapse
|
46
|
Vigneshwaran R, Ezhilarasan D, Rajeshkumar S. Inorganic titanium dioxide nanoparticles induces cytotoxicity in colon cancer cells. INORG CHEM COMMUN 2021. [DOI: 10.1016/j.inoche.2021.108920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
47
|
Ezhilarasan D. Deciphering the toxicological role of Porphyromonas gingivalis derived endotoxins in liver diseases. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 88:103755. [PMID: 34662732 DOI: 10.1016/j.etap.2021.103755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 06/13/2023]
Abstract
Periodontitis is a most prevalent and infectious multifactorial inflammatory disease and is characterized by the progressive destruction of the tooth-supporting tissues. Porphyromonas gingivalis, a Gram‑negative oral anaerobe, mainly causes periodontitis and it is one of the most important risk factors responsible for aggravation of existing systemic diseases. Several experimental and clinical studies have shown the positive association between periodontitis and different forms of liver disease. Periodontal diseases increase the prevalence of non-alcoholic fatty liver diseases and cirrhosis. Infected periodontium and pathogens in the periodontal microenvironments release pathogen-associated molecular patterns such as peptidoglycan, lipopolysaccharides, gingipain, fimbria, bacterial DNA, etc, and damage-associated molecular patterns such as interleukins-1α, β, - 8, and galectin-3, etc. These virulence factors and cytokines enter the bloodstream, disseminate into the whole body, and induce a variety of systemic pathological effects, including liver diseases (steatosis and fibrosis). Maintaining oral hygiene by scaling and root planning significantly improves liver damage in patients with periodontitis. Dentists and physicians should have more awareness in understanding the bidirectional nature of the relationship between oral and systemic diseases. Importantly, periodontitis condition aggravates simple fatty liver into fibrotic disease and therefore, the aim of this review is to understand the possible link between periodontitis and liver diseases.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, The Blue Lab, Molecular Medicine and Toxicology Division, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600 077, India.
| |
Collapse
|
48
|
Ezhilararasan D, Lakshmi T, Raut B. Novel Nano-Based Drug Delivery Systems Targeting Hepatic Stellate Cells in the Fibrotic Liver. JOURNAL OF NANOMATERIALS 2021; 2021:1-9. [DOI: 10.1155/2021/4674046] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Hepatic stellate cells (HSCs) exist in the liver’s perisinusoidal space, are phenotypically activated, and acquire myofibroblast-like phenotype. This phenotypic transformation is accountable for the accumulation and production of various extracellular matrix (ECM) proteins, involving different fibril-forming collagens in the perisinusoidal space, producing altered hepatic function and portal hypertension and increased vascular resistance, fibrosis, cirrhosis, and hepatocellular carcinoma. The activated HSCs/myofibroblasts are principal collagen-producing cells in the damaged liver. Therefore, fibrosis treatments are often targeting HSCs. HSCs store most of the total body’s retinol in their cytoplasm, and hence, antifibrotic nanomedicines are often targeted with vitamin A decoration. Vitamin A-decorated nanomedicines with siRNAs for transforming growth factor-beta, collagen, and connective tissue growth factors target to inhibit fibrogenesis and the ECM-associated gene expressions, leading to fibrosis regression. Similarly, a variety of miRNAs play pro- and antifibrotic function. In the fibrotic liver, the profibrotic miRNAs are targeted with their respective antagomir and the antifibrotic miRNAs are targeted with their respective agomirs along with HSC-specific nanodecoration. These miRNA treatments reduce fibrogenesis by downregulation of ECM-related gene expressions. However, liver fibrosis is caused by the upregulation of a different type of profibrotic signaling pathways associated with ECM accumulation in the fibrotic liver. Therefore, specific gene silencing by siRNAs or targeting particularly miRNA may also not effectively reduce fibrosis to a greater extent. However, nanodecoration of a drug is useful to deliver drugs into activated HSCs in the injured liver. Therefore, the aim of this review is to focus on targeted drug delivery towards activated HSCs in the persistently damaged liver.
Collapse
Affiliation(s)
- Devaraj Ezhilararasan
- Department of Pharmacology, The Blue Lab, Molecular Medicine and Toxicology Division, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu 600077, India
| | - Thangavelu Lakshmi
- Department of Pharmacology, The Blue Lab, Molecular Medicine and Toxicology Division, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu 600077, India
| | - Biond Raut
- Department of Pharmacology, Kathmandu Medical College Bhaktapur Duwakot, Kathmandu University, Dhulikhel, Kavre, Nepal
| |
Collapse
|
49
|
Zhou F, She W, He L, Zhu J, Gu L. The effect of anthocyanins supplementation on liver enzymes among patients with metabolic disorders: A systematic review and meta-analysis of randomized clinical trials. Phytother Res 2021; 36:53-61. [PMID: 34510592 DOI: 10.1002/ptr.7280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/10/2021] [Accepted: 08/26/2021] [Indexed: 12/16/2022]
Abstract
The present study aims to summarize and quantitatively examine the available evidence on the effectiveness of anthocyanin supplementation on liver enzymes among patients with metabolic disorders, by employing a systematic review and meta-analytic approach. Online databases including PubMed/Medline, Scopus, ISI Web of Science, and Cochrane Library were searched up to June 2020 for randomized controlled trials (RCTs) that examined the effect of anthocyanin supplementation on serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) among patients with metabolic disorders. To estimate the overall effect of anthocyanin supplementation, we employed the random-effects model. In total, 12 RCTs were included in the systematic review. Pooled analysis did not show any significant changes in ALT (WMD: -0.92 U/L, 95% CI: -4.19 to 2.35, p = .58; I2 = 91.3%) and AST (WMD: -1.22 U/L, 95% CI: -3.43 to 0.99, p = .28; I2 = 87.0) concentrations after supplementation with anthocyanin. The dose and duration of supplementation were the potential sources of heterogeneity among most of the trials. However, subgroup analysis showed that the effect is not statistically significant in all subgroups. Overall, in our study, anthocyanin does not have any effect on liver enzyme levels significantly. However, future high-quality studies are still needed to confirm the results.
Collapse
Affiliation(s)
- Fengling Zhou
- Pediatrics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei She
- Clinical Laboratories, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lin He
- Department of Pharmacy, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiuqun Zhu
- Department of Pharmacy, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lu Gu
- Department of Radiation, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
50
|
Ezhilarasan D. Hepatic stellate cells in the injured liver: Perspectives beyond hepatic fibrosis. J Cell Physiol 2021; 237:436-449. [PMID: 34514599 DOI: 10.1002/jcp.30582] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/22/2021] [Accepted: 08/30/2021] [Indexed: 12/14/2022]
Abstract
Over the last two decades, our understanding of the pathological role of hepatic stellate cells (HSCs) in fibrotic liver disease has increased dramatically. As HSCs are identified as the principal collagen-producing cells in the injured liver, several experimental and clinical studies have targeted HSCs to treat liver fibrosis. However, HSCs also play a critical role in developing nonfibrotic liver diseases such as cholestasis, portal hypertension, and hepatocellular carcinoma (HCC). Therefore, this review exclusively focuses on the role of activated HSCs beyond hepatic fibrosis. In cholestasis conditions, elevated bile salts and bile acids activate HSCs to secrete collagen and other extracellular matrix products, which cause biliary fibrosis and cholangitis. In the chronically injured liver, autocrine and paracrine signaling from liver sinusoidal endothelial cells activates HSCs to induce portal hypertension via endothelin-1 release. In the tumor microenvironment (TME), activated HSCs are the major source of cancer-associated fibroblasts (CAF). The crosstalk between activated HSC/CAF and tumor cells is associated with tumor cell proliferation, migration, metastasis, and chemoresistance. In TME, activated HSCs convert macrophages to tumor-associated macrophages and induce the differentiation of dendritic cells (DCs) and monocytes to regulatory DCs and myeloid-derived suppressor cells, respectively. This differentiation, in turn, increases T cells proliferation and induces their apoptosis leading to reduced immune surveillance in TME. Thus, HSCs activation in chronically injured liver is a critical process involved in the progression of cholestasis, portal hypertension, and liver cancer.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, The Blue Lab, Molecular Medicine and Toxicology Division, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| |
Collapse
|