1
|
Sultana M, Islam MA, Khairnar R, Kumar S. A guide to pathophysiology, signaling pathways, and preclinical models of liver fibrosis. Mol Cell Endocrinol 2025; 598:112448. [PMID: 39755140 DOI: 10.1016/j.mce.2024.112448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 01/06/2025]
Abstract
Liver fibrosis is potentially a reversible form of liver disease that evolved from the early stage of liver scarring as a consequence of chronic liver injuries. Recurrent injuries in the liver without any appropriate medication cause the injuries to get intense and deeper, which gradually leads to the progression of irreversible cirrhosis or carcinoma. Unfortunately, there are no approved treatment strategies for reversing hepatic fibrosis, making it one of the significant risk factors for developing advanced liver disorders and liver disease-associated mortality. Consequently, the interpretation of the fundamental mechanisms, etiology, and pathogenesis is crucial for identifying the potential therapeutic target as well as evaluating novel anti-fibrotic therapy. However, despite innumerable research, the functional mechanism and disease characteristics are still obscure. To accelerate the understanding of underlying disease pathophysiology, molecular pathways and disease progression mechanism, it is crucial to mimic human liver disease through the formation of precise disease models. Although various in vitro and in vivo liver fibrotic models have emerged and developed already, a perfect clinical model replicating human liver diseases is yet to be established, which is one of the major challenges in discovering proper therapeutics. This review paper will shed light on pathophysiology, signaling pathways, preclinical models of liver fibrosis, and their limitations.
Collapse
Affiliation(s)
- Mehonaz Sultana
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Md Asrarul Islam
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Rhema Khairnar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Sunil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| |
Collapse
|
2
|
Yaribeygi H, Kashian K, Moghaddam KI, Karim SR, Bagheri N, Karav S, Jamialahmadi T, Rizzo M, Sahebkar A. Hepatic effects of GLP-1 mimetics in diabetic milieu: A mechanistic review of involved pathways. J Diabetes Complications 2025; 39:108928. [PMID: 39644538 DOI: 10.1016/j.jdiacomp.2024.108928] [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: 09/07/2024] [Revised: 11/25/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Patients with diabetic are at a higher risk of developing hepatic disorders compared to non-diabetic individuals. This increased risk can be attributed to the diabetic environment, which triggers and exacerbates harmful pathways involved in both diabetic complications and hepatic disorders. Therefore, it is important to consider the use of antidiabetic agents that offer benefits beyond glycemic control and have positive effects on liver tissues. Glucagon-like peptide-1 (GLP-1) mimetics are a novel class of antidiabetic medications known for their potent blood sugar-lowering effects. Emerging evidence suggests that these drugs also have favorable effects on the liver. However, the precise effects and underlying mechanisms are not yet fully understood. In this review, we aim to provide a mechanistic perspective on the liver benefits of GLP-1 mimetics and outline the mediating mechanisms involved.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Kiana Kashian
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | | | | | - Narges Bagheri
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Manfredi Rizzo
- School of Medicine, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (Promise), University of Palermo, Italy; Department of Biochemistry, Mohamed Bin Rashid University, Dubai, United Arab Emirates
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Prajapati J, Bhatt N, Rawal R. Hepatoprotective effects of phytochemicals and plant extracts against chemotherapy-induced liver damage in animal models: a systematic review. Arch Toxicol 2024:10.1007/s00204-024-03928-7. [PMID: 39729113 DOI: 10.1007/s00204-024-03928-7] [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: 10/12/2024] [Accepted: 11/28/2024] [Indexed: 12/28/2024]
Abstract
Chemotherapy, a cornerstone of cancer treatment, is frequently marred by its hepatotoxic effects, which can significantly impede therapeutic efficacy. This systematic review meticulously evaluates the hepatoprotective properties of phytochemicals and plant extracts against chemotherapy-induced liver damage, primarily in experimental animal models. Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, an exhaustive search was conducted across databases like SCOPUS, PubMed, and Web of Science, culminating in the inclusion of 61 pertinent studies. These studies illustrate those natural compounds, spanning a diverse array of phytochemicals and plant extracts that can effectively mitigate biochemical markers of liver damage, enhance antioxidant defences, and modulate inflammatory responses in model organisms subjected to hepatotoxic chemotherapeutic agents such as cyclophosphamide, cisplatin, and doxorubicin. Notably, the natural agents reviewed have demonstrated significant reductions in liver enzymes, improved histopathological outcomes, and bolstered cellular antioxidant capacities. The systematic synthesis of data underscores the potential of these natural substances to diminish liver toxicity associated with chemotherapy in preclinical settings. However, the review also highlights critical gaps in research, notably the underreporting of molecular mechanisms and inconsistent data on clinical translatability. To optimize the therapeutic utility of these compounds, future studies should focus on detailed molecular analyses and rigorous clinical trials to validate efficacy and safety, paving the way for integrated approaches in oncological care that minimize hepatic complications.
Collapse
Affiliation(s)
- Jignesh Prajapati
- Department of Biochemistry and Forensic Science, University School of Sciences, Gujarat University, Ahmedabad, 380009, Gujarat, India
- INVENTAYU Private Limited, AIC-LMCP Foundation, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Narendra Bhatt
- INVENTAYU Private Limited, AIC-LMCP Foundation, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380009, Gujarat, India.
| | - Rakesh Rawal
- Department of Medical Biotechnology, Gujarat Biotechnology University, GIFT City, Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
4
|
Nie P, Hu L, You T, Jia T, Xu H. Lead Mediated Lipopolysaccharides Exacerbates Fatty Liver Processes in High-Fat Diets-Induced Mice. ENVIRONMENTAL TOXICOLOGY 2024. [PMID: 39715151 DOI: 10.1002/tox.24463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 10/10/2024] [Accepted: 12/08/2024] [Indexed: 12/25/2024]
Abstract
Obesity leads to a variety of health risks, and lead, which is ranked second in Agency for Toxic Substances and Disease Registry's priority list of harmful substances, may be more harmful to individuals that are obese. C57BL/6 mice were fed a normal diet or a high-fat diet with or without exposure to 1 g/L lead exposure in drinking water for 8 consecutive weeks. Serum and hepatic biochemistry analysis, histopathological observation, and RT-qPCR were used to explore the potential mechanism of liver damage in obese individuals after Pb exposure, and fecal microbiota transplantation was performed to investigate the role of the gut microbiota in the progression of fatty liver disease. We found that the progression of fatty liver disease induced by high-fat diets was accelerated by chronic lead intake. In addition, the occurrences of liver injury in recipient mice suggested the role of the gut microbiota. These findings indicated that the combination of lead and a HFD exacerbated hepatic lipotoxicity by activating LPS-mediated inflammation, and that gut microbiota disorders and impaired intestinal barrier function play pivotal roles in the progression of fatty liver disease.
Collapse
Affiliation(s)
- Penghui Nie
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Liehai Hu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Tao You
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Tiantian Jia
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- International Institute of Food Innovation co. Ltd., Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
5
|
Abdelrahman RS, Elnfarawy AA, Nashy AE, Abdelsalam RA, Zaghloul MS. Targeting angiogenic and proliferative mediators by montelukast & trimetazidine Ameliorates thioacetamide-induced liver fibrosis in rats. Toxicol Appl Pharmacol 2024; 495:117208. [PMID: 39716576 DOI: 10.1016/j.taap.2024.117208] [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: 09/16/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024]
Abstract
Liver fibrosis is a significant health complication with the potential to result in serious mortality and morbidity. However, there is no standard treatment due to its complex pathogenesis. The drug montelukast reversibly and selectively antagonizes the cysteinyl-leukotrienes-1 receptor and reduces inflammation; thus, it is used in the treatment of asthma. Trimetazidine, an anti-anginal agent, selectively inhibits the activity of mitochondrial long-chain 3-ketoacyl-CoA thiolase, inhibition of free fatty acid (FFA) oxidation. This study explores the efficacy of montelukast (5 and 10 mg/kg) and trimetazidine (10-20 mg/kg) against liver fibrosis induced by thioacetamide (TAA) in rats. Impaired liver function tests were significantly improved by montelukast and trimetazidine. The antioxidant and anti-inflammatory effects of montelukast and trimetazidine were proved by the inhibition of malondialdehyde (MDA) and nitric oxide (NO) accumulation, with elevation of glutathione (GSH) and superoxide dismutase activity, decreased heat shock protein (HSP-70) expression, and a decline in interleukin-6 (IL-6) and tumor necrosis factor (TNF-α) levels in liver tissue. Also, the antifibrotic effects were explored by reducing levels of hydroxyproline and alpha-smooth muscle actin (α-SMA) expression in liver tissue and attenuating hepatic expression of hepatic expression of angiogenic mediator vascular endothelium growth factor (VEGF) and proliferative mediator Antigen Kiel 67 (Ki-67).
Collapse
Affiliation(s)
- Rehab S Abdelrahman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Taibah University, Al-Madina Al-Munawwarah, 30001, Saudi Arabia.
| | - Ahmed A Elnfarawy
- Biotechnology Lab, Central Administration of Biological and Innovative Products and Clinical Studies, Giza, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | | | - Ramy A Abdelsalam
- Lecturer of Pathology, Faculty of Medicine, Mansoura University, Egypt
| | - Marwa S Zaghloul
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura National University, Gamasa, 7731168, Egypt
| |
Collapse
|
6
|
Alfwuaires MA. Rosmarinic acid protects against cyclophosphamide-induced hepatotoxicity via inhibition of oxidative stress, inflammation, and apoptosis and upregulation of Nrf2 in mice. J Mol Histol 2024; 56:49. [PMID: 39702535 DOI: 10.1007/s10735-024-10290-6] [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: 09/17/2024] [Accepted: 10/29/2024] [Indexed: 12/21/2024]
Abstract
Cyclophosphamide (CP) is widely used in chemotherapy to treat various types of cancer. However, it is toxic to the liver and other organs. Rosmarinic acid (RA) possesses anti-inflammatory, antioxidant, and cytoprotective properties. This study investigated the protective effects of RA against CP-induced liver injury in mice. Mice were treated with RA (25, 50, and 100 mg/kg) for 15 days and followed by a single injection of CP on day 16th. CP injection resulted in an elevation in serum AST, ALT, and ALP, along with multiple histopathological alterations in the liver. CP also induced increased levels of MDA and NO, associated with declined GSH, SOD and CAT. RA pretreatment prevented liver injury, alleviated the enhanced levels of MDA and NO, and restored antioxidants defenses, hence avoiding the oxidative injury in the liver. Moreover, RA pretreatment attenuated NF-κB p65 and proinflammatory cytokines levels. Liver of CP-injected mice also showed a decrease in Bcl2, accompanied with elevated BAX and caspase-3 expression, an effect that RA pretreatment alleviated. In addition, pretreatment of CP-administrated mice with RA restored the Nrf2 expression in the liver. Taken together, this study suggests a potential application value of RA in preventing CP hepatotoxicity and sheds light on the possible mechanism.
Collapse
Affiliation(s)
- Manal A Alfwuaires
- Department of Biological Sciences, Faculty of Science, King Faisal University, 31982, Al Hofuf, Al-Ahsa, Saudi Arabia.
| |
Collapse
|
7
|
Yao L, Chen T. A combined association of alanine aminotransferase, aspartate transaminase and bilirubin with sleep duration in aged 16-85 years (2005-2010). Medicine (Baltimore) 2024; 103:e40915. [PMID: 39654161 PMCID: PMC11630931 DOI: 10.1097/md.0000000000040915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/13/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024] Open
Abstract
Sleep is a vital restorative process that plays a pivotal role in maintaining the delicate equilibrium of mental and physical well-being. Both short and long sleep duration are associated with a range of adverse health outcomes. Numerous studies have consistently demonstrated a robust association between sleep duration and liver disease. In this study, we conducted statistical tests and performed subgroup analyses to explore potential variations in this association across different contexts, aiming to elucidate the correlation between ALT, AST, and TB with sleep duration. This cross-sectional investigation utilized datasets from the National Health and Nutrition Examination Survey 2005 to 2010. Multivariate linear regression models were used to examine the linear association between ALT, AST, and TB with sleep duration. Test for interaction is commonly conducted using multivariabte models to assess statistically significant subgroup disparities. Fitted smoothied curves and threshold effect analyses were employed to depict nonlinear relationships. The study enrolled 17,491 participants aged 16 to 85 years who met the inclusion and exclusion criteria, with a mean age of the participants was 45.58 ± 19.94 years. Multivariate linear regression analysis showed a significant positive association between sleep duration and ALT [-0.23 (-0.45, -0.00) 0.0455] and AST[-0.20 (-0.38, -0.01) 0.0338] in Model 3. Using a two-segment linear regression model, we found an U-shaped relationship and significant inflection point between between ALT and AST with sleep duration. The present study unveiled a significant inverse correlation between sleep duration and levels of ALT and AST, while no significant association was observed with TB levels. Furthermore, variations in the optimal sleep duration for liver function recovery were identified across diverse populations, thereby offering valuable healthcare recommendations to public.
Collapse
Affiliation(s)
- Lishuai Yao
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Tiantian Chen
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
8
|
Subramaniam NK, Mann KK. Mechanisms of Metal-Induced Hepatic Inflammation. Curr Environ Health Rep 2024; 11:547-556. [PMID: 39499483 DOI: 10.1007/s40572-024-00463-6] [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] [Accepted: 09/16/2024] [Indexed: 11/07/2024]
Abstract
PURPOSE OF REVIEW Worldwide, there is an increasing prevalence of hepatic diseases. The most common diseases include alcoholic-associated liver disease (ALD), metabolic dysfunction-associated fatty liver disease/ metabolic dysfunction-associated steatohepatitis (MAFLD/MASH) and viral hepatitis. While there are many important mediators of these diseases, there is increasing recognition of the importance of the inflammatory immune response in hepatic disease pathogenesis. RECENT FINDINGS Hepatic inflammation triggers the onset and progression of liver diseases. Chronic and sustained inflammation can lead to fibrosis, then cirrhosis and eventually end-stage cancer, hepatocellular carcinoma. Importantly, growing evidence suggest that metal exposure plays a role in hepatic disease pathogenesis. While in recent years, studies have linked metal exposure and hepatic steatosis, studies emphasizing metal-induced hepatic inflammation are limited. Hepatic inflammation is an important hallmark of fatty liver disease. This review aims to summarize the mechanisms of arsenic (As), cadmium (Cd) and chromium (Cr)-induced hepatic inflammation as they contribute to hepatic toxicity and to identify data gaps for future investigation.
Collapse
Affiliation(s)
| | - Koren K Mann
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada.
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada.
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Cote Ste Catherine Rd. Rm 202.1, Montréal, Québec, H3T 1E2, Canada.
| |
Collapse
|
9
|
Hassan FE, El-Mosallamy AEMK, Khalifa MM, Aljuaydi SH, Ali ME, Hosny SA, Bastawy N. Evaluating the potential impact of sodium-glucose cotransporter-2 inhibitor "canagliflozin" on the hepatic damage triggered by hypertension in rats. Microsc Res Tech 2024; 87:2929-2942. [PMID: 39051105 DOI: 10.1002/jemt.24665] [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: 01/03/2024] [Revised: 06/06/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Hypertension (HTN) is a prevalent chronic disease. HTN and liver disease association is extensively noted. Thus, finding a medication that can alleviate HTN and its accompanying liver insult would be promising. This study investigated the potential impacts of canagliflozin "sodium-glucose cotransporter-2 inhibitor" on the liver of the Nω-nitro-L-arginine methyl ester (L-NAME)-induced HTN rat model. Twenty-four adult male rats were divided into four groups; negative control group, canagliflozin group, L-NAME group: 50 mg/kg of L-NAME was injected daily for 5 weeks and L-NAME + canagliflozin group: 1 week after L-NAME injection both L-NAME + canagliflozin (40 mg/kg) were given concomitantly daily for further 4 weeks. Liver functions, serum lipid profile, hepatic oxidative/nitrative stress biomarkers, gene expression of lipogenic enzymes, B-cell lymphoma 2 (Bcl2), and DNA fragmentation, were measured. Besides, hepatic histology and immunohistochemistry of nuclear factor kappa B (NF-κB) and endothelial nitric oxide synthase (eNOS) were assessed. Canagliflozin improved hepatic lipogenesis via the downregulation of fatty acid synthase (FAS) and transcriptional regulatory element binding protein 1c (SREBP1c) genes leading to an improved serum lipid profile. Further, canagliflozin modified the eNOS/inducible nitric oxide synthase (iNOS) pathway and decreased the NF-κB immunoreactivity besides restoring the oxidants-antioxidants balance; increased reduced glutathione concomitant with declined malondialdehyde. This improvement of the liver was mirrored by the significant restoration of liver architecture and confirmed by the preserved liver DNA content and upregulation of the antiapoptotic Bcl2 mRNA level and attenuation of the alanine transaminase, aspartate aminotransferase. In conclusion, canagliflozin is a promising anti-hypertensive and hepatic-supportive medication. RESEARCH HIGHLIGHTS: Canagliflozin's antioxidant, anti-inflammatory, anti-lipogenic, and antiapoptotic characteristics mitigate remote liver compromise caused by hypertension. Canagliflozin can be exploited as a hepatoprotective and antihypertensive medication.
Collapse
Affiliation(s)
- Fatma E Hassan
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
- General Medicine Practice Program, Department of Physiology, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Aliaa E M K El-Mosallamy
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Center, Giza, Egypt
| | - Mohamed Mansour Khalifa
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
- Department of Human Physiology, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Samira H Aljuaydi
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Merhan E Ali
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Sara Adel Hosny
- Histology Department, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Nermeen Bastawy
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
10
|
Lubau NSA, Chengebroyen N, Subramaniyan V. Investigation of Uncovering Molecular Mechanisms of Alcohol-Induced Female Infertility-A Rational Approach. Reprod Sci 2024; 31:3660-3672. [PMID: 39485609 PMCID: PMC11611948 DOI: 10.1007/s43032-024-01692-8] [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: 04/11/2024] [Accepted: 09/06/2024] [Indexed: 11/03/2024]
Abstract
This study aimed to investigate the molecular mechanisms by which chronic alcohol consumption impacts female infertility, highlighting significant societal implications. By conducting a comprehensive literature review, we examined existing evidence on the association between long-term alcohol use and female reproductive health. Relevant studies were identified through systematic searches of electronic databases and key journals. We synthesized information on the molecular pathways affected by alcohol consumption, with particular emphasis on oxidative stress, inflammation, and hormonal disruptions. Additionally, we reviewed efforts to address alcohol-related health issues, including public health interventions, regulatory measures, and educational initiatives. Our study found strong evidence linking chronic alcohol consumption to increased mortality rates and a range of preventable diseases globally. Alcohol's effects extend beyond physiological consequences to psychological, social, and economic burdens. Chronic alcohol consumption disrupts hormonal balance and reproductive function, contributing to female infertility. Future research should focus on quantifying mortality risks associated with alcohol consumption, understanding gender-specific patterns in alcohol-related health outcomes, and elucidating the molecular mechanisms underlying female infertility. Addressing these gaps will inform strategies to mitigate the burden of alcohol-induced health issues and promote overall well-being. Collaborative efforts among diverse stakeholders are essential for advancing research agendas and translating findings into effective interventions.
Collapse
Affiliation(s)
- Natasha Sura Anak Lubau
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Neevashini Chengebroyen
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia.
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Jalan University, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
11
|
Gulzar F, Chhikara N, Kumar P, Ahmad S, Yadav S, Gayen JR, Tamrakar AK. ER stress aggravates NOD1-mediated inflammatory response leading to impaired nutrient metabolism in hepatoma cells. Biochem Biophys Res Commun 2024; 735:150827. [PMID: 39423570 DOI: 10.1016/j.bbrc.2024.150827] [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: 07/15/2024] [Revised: 09/23/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Nucleotide-binding Oligomerization Domain 1 (NOD1) is a cytosolic pattern recognition receptor that senses specific bacterial peptidoglycan moieties, leading to the induction of inflammatory response. Besides, sensing peptidoglycan, NOD1 has been reported to sense metabolic disturbances including the ER stress-induced unfolded protein response (UPR). However, the underpinning crosstalk between the NOD1 activating microbial ligands and the metabolic cues to alter metabolic response is not yet comprehensively defined. Here, we show that underlying ER stress aggravated peptidoglycan-induced NOD1-mediated inflammatory response in hepatoma cells. The HepG2 cells, undergoing ER stress induced by thapsigargin exhibited an amplified inflammatory response induced by peptidoglycan ligand of NOD1 (i.e. iE-DAP). This aggravated inflammatory response disrupted lipid and glucose metabolism, characterized by de novo lipogenic response, and increased gluconeogenesis in HepG2 cells. Further, we characterized that the aggravation of NOD1-induced inflammatory response was dependent on inositol-requiring enzyme 1-α (IRE1-α) and protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) activation, in conjunction with calcium flux. Altogether, our findings suggest that differential UPR activation makes liver cells more sensitive towards bacterial-derived ligands to pronounce inflammatory response in a NOD1-dependent manner that impairs hepatic nutrient metabolism.
Collapse
Affiliation(s)
- Farah Gulzar
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Nikita Chhikara
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, UP, India
| | - Pawan Kumar
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, UP, India
| | - Shadab Ahmad
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, UP, India
| | - Shubhi Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, UP, India
| | - Jiaur R Gayen
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, UP, India
| | - Akhilesh K Tamrakar
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, UP, India.
| |
Collapse
|
12
|
Bai Z, Li H, Jiao B. Potential Therapeutic Effect of Sinigrin on Diethylnitrosamine-Induced Liver Cancer in Mice: Exploring the Involvement of Nrf-2/HO-1, PI3K-Akt-mTOR Signaling Pathways, and Apoptosis. ACS OMEGA 2024; 9:46064-46073. [PMID: 39583716 PMCID: PMC11579720 DOI: 10.1021/acsomega.4c06203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/01/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024]
Abstract
Sinigrin is a glucosinolate present in plants of the family Brassicaceae and has been considered for its anticancer potential. This study examines the efficacy of sinigrin on the liver cancer caused by diethylnitrosamine (DEN) in mice through the analysis of its impact on the Nrf-2/HO-1, PI3K-Akt-mTOR, and apoptotic pathways. Development of liver cancer was induced by intraperitoneal injection at the age of 14 days with DEN (25 mg/kg) in mice. Thereafter, sinigrin was orally administered at doses of 10 and 20 mg/kg body weight per day the last 28 days. At the end of 10 weeks, mice were sacrificed and then we conducted hepatic biochemical and molecular assessments. Sinigrin reduced the serum level of alanine aminotransferase (ALT), aspartate aminotransferase (AST), gamma-glutamyl transferase (GGT), alkaline phosphatase (ALP), lactate dehydrogenase (LDH), alpha-fetoprotein (AFP), and bilirubin but increased total protein, and albumin, levels. Sinigrin increased the antioxidant enzymes (SOD, CAT, GPx, and GST) as indicated by reduced 8-OHdG, TBARS and increased glutathione. Sinigrin reduced the levels of inflammatory cytokines (IL-6, IL-1β, TNF-α, and NF-κB p65) and PI3K/AKT/mTOR signaling pathway. Sinigrin also activated the intrinsic mitochondrial apoptosis pathway mediated by p53, downregulated antiapoptotic proteins (Bcl-2), up-regulated pro-apoptosis regulatory proteins like Bax and caspase-3. All these results indicate that the protective effects of sinigrin against liver cancer are likely to be applied as an effective therapeutic agent through its antioxidant and pro-apoptotic activities.
Collapse
Affiliation(s)
- Zhe Bai
- Department
of Hepatobiliary Pancreatic and Gastrosurgery, Shanxi Province Cancer
Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital
Affiliated to Shanxi Medical University, Xinghualing District Workers New Street 3, Taiyuan 030013, China
| | - Hui Li
- Department
of Gastroenterology, The First Hospital
of Shanxi Medical University, No. 85, Jiefang South Road, Taiyuan, Shanxi 030001, China
| | - Baoping Jiao
- Department
of Hepatobiliary Pancreatic and Gastrosurgery, Shanxi Province Cancer
Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital
Affiliated to Shanxi Medical University, Xinghualing District Workers New Street 3, Taiyuan 030013, China
| |
Collapse
|
13
|
Yang Y, Chen Y, Feng D, Wu H, Long C, Zhang J, Wang J, Zhou B, Li S, Xiang S. Ficus hirta Vahl. ameliorates liver fibrosis by triggering hepatic stellate cell ferroptosis through GSH/GPX4 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118557. [PMID: 39009327 DOI: 10.1016/j.jep.2024.118557] [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: 03/07/2024] [Revised: 06/28/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ficus hirta Vahl., a traditional Chinese medicine commonly used in the Lingnan region, has been extensively used for liver disease treatment in China. Its notable antioxidant and anti-inflammatory properties have been reported in previous studies. However, its potential effect and underlying mechanism on liver fibrosis remains unclear. AIM OF STUDY This study was aimed to investigate the effect and its underlying mechanism of Ficus hirta Vahl on liver fibrosis in vitro and in vivo. MATERIALS AND METHODS The main components of Ficus hirta Vahl in blood were investigated by using UPLC-Q/TOF-MS/MS. Two animal models of liver fibrosis, the CCl4 and MCD induced mice, were used to assess the efficacy of Ficus hirta Vahl on liver fibrosis. Metabolomics was used to detect the level of metabolites in the serum of liver fibrosis mice after Ficus hirta Vahl treatment. Furthermore, the mechanism was validated in vitro using the human liver stellate cell line LX-2. The binding affinities of the active ingredients of Ficus hirta Vahl to the main targets of liver fibrosis were also determined. Finally, we identified the key active ingredients responsible for the treatment of liver fibrosis in vivo. RESULTS Fibrosis and inflammatory markers were significant down-regulation in both CCl4 and MCD induced liver fibrosis mice after Ficus hirta Vahl administration in a dose-dependent manner. We found that Ficus hirta Vahl may primarily exert its effect on liver fibrosis through the glutathione metabolic pathway. Importantly, the glutathione metabolic pathway is closely associated with ferroptosis, and our subsequent in vitro experiments provided evidence supporting this association. Ficus hirta Vahl was found to modulate the GSH/GPX4 pathway, ultimately leading to the amelioration of liver fibrosis. Moreover, using serum pharmacochemistry and molecular docking, we successfully identified apigenin as a probable efficacious monomer for the management of liver fibrosis and subsequently validated its efficacy in mice with CCl4-induced hepatic fibrosis. CONCLUSION Ficus hirta Vahl triggered the ferroptosis of hepatic stellate cell by regulating the GSH/GPX4 pathway, thereby alleviating liver fibrosis in mice. Moreover, apigenin is a key compound in Ficus hirta Vahl responsible for the effective treatment of liver fibrosis.
Collapse
Affiliation(s)
- Yuxuan Yang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China; School of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Yanchun Chen
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China; School of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Dongge Feng
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China; School of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Huixing Wu
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, PR China
| | - Changrui Long
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, PR China
| | - Jianping Zhang
- School of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Jinghao Wang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China
| | - Benjie Zhou
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, PR China; Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, 518107, PR China.
| | - Shasha Li
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China.
| | - Shijian Xiang
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, PR China; Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, 518107, PR China.
| |
Collapse
|
14
|
Miracle CE, McCallister CL, Egleton RD, Salisbury TB. Mechanisms by which obesity regulates inflammation and anti-tumor immunity in cancer. Biochem Biophys Res Commun 2024; 733:150437. [PMID: 39074412 PMCID: PMC11455618 DOI: 10.1016/j.bbrc.2024.150437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/12/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024]
Abstract
Obesity is associated with an increased risk for 13 different cancers. The increased risk for cancer in obesity is mediated by obesity-associated changes in the immune system. Obesity has distinct effects on different types of inflammation that are tied to tumorigenesis. For example, obesity promotes chronic inflammation in adipose tissue that is tumor-promoting in peripheral tissues. Conversely, obesity inhibits acute inflammation that rejects tumors. Obesity therefore promotes cancer by differentially regulating chronic versus acute inflammation. Given that obesity is chronic, the initial inflammation in adipose tissue will lead to systemic inflammation that could induce compensatory anti-inflammatory reactions in peripheral tissues to suppress chronic inflammation. The overall effect of obesity in peripheral tissues is therefore dependent on the duration and severity of obesity. Adipose tissue is a complex tissue that is composed of many cell types in addition to adipocytes. Further, adipose tissue cellularity is different at different anatomical sites throughout the body. Consequently, the sensitivity of adipose tissue to obesity is dependent on the anatomical location of the adipose depot. For example, obesity induces more inflammation in visceral than subcutaneous adipose tissue. Based on these studies, the mechanisms by which obesity promotes tumorigenesis are multifactorial and immune cell type-specific. The objective of our paper is to discuss the cellular mechanisms by which obesity promotes tumorigenesis by regulating distinct types of inflammation in adipose tissue and the tumor microenvironment.
Collapse
Affiliation(s)
- Cora E Miracle
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| | - Chelsea L McCallister
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| | - Richard D Egleton
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| | - Travis B Salisbury
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| |
Collapse
|
15
|
Selvarani R, Nguyen HM, Pazhanivel N, Raman M, Lee S, Wolf RF, Deepa SS, Richardson A. The role of inflammation induced by necroptosis in the development of fibrosis and liver cancer in novel knockin mouse models fed a western diet. GeroScience 2024:10.1007/s11357-024-01418-3. [PMID: 39514172 DOI: 10.1007/s11357-024-01418-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Non-resolving, chronic inflammation (inflammaging) is believed to play an important role in aging and age-related diseases. The goal of this study was to determine if inflammation induced by necroptosis arising from the liver plays a role in chronic liver disease (CLD) and liver cancer in mice fed a western diet (WD). Necroptosis was induced in liver using two knockin (KI) mouse models that overexpress genes involved in necroptosis (Ripk3 or Mlkl) specifically in liver (i.e., hRipk3-KI and hMlkl-KI mice). These mice and control mice (not overexpressing Ripk3 or Mlkl) were fed a WD (high in fat, sucrose, and cholesterol) starting at 2 months of age for 3, 6, and 12 months. Feeding the WD induced necroptosis in the control mice, which was further elevated in the hRipk3-KI and hMlkl-KI mice and was associated with a significant increase in inflammation in the livers of the hRipk3-KI and hMlkl-KI mice compared to control mice fed the WD. Overexpressing Ripk3 or Mlkl significantly increased steatosis and fibrosis compared to control mice fed the WD. Mice fed the WD for 12 months developed liver tumors (hepatocellular adenomas): 28% of the control mice developing tumors compared to 62% of the hRipk3-KI and hMlkl-KI mice. The hRipk3-KI and hMlkl-KI mice showed significantly more and larger tumor nodules. Our study provides the first direct evidence that inflammation induced by necroptosis arising from hepatocytes can lead to the progression of hepatic steatosis to fibrosis in obese mice that eventually results in an increased incidence in hepatocellular adenomas.
Collapse
Affiliation(s)
- Ramasamy Selvarani
- Biochemistry & Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Natesan Pazhanivel
- Department of Veterinary Pathology, TANUVAS, Chennai City, Tamilnadu, India
| | | | - Sunho Lee
- Biochemistry & Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Roman F Wolf
- Oklahoma Veteran Affairs Medical Center, Oklahoma City, OK, USA
| | - Sathyaseelan S Deepa
- Biochemistry & Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience & Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Arlan Richardson
- Biochemistry & Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Stephenson Cancer Center, Oklahoma City, OK, USA.
- Oklahoma Center for Geroscience & Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Veteran Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
16
|
Ojo OA, Adeyemo TR, Iyobhebhe M, Adams MD, Asaleye RM, Evbuomwan IO, Abdurrahman J, Maduakolam-Aniobi TC, Nwonuma CO, Odesanmi OE, Ojo AB. Beta vulgaris L. beetroot protects against iron-induced liver injury by restoring antioxidant pathways and regulating cellular functions. Sci Rep 2024; 14:25205. [PMID: 39448782 PMCID: PMC11502780 DOI: 10.1038/s41598-024-77503-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 10/23/2024] [Indexed: 10/26/2024] Open
Abstract
Beta vulgaris L. is a root vegetable that is consumed mainly as a food additive. This study aimed to describe the protective effect of B. vulgaris on Fe2+-mediated oxidative liver damage through in vitro, ex vivo, and in silico studies to establish a strong rationale for its protective effect. To induce oxidative damage, we incubated the livers of healthy male rats with 0.1 mM FeSO4 to induce oxidative injury and coincubated them with an aqueous extract of B. vulgaris root (BVFE) (15-240 µg/mL). Induction of liver damage significantly (p < .05) decreased the levels of GSH, SOD, CAT, and ENTPDase activities, with a corresponding increase in MDA and NO levels and Na+/K+ ATPase, G6 Pase, and F-1,6-BPase enzyme activities. BVFE treatment (p < .05) reduced these levels and activities to almost normal levels, with the most prominent effects observed at 240 µg/mL BVFE. An HPLC investigation revealed sixteen compounds in BVFE, with quercetin being the most abundant. Chlorogenic acid and iso-orientation showed the highest binding affinities for G6 Pase and Na+/K + ATPase, respectively. These findings suggest that B. vulgaris can protect against Fe2+-mediated liver damage by suppressing oxidative stress and cholinergic and purinergic activities while regulating gluconeogenesis. Overall, the hepatoprotective activity of this extract might be driven by the synergistic effect of the identified compounds and their probable interactions with target proteins.
Collapse
Affiliation(s)
- Oluwafemi Adeleke Ojo
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo, 232101, Nigeria.
| | | | | | - Moses Dele Adams
- Clinical Biochemistry, Phytopharmacology and Biochemical Toxicology Research Laboratory (CBPBT-RL), Department of Biochemistry, Baze University, Abuja, Nigeria
| | | | | | | | | | | | | | - Adebola Busola Ojo
- Department of Environmental Management and Toxicology, University of Ilesa, Ilesa, Nigeria
| |
Collapse
|
17
|
Zhang H, Zong R, Wu H, Jiang J, Liu C, Liu S. Transcription factor ASCL1 targets SLC6A13 to inhibit the progression of hepatocellular carcinoma via the glycine-inflammasome signaling. BIOMOLECULES & BIOMEDICINE 2024; 24:1606-1619. [PMID: 38780447 PMCID: PMC11496862 DOI: 10.17305/bb.2024.10328] [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: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024]
Abstract
Hepatocellular carcinoma (HCC), the most common primary liver cancer, typically arises from chronic liver conditions such as hepatitis, cirrhosis, or other chronic liver diseases, and is characterized by its aggressive nature and poor prognosis. The purpose of this research was to clarify the function of achaete-scute family bHLH transcription factor 1 (ASCL1) and solute carrier family 6 member 13 (SLC6A13) in influencing tumor cell behavior, inflammatory responses, and the regulation of inflammasomes. We analyzed the differentially expressed genes (DEGs) in the Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) database, as well as in the GSE14520 and GSE67764 datasets, to identify the expression changes of SLC6A13 in liver cancer. The prognostic significance of SLC6A13 in LIHC was assessed through Kaplan-Meier survival curve analysis. Transcriptional regulation of SLC6A13 by ASCL1 was explored using the Joint Annotation of the Human Genome and other species by the Systematic Pipeline for the Annotation of Regulatory Regions (JASPAR) database and dual-luciferase assays. In vitro experiments investigated the impact of ASCL1 and SLC6A13 overexpression on HCC cell growth. Additionally, the effects of ethanol treatment and glycine modulation on the inflammatory response in HCC cell lines were evaluated. HCC samples showed reduced levels of SLC6A13, which correlates with a better prognosis for liver metastases. Elevated SLC6A13 expression correlated with improved overall survival (OS), progression-free survival (PFS), recurrence-free survival (RFS), and disease-specific survival (DSS). ASCL1 upregulated SLC6A13 and inhibited proliferation, migration, and invasion of HCC cells. Ethanol induced the production of inflammatory cytokines, which was enhanced by overexpression of SLC6A13 but counteracted by glycine. This study highlighted elevated expression of SLC6A13 in LIHC which has been correlated with improved OS, PFS, RFS, and DSS. Overexpression of SLC6A13 and ASCL1 in HCC cells enhanced inflammasome activation, which was exacerbated by ethanol and attenuated by glycine.
Collapse
|