1
|
Cheng S, Xiao X, Wang D, Wang X, Yang M. Lactate and lactylation in liver diseases: energy metabolism, inflammatory immunity and tumor microenvironment. Front Immunol 2025; 16:1581582. [PMID: 40421024 PMCID: PMC12104064 DOI: 10.3389/fimmu.2025.1581582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 04/21/2025] [Indexed: 05/28/2025] Open
Abstract
Liver diseases pose a significant threat to human health. Lactate, a byproduct of glycolysis, serves various biological functions, including acting as an energy source, a signaling molecule, and a substrate for lactylation. Lactylation is a novel lactate-dependent post-translational modification that plays a role in tumor proliferation, the regulation of immune cell function, and the modulation of gene expression. In this paper, we summarize the roles of lactate and lactylation in energy metabolism, inflammatory immunity, and the tumor microenvironment, while also elucidating recent research advancements regarding lactate and lactylation in the context of hepatic fibrosis, non-alcoholic fatty liver disease, and hepatocellular carcinoma. Furthermore, lactate and lactylation are proposed as promising new targets for the treatment of liver diseases.
Collapse
Affiliation(s)
| | | | | | | | - Minlan Yang
- School of Medicine, Yangtze University, Jingzhou, China
| |
Collapse
|
2
|
Cheng X, Kang L, Liu J, Wang Q, Zhang Z, Zhang L, Xie Y, Chang L, Zeng D, Tian L, Zhang L, Xu P, Li Y. Proteomics and phosphoproteomics revealed dysregulated kinases and potential therapy for liver fibrosis. Mol Cell Proteomics 2025:100991. [PMID: 40368138 DOI: 10.1016/j.mcpro.2025.100991] [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: 08/23/2024] [Revised: 04/30/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025] Open
Abstract
Liver fibrosis is the initial stage of most liver diseases, and it is also a pathological process involving the liver in the late stages of many metabolic diseases. Therefore, it is important to systematically understand the pathological mechanism of liver fibrosis and seek therapeutic approaches for intervention and treatment of liver fibrosis. Disordered proteins and their post-translational modifications, such as phosphorylation, play vital roles in the occurrence and development of liver fibrosis. However, the regulatory mechanisms that govern this process remain poorly understood. In this study, we analyzed and quantified the liver proteome and phosphoproteome of CCl4-induced early liver fibrosis model in mice. Proteomic analysis revealed that the pathways involved in extracellular matrix (ECM) recombination, collagen formation, metabolism and other related disorders, and protein phosphorylation modification pathways were also significantly enriched. In addition, western blotting and phosphoproteomics demonstrated that phosphorylation levels were elevated in the context of liver fibrosis. A total of 13,152 phosphosites were identified, with 952 sites increased while only 156 ones decreased. Furthermore, the upregulated phosphorylation sites, which exhibited no change at the proteome level mainly shared a common [xxxSPxxx] motif. Consequently, the kinases-substrates analysis ascertained the overactive kinases of these up-regulated substrates, which ultimately led to the identification of 13 significantly altered kinases within this dataset. These kinases were mainly catalogued into the STE, CMGC, and CAMK kinase families. Among them, STK4, GSK3α and CDK11B were subsequently validated though cellular and animal experiments, and the results demonstrated that their inhibitors could effectively reduce the activation of hepatic stellate cells and ECM production. These kinases may represent potential therapeutic targets for liver fibrosis, and their inhibitors may serve as promising anti- hepatic fibrosis drugs.
Collapse
Affiliation(s)
- Xinyu Cheng
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Li Kang
- School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Jinfang Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China; TaiKang Medical School (School of Basic Medical Sciences), Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, P. R. China
| | - Qingye Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Zhenpeng Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Li Zhang
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Yuping Xie
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Lei Chang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Daobing Zeng
- General Surgery Department, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Lantian Tian
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, P. R. China
| | - Lingqiang Zhang
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China.
| | - Ping Xu
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China; School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China; College of Life Sciences, Hebei University, 071002 Baoding, China; TaiKang Medical School (School of Basic Medical Sciences), Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, P. R. China.
| | - Yanchang Li
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China; College of Life Sciences, Hebei University, 071002 Baoding, China.
| |
Collapse
|
3
|
Wang X, Li M, Liu X, Sun G, Zhang D, Sun L, Yin Y, Zhang W, Hao J. Isolation of Murine Pancreatic Stellate Cells and the Establishment of a New ex-vivo Activation Model. Clin Exp Gastroenterol 2025; 18:79-89. [PMID: 40357130 PMCID: PMC12067667 DOI: 10.2147/ceg.s507384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Background Pancreatic stellate cells (PSCs) are critical in the development of pancreatic fibrosis. In vitro, cell attachment itself can promote cell activation. Currently, there is a lack of methods for isolating activated PSCs that are unaffected by cell attachment. This study aims to identify effective methods for isolating quiescent and activated murine PSCs (mPSCs) and to evaluate the potential of caerulein in inducing mPSC activation in an ex vivo model. Methods Pancreatic tissue from mice was digested with collagenase P (1.17 U/mL), Pronase (0.5 mg/mL), and DNase I (0.01 mg/mL). Quiescent and activated mPSCs were isolated using a Nycodenz gradient. Immunostaining for α-smooth muscle actin (α-SMA), Desmin, glial fibrillary acidic protein (GFAP), vimentin, CK19, and CD68 was performed to confirm cell purity. Real-time quantitative PCR (RT-PCR) and RNA sequencing assessed the activation phenotype following caerulein treatment. Results Quiescent and activated mPSCs were successfully isolated using the Nycodenz gradient, with cells exhibiting typical stellate morphology and positive staining for α-SMA, Desmin and vimentin. Oil Red O staining confirmed lipid droplets in quiescent mPSCs. In the caerulein-treated group, mPSC activation was significantly greater than in the saline-treated control group. RT-PCR revealed progressive upregulation of acta2 (**p<0.01, d4 compared to d2, ## p<0.01,d7 compared to d4,**p<0.01,d7 compared to d2), col1a (**p<0.01, d4 compared to d2,**p<0.01,d7 compared to d2), and actg2 (**p<0.01, d4 compared to d2, ## p<0.01,d7 compared to d4, **p<0.01,d7 compared to d2) mRNA levels at 2, 4, and 7 days post-adhesion. Fibroblast markers were also upregulated, and KEGG and GO enrichment analyses identified key pathways involved in ECM-receptor interactions, cell cycle regulation, PI3K-Akt signaling, and extracellular matrix remodeling. Conclusion The Nycodenz gradient efficiently isolates quiescent mPSCs, and short-term caerulein treatment effectively activates mPSCs ex vivo, providing a valuable model for studying mPSC activation and related signaling pathways.
Collapse
Affiliation(s)
- Xinye Wang
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
| | - Miaomiao Li
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
| | - Xinjuan Liu
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
| | - Guangyong Sun
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
| | - Dong Zhang
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
| | - Lijun Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, People’s Republic of China
| | - Yue Yin
- Department of Pharmacology, School of Basic Medical Sciences, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, People’s Republic of China
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, People’s Republic of China
| | - Jianyu Hao
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
| |
Collapse
|
4
|
Pang J, Xu D, Zhang X, Qu J, Jiang J, Suo J, Li T, Li Y, Peng Z. TIMP2-mediated mitochondrial fragmentation and glycolytic reprogramming drive renal fibrogenesis following ischemia-reperfusion injury. Free Radic Biol Med 2025; 232:244-259. [PMID: 39986488 DOI: 10.1016/j.freeradbiomed.2025.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
Acute kidney injury (AKI) triggers renal structural and functional abnormalities through inflammatory and fibrotic signaling pathways, ultimately progressing to chronic kidney disease (CKD). The mechanisms underlying AKI-to-CKD transition are complex, with hypoxia, mitochondrial dysfunction, and metabolic reprogramming as critical contributors. Public data analysis demonstrated significant upregulation of tissue inhibitors of metalloproteinases (Timp2) in renal biopsy tissues of CKD patients. In both ischemia/reperfusion (I/R) and unilateral ureteral obstruction (UUO) models, Timp2 upregulation was observed. Tubule-specific Timp2 knockout markedly attenuated renal fibrosis. RNA-sequencing revealed Timp2's association with mitochondrial dynamics and glycolysis in I/R mice. Timp2 deletion improved mitochondrial morphology and suppressed glycolytic enzyme expression. In vitro, TGF-β1-treated Timp2-knockdown HK-2 cells exhibited inhibited Drp1 expression, restored Mfn2 levels, alleviated mitochondrial fragmentation, and elevated mitochondrial membrane potential. Additionally, Pfkfb3 and HIF-1α were downregulated, accompanied by reduced extracellular acidification rate (ECAR), PFK activity, and lactate production. Mechanistically, Timp2 interacts with the extracellular domain of Sdc4 in an autocrine manner, activating the Hedgehog (Hh) signaling pathway. Cyclopamine partially rescued Timp2 overexpression-induced mitochondrial dysfunction, suppressed Pfkfb3-mediated glycolysis, and diminished collagen deposition. This study is the first to demonstrate that Timp2 in TECs exacerbates Hh signaling, promoting mitochondrial fragmentation and metabolic reprogramming to accelerate I/R-induced renal fibrosis.
Collapse
Affiliation(s)
- Jingjing Pang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Dongxue Xu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China.
| | - Xiaoyu Zhang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Jiacheng Qu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Jun Jiang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Jinmeng Suo
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Tianlong Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Yiming Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China.
| | - Zhiyong Peng
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Intensive Care Unit of the Second Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China.
| |
Collapse
|
5
|
Roh YJ, Kim H, Choi DW. Metabolic Sparks in the Liver: Metabolic and Epigenetic Reprogramming in Hepatic Stellate Cells Activation and Its Implications for Human Metabolic Diseases. Diabetes Metab J 2025; 49:368-385. [PMID: 40367987 PMCID: PMC12086559 DOI: 10.4093/dmj.2025.0195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2025] [Accepted: 04/17/2025] [Indexed: 05/16/2025] Open
Abstract
The liver plays a fundamental role in metabolic homeostasis, integrating systemic fuel utilization with the progression of various metabolic diseases. Hepatic stellate cells (HSCs) are a key nonparenchymal cell type in the liver, which is essential for maintaining hepatic architecture in their quiescent state. However, upon chronic liver injury or metabolic stress, HSCs become activated, leading to excessive extracellular matrix deposition and pro-fibrotic signaling, ultimately positioning them as key players in liver pathology. Emerging evidence highlights the critical roles of metabolic reprogramming and epigenetic regulation in HSCs activation. HSCs activation is driven by both intrinsic fuel metabolism reprogramming and extrinsic metabolic cues from the microenvironment, while the metabolic intermediates actively reshape the epigenetic landscape, reinforcing fibrogenic transcriptional programs. In this review, we summarize recent advances in understanding how metabolic and epigenetic alterations drive HSCs activation, thereby shaping transcriptional programs that sustain fibrosis, and discuss potential therapeutic strategies to target these interconnected pathways in human metabolic diseases.
Collapse
Affiliation(s)
- Yeon Jin Roh
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Hyeonki Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Dong Wook Choi
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| |
Collapse
|
6
|
Xie SS, Hou R, Gao L, Yang Q, Li W, Dong ZH, Dong YH, Li SJ, Ma WX, Gao YY, Xu L, Li C, Chen Y, Yu JT, Wang JN, Ji ML, He RB, Suo XG, Liu MM, Jin J, Wen JG, Yang C, Meng XM. IGF-Binding Protein 7 and Cadmium-Induced Hepatorenal Fibrosis. J Am Soc Nephrol 2025:00001751-990000000-00624. [PMID: 40208692 DOI: 10.1681/asn.0000000698] [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: 11/07/2024] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
Key Points
IGF-binding protein 7 (IGFBP7) expression was elevated in kidney and liver tissues of mice subjected to chronic cadmium exposure.IGFBP7 deficiency protected against cadmium-induced hepatorenal dysfunction and fibrosis.Inhibition of the IGFBP7/α-enolase/H3K18la axis may be a potential therapeutic intervention for cadmium-induced hepatorenal fibrosis.
Background
Chronic cadmium exposure can induce the onset and progression of hepatorenal fibrosis; however, its molecular basis is unclear. IGF-binding protein 7 (IGFBP7) is not only a biomarker of AKI but also plays a functional role in promoting kidney injury and inflammation. Abnormal repair of AKI causes kidney fibrosis and CKD. IGFBP7 has also been reported as a more sensitive biomarker for liver fibrosis. However, its role in hepatorenal fibrosis requires further investigation.
Methods
IGFBP7 global and conditional knockout mice were used to determine the role of IGFBP7 in cadmium-induced hepatorenal fibrosis. Then, liquid chromatography–mass spectrometry, truncated mutants, coimmunoprecipitation, and microscale thermophoresis were used to unravel the downstream mechanisms.
Results
IGFBP7 expression was significantly elevated in kidney and liver tissues of mice subjected to chronic cadmium exposure. IGFBP7 deficiency attenuated cadmium-induced hepatorenal dysfunction and fibrosis, whereas restoration of IGFBP7 expression in IGFBP7-deficient mice reproduced hepatorenal fibrosis. Mechanistically, IGFBP7 interacted with α-enolase (ENO1) and inhibited its ubiquitination and degradation. Upregulated ENO1 further promoted glucose metabolic reprogramming and lactate accumulation. Conversely, lactate accumulation enhanced IGFBP7 transcription and expression through histone H3K18 lactylation. Importantly, therapy targeting IGFBP7 significantly ameliorated cadmium-induced hepatorenal fibrosis.
Conclusions
IGFBP7 promoted cadmium-induced hepatorenal fibrosis by enhancing ENO1-driven abnormal glycolysis and lactate accumulation.
Collapse
Affiliation(s)
- Shuai-Shuai Xie
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Rui Hou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Li Gao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Qin Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Ze-Hui Dong
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Yu-Hang Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Shuang-Jian Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Wen-Xian Ma
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Ying-Ying Gao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Long Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Chao Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Ying Chen
- Anhui Provincial Chest Hospital, Hefei, China
| | - Ju-Tao Yu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Jia-Nan Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Ming-Lu Ji
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Ruo-Bing He
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Xiao-Guo Suo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Ming-Ming Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Juan Jin
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jia-Gen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Chen Yang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| |
Collapse
|
7
|
Xiang X, Shao Y, Xiang L, Jiao Q, Zhang W, Qin Y, Chen Y. Suppression of Liver Fibrogenesis with Photothermal Sorafenib Nanovesicles via Selectively Inhibiting Glycolysis and Amplification of Active HSCs. Mol Pharm 2025; 22:1939-1957. [PMID: 40053386 DOI: 10.1021/acs.molpharmaceut.4c01135] [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] [Indexed: 04/08/2025]
Abstract
As the major driving factor of hepatic fibrosis, the activated hepatic stellate cells (aHSCs) rely on active glycolysis to support their aberrant proliferation and secretion of the extracellular matrix. Sorafenib (Sor) can combat liver fibrosis by suppressing HIF-1α and glycolysis, but its poor solubility, rapid metabolism, and low bioavailability restrict such a clinical application. Here, Sor was loaded onto polydopamine nanoparticles and then encapsulated by a retinoid-decorated red blood cell membrane, yielding HSC-targeted Sor nanovesicles (PDA/Sor@RMV-VA) with a high Sor-loading capacity and photothermally controlled drug release for antifibrotic treatment. These Sor RMVs not only exhibited a good particle size, dispersity and biocompatibility, prolonged circulation time, enhanced aHSC targetability, and hepatic accumulation both in vitro and in vivo, but also displayed a mild photothermal activity proper for promoting sorafenib release and accumulation in CCl4-induced fibrotic mouse livers without incurring phototoxicity. Compared with nontargeting Sor formulations, PDA/Sor@RMV-VA more effectively downregulated HIF-1α and glycolytic enzyme in both cultured aHSCs and fibrotic mice and reversed myofibroblast phenotype and amplification of aHSCs and thus more significantly improved liver damage, inflammation, and fibrosis, all of which could be even further advanced with NIR irradiation. These results fully demonstrate the antifibrotic power and therapeutic potential of PDA/Sor@RMV-VA as an antifibrotic nanomedicine, which would support a new clinical treatment for hepatic fibrosis.
Collapse
Affiliation(s)
- Xianjing Xiang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yaru Shao
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Li Xiang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| | - Qiangqiang Jiao
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Wenhui Zhang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yuting Qin
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yuping Chen
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
- MOE Key Laboratory of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| |
Collapse
|
8
|
Herrera-Sánchez MP, Rodríguez-Hernández R, Rondón-Barragán IS. Comparative Transcriptome Analysis of Hens' Livers in Conventional Cage vs. Cage-Free Egg Production Systems. Vet Med Int 2025; 2025:3041254. [PMID: 40160973 PMCID: PMC11952924 DOI: 10.1155/vmi/3041254] [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: 10/31/2024] [Accepted: 02/22/2025] [Indexed: 04/02/2025] Open
Abstract
Different conditions of production systems including stocking density, thermal conditions, and behavior restriction can have a significant detrimental effect on the health and performance of laying hens. The conventional cage system is one of the systems that have been reported to cause stress problems in birds, due to social and behavioral stress. Emerging technologies have facilitated a deeper understanding of animal responses to various scenarios and can be an additional tool to conventional ones to assess animal welfare, where transcriptomic analysis has the potential to show the genetic changes that occur in response to stress. According to this, the aim of this work was to characterize the liver transcriptome of hens housed under two egg production systems (conventional cage and cage-free). Liver tissue from Hy-Line Brown hens housed in conventional cage (n = 3) and cage-free (n = 3) production systems at week 80 of age was processed using the Illumina platform to identify differentially expressed genes with a padj < 0.05. Regarding the differentially expressed genes, 138 genes were found, of which 81 were upregulated and 57 downregulated. Some of the genes of interest were TENM2, GRIN2C, and ACACB, which would indicate greater fat synthesis in the liver of caged hens. The enriched KEGG pathways were DNA replication and the cell cycle. In conclusion, it was identified that the cage production system may influence DNA replication and the cell cycle since the genes related to these terms were found suppressed, which would indicate cellular instability.
Collapse
Affiliation(s)
- María Paula Herrera-Sánchez
- Poultry Research Group, Laboratory of Immunology and Molecular Biology, Faculty of Veterinary Medicine and Zootechnics, Universidad del Tolima, Altos de Santa Helena, Ibagué 730006299, Tolima, Colombia
- Immunobiology and Pathogenesis Research Group, Laboratory of Immunology and Molecular Biology, Faculty of Veterinary Medicine and Zootechnics, Universidad del Tolima, Altos de Santa Helena, Ibagué 730006299, Tolima, Colombia
| | - Roy Rodríguez-Hernández
- Poultry Research Group, Laboratory of Immunology and Molecular Biology, Faculty of Veterinary Medicine and Zootechnics, Universidad del Tolima, Altos de Santa Helena, Ibagué 730006299, Tolima, Colombia
| | - Iang Schroniltgen Rondón-Barragán
- Poultry Research Group, Laboratory of Immunology and Molecular Biology, Faculty of Veterinary Medicine and Zootechnics, Universidad del Tolima, Altos de Santa Helena, Ibagué 730006299, Tolima, Colombia
- Immunobiology and Pathogenesis Research Group, Laboratory of Immunology and Molecular Biology, Faculty of Veterinary Medicine and Zootechnics, Universidad del Tolima, Altos de Santa Helena, Ibagué 730006299, Tolima, Colombia
| |
Collapse
|
9
|
Peng TY, Lu JM, Zheng XL, Zeng C, He YH. The role of lactate metabolism and lactylation in pulmonary arterial hypertension. Respir Res 2025; 26:99. [PMID: 40075458 PMCID: PMC11905457 DOI: 10.1186/s12931-025-03163-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex and progressive disease characterized by elevated pulmonary artery pressure and vascular remodeling. Recent studies have underscored the pivotal role of metabolic dysregulation and epigenetic modifications in the pathogenesis of PAH. Lactate, a byproduct of glycolysis, is now recognized as a key molecule that links cellular metabolism with activity regulation. Recent findings indicate that, in addition to altered glycolytic activity and dysregulated. Lactate homeostasis and lactylation-a novel epigenetic modification-also play a significant role in the development of PAH. This review synthesizes current knowledge regarding the relationship between altered glycolytic activity and PAH, with a particular focus on the cumulative effects of lactate in pulmonary vascular cells. Furthermore, lactylation, an emerging epigenetic modification, is discussed in the context of PAH. By elucidating the complex interplay between lactate metabolism and lactylation in PAH, this review aims to provide insights into potential therapeutic targets. Understanding these metabolic pathways may lead to innovative strategies for managing PAH and improving patient outcomes. Future research should focus on the underlying mechanisms through which lactylation influences the pathophysiology of PAH, thereby aiding in the development of targeted interventions.
Collapse
Affiliation(s)
- Tong-Yu Peng
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jun-Mi Lu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xia-Lei Zheng
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Cheng Zeng
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yu-Hu He
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
10
|
Delgado ME, Naranjo-Suarez S, Ramírez-Pedraza M, Cárdenas BI, Gallardo-Martínez C, Balvey A, Belloc E, Martín J, Boyle M, Méndez R, Fernandez M. CPEB4 modulates liver cancer progression by translationally regulating hepcidin expression and sensitivity to ferroptosis. JHEP Rep 2025; 7:101296. [PMID: 39980747 PMCID: PMC11840500 DOI: 10.1016/j.jhepr.2024.101296] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 02/22/2025] Open
Abstract
Background & Aims Liver cancer is a significant global health issue, with its incidence rising in parallel with the obesity epidemic. The limited therapeutic options available emphasize the need for a better understanding of the molecular pathways involved in its pathogenesis. While much of the previous research has focused on transcriptional changes, this study examines translational alterations, specifically the role of cytoplasmic polyadenylation element binding protein 4 (CPEB4), a key regulator of translation. Methods We analyzed publicly available patient databases and conducted studies using human and mouse liver cancer cells, xenograft and allograft models, mouse models of high-fat diet-related liver cancer, and CPEB4 knockout and knockdown mice and cell lines. Results Patient data analysis (n = 87) showed a strong correlation between low CPEB4 levels and reduced survival rates (p <0.001). In mouse models of diet-induced liver cancer (n = 10-15 per group), both systemic and hepatocyte-specific CPEB4 knockout mice exhibited significantly increased tumor burden compared with wild-type controls (p <0.05). In vitro studies using human and murine liver cancer cells (n = 3 biological replicates) demonstrated reduced sensitivity to ferroptosis upon CPEB4 depletion when induced by erastin or RSL3 (p <0.01). Mechanistically, CPEB4 deficiency suppressed hepcidin expression, leading to elevated ferroportin levels, decreased intracellular iron accumulation, and reduced lipid peroxidation (p <0.05). Conclusions This study uncovers a novel CPEB4-dependent mechanism linking translational control to liver cancer progression and ferroptosis regulation. Therapeutic strategies targeting CPEB4-mediated pathways hold promise for advancing treatment options in liver cancer. Impact and implications This study addresses the pressing need for improved therapies in liver cancer, particularly given its increasing prevalence linked to obesity and metabolic-associated fatty liver disease. By uncovering the role of the RNA-binding protein cytoplasmic polyadenylation element binding protein 4 (CPEB4) in modulating iron regulation and cancer cell sensitivity to ferroptosis, our research highlights a new translational mechanism with potential therapeutic relevance. These findings are particularly significant for clinicians, researchers, and policymakers focused on advancing targeted treatments for hepatocellular carcinoma. If further validated in human clinical studies, targeting CPEB4-mediated pathways could help develop treatments that enhance cancer cell susceptibility to ferroptosis, offering a promising strategy for improving outcomes in patients with advanced liver cancer. Limitations of the study include the need for further clinical validation to confirm these preclinical findings in human disease contexts.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eulalia Belloc
- Institute for Research in Biomedicine (IRB), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Judit Martín
- Institute for Research in Biomedicine (IRB), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mark Boyle
- FRCB-IDIBAPS Biomedical Research Institute; Barcelona, Spain
| | - Raúl Méndez
- Institute for Research in Biomedicine (IRB), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA); Barcelona, Spain
| | | |
Collapse
|
11
|
Li X, Wang J, Chen Y, Li P, Wen H, Xu X, Wang J, Xu Y, Chen Y, Song J, Lu W, Zhu D, Fu X. Estrogen Oppositely Regulates Pulmonary Hypertension via METTL3/PFKFB3 under Normoxia and Hypoxia. Am J Respir Cell Mol Biol 2025; 72:258-271. [PMID: 39265182 DOI: 10.1165/rcmb.2024-0042oc] [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: 01/08/2024] [Accepted: 09/12/2024] [Indexed: 09/14/2024] Open
Abstract
Despite extensive investigation into estrogen's role in pulmonary hypertension (PH) development, its effects, whether beneficial or detrimental, remain contentious. This study aimed to elucidate estrogen's potential role in PH under normoxic and hypoxic conditions. Using norfenfluramine- and hypoxia-induced rat models of PH, the study evaluated the impact of 17β-estradiol (E2) on PH progression. E2 promoted PH development under normoxia while providing protection under hypoxia. Mechanistically, under normoxia, E2 upregulated METTL3 (methyltransferase-like 3) gene transcription and protein via an estrogen response element-dependent pathway, which in turn increased the N6-methyladenosine methylation and translational efficiency of PFKFB3 (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoform 3) mRNA, leading to increased PFKFB3 protein levels and enhanced proliferation and migration of pulmonary artery smooth muscle cells. Conversely, under hypoxia, E2 downregulated METTL3 transcription through a hypoxia response element-dependent mechanism driven by increased HIF-1α (hypoxia-inducible factor 1α) levels, resulting in reduced PFKFB3 protein expression and diminished pulmonary artery smooth muscle cell proliferation and migration. METTL3 and PFKFB3 proteins are upregulated in the pulmonary arteries of patients with pulmonary arterial hypertension. Collectively, these findings suggest that E2 exerts differential effects on PH progression via dual regulation of the METTL3/PFKFB3 protein under normoxic and hypoxic conditions, positioning the METTL3/PFKFB3 protein as a potential therapeutic target for PH treatment.
Collapse
Affiliation(s)
- Xiaosa Li
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiale Wang
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuqin Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; and
| | - Ping Li
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hao Wen
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xingyan Xu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; and
| | - Yiming Xu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yingying Chen
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; and
| | - Dongxing Zhu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaodong Fu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
12
|
Xie L, Song D, Ouyang Z, Ning Y, Liu X, Li L, Xia W, Yang Y. USP27 promotes glycolysis and hepatocellular carcinoma progression by stabilizing PFKFB3 through deubiquitination. Cell Signal 2025; 127:111585. [PMID: 39746496 DOI: 10.1016/j.cellsig.2024.111585] [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: 11/10/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Hepatocellular carcinoma (HCC) is associated with a dismal prognosis, primarily due to its high rates of metastasis and recurrence. Metabolic reprogramming, specifically enhanced glycolysis, is a prominent feature of cancer progression. This study identifies ubiquitin-specific peptidase 27 X-linked (USP27) as a significant regulator of glycolysis in HCC. We demonstrate that USP27 stabilizes PFKFB3, a key glycolytic enzyme, through deubiquitination, thereby increasing glycolytic activity and facilitating tumor progression. Furthermore, we reveal that CTCF, a well-known transcription factor, directly binds to the USP27 promoter and upregulates its expression, thereby establishing a connection between transcriptional regulation and metabolic reprogramming in HCC. Knockdown of USP27 or CTCF in HCC cells considerably decreased glycolysis and proliferation, while overexpression had the opposite effect. In vivo studies confirmed that USP27 knockdown suppresses HCC growth and metastasis. Our findings establish the CTCF/USP27/PFKFB3 axis as a novel mechanism driving HCC progression through glycolysis, indicating that targeting this pathway could offer new therapeutic opportunities. These results provide valuable insights into the molecular mechanisms underlying HCC and emphasize the potential of targeting USP27-mediated metabolic pathways as a strategy for cancer treatment.
Collapse
Affiliation(s)
- Longhui Xie
- Department of Hepatobiliary Pancreatic Spleen Surgery, The Central Hospital of Yongzhou, Yongzhou 425000, PR China
| | - Dekun Song
- Department of Hepatobiliary Surgery, Binzhou People's Hospital, Binzhou 256600, PR China
| | - Zhengsheng Ouyang
- Department of Hepatobiliary Pancreatic Spleen Surgery, The Central Hospital of Yongzhou, Yongzhou 425000, PR China; Department of clinical medicine, YongZhou Vocational Technical College, Yongzhou 425000, PR China
| | - Yinkuan Ning
- Department of Interventional Vascular Surgery, The Central Hospital of Shaoyang, Shaoyang 422000, PR China
| | - Xintao Liu
- Department of Hepatobiliary Pancreatic Spleen Surgery, The Central Hospital of Yongzhou, Yongzhou 425000, PR China
| | - Lai Li
- Department of Hepatobiliary Pancreatic Spleen Surgery, The Central Hospital of Yongzhou, Yongzhou 425000, PR China
| | - Wangning Xia
- Department of Hepatobiliary Pancreatic Spleen Surgery, The Central Hospital of Yongzhou, Yongzhou 425000, PR China
| | - Yang Yang
- Department of Oncology, The Central Hospital of Shaoyang, Shaoyang 422000, PR China.
| |
Collapse
|
13
|
He J, Li X, Yu H, Xu C, Tian R, Zhou P, Yin Z. Inflammation-induced PFKFB3-mediated glycolysis promoting myometrium contraction through the PI3K-Akt-mTOR pathway in preterm birth mice. Am J Physiol Cell Physiol 2025; 328:C895-C907. [PMID: 39907705 DOI: 10.1152/ajpcell.00704.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/21/2024] [Accepted: 01/06/2025] [Indexed: 02/06/2025]
Abstract
Inflammation is a significant risk factor for preterm birth. Inflammation enhances glycolytic processes in various cell types and contributes to the development of myometrial contractions. However, the potential of inflammation to activate glycolysis in pregnant murine uterine smooth muscle cells (mUSMCs) and its role in promoting inflammatory preterm birth remain unexplored. In this study, lipopolysaccharide was employed to establish both cell and animal inflammation models. We found that inflammation of mUSMCs during late pregnancy could initiate glycolysis and promote cell contraction. Subsequently, the inhibition of glycolysis using the glycolysis inhibitor 2-deoxyglucose can reverse inflammation-induced cell contraction. The expression of 6-phosphofructokinase 2 kinase (PFKFB3) was significantly upregulated in mUSMCs following lipopolysaccharide stimulation. In addition, lactate accumulation and enhanced contraction were observed. Inhibition of PFKFB3 reversed the lactate accumulation and enhanced contraction induced by inflammation. We also found that inflammation activated the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt)-mammalian target of the rapamycin (mTOR) pathway, leading to the upregulation of PFKFB3 expression. The PI3K-Akt pathway inhibitor LY294002 and the mTOR pathway inhibitor rapamycin effectively inhibited the upregulation of PFKFB3 protein expression, lactate production, and the enhancement of cell contraction induced by lipopolysaccharide. This study indicates that inflammation regulates PFKFB3 through the PI3K-Akt-mTOR pathway, which enhances the glycolytic process in pregnant mUSMCs, ultimately leading to myometrial contraction.NEW & NOTEWORTHY Expression of PFKFB3, a key enzyme in glycolysis, was significantly upregulated both in the mUSMCs and myometrium of mice during late pregnancy after lipopolysaccharide stimulation. Activation of the PI3K-Akt-mTOR pathway enhanced PFKFB3 expression, which is involved in the initiation of glycolysis. Inflammation-activated PFKFB3 via the PI3K-Akt-mTOR pathway, which enhances the cellular glycolytic process and thus promotes myometrium contraction in pregnancy.
Collapse
Affiliation(s)
- Jing He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Department of Obstetrics and Gynecology, Anqing Medical Center of Anhui Medical University, Anqing, People's Republic of China
| | - Xuan Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei, People's Republic of China
| | - Huihui Yu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei, People's Republic of China
| | - Chenyi Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei, People's Republic of China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, People's Republic of China
| | - Ruixian Tian
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei, People's Republic of China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, People's Republic of China
| | - Ping Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei, People's Republic of China
| | - Zongzhi Yin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei, People's Republic of China
- Center for Big Data and Population Health of IHM, Hefei, People's Republic of China
| |
Collapse
|
14
|
Fernandez M, Mendez R. Cytoplasmic regulation of the poly(A) tail length as a potential therapeutic target. RNA (NEW YORK, N.Y.) 2025; 31:402-415. [PMID: 39805658 PMCID: PMC11874964 DOI: 10.1261/rna.080333.124] [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: 11/21/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025]
Abstract
Virtually all mRNAs acquire a poly(A) tail cotranscriptionally, but its length is dynamically regulated in the cytoplasm in a transcript-specific manner. The length of the poly(A) tail plays a crucial role in determining mRNA translation, stability, and localization. This dynamic regulation of poly(A) tail length is widely used to create posttranscriptional gene expression programs, allowing for precise temporal and spatial control. Dysregulation of poly(A) tail length has been linked to various diseases, including cancers, inflammatory and cardiovascular disorders, and neurological syndromes. Cytoplasmic poly(A) tail length is maintained by a dynamic equilibrium between cis-acting elements and cognate factors that promote deadenylation or polyadenylation, enabling rapid gene expression reprogramming in response to internal and external cellular cues. While cytoplasmic deadenylation and its pathophysiological implications have been extensively studied, cytoplasmic polyadenylation and its therapeutic potential remain less explored. This review discusses the distribution, regulation, and mechanisms of cytoplasmic polyadenylation element-binding proteins(CPEBs), highlighting their dual roles in either promoting or repressing gene expression depending on cellular context. We also explore their involvement in diseases such as tumor progression and metastasis, along with their potential as targets for novel therapeutic strategies.
Collapse
Affiliation(s)
| | - Raul Mendez
- Institute for Research in Biomedicine (IRB), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
15
|
Jain S. Can Schistosoma japonicum infection cause liver cancer? J Helminthol 2025; 99:e11. [PMID: 39924660 DOI: 10.1017/s0022149x24000762] [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] [Indexed: 02/11/2025]
Abstract
A co-relation between Schistosoma japonicum (Sj) and liver cancer (LC) in humans has been reported in the literature; however, this association is circumstantial. Due to the inconclusive nature of this association, the International Agency for Research on Cancer has placed Sj in Group 2B for LC, signifying it to be a 'possible carcinogen'. Many epidemiological, pathological and clinical studies have identified multiple factors, linked with Sj infection, which can lead to liver carcinogenesis. These factors include chronic inflammation in response to deposited eggs (which leads to fibrosis, cirrhosis and chromosomal instability at cellular level), hepatotoxic effects of egg-antigens, co-infection with hepatitis viruses, and up-regulation of glycolysis linked genes among others which predisposes hepatic tissue towards malignant transformation. The objective of this work is to present the current understanding on the association of Sj infection with LC. Mechanisms and factors linked with Sj infection that can lead to LC are emphasized, along with measures to diagnose and treat it. A comparison of liver carcinogenesis is also provided for cases linked with and independent of Sj infection. It appears that Sj, alone or with another carcinogen, is an important factor in liver carcinogenesis, but further studies are warranted to conclusively label 'infection with Sj alone' as a liver carcinogen.
Collapse
Affiliation(s)
- S Jain
- Independent Researcher, Institute for Globally Distributed Open Research and Education (IGDORE), Rewari, Haryana, India
| |
Collapse
|
16
|
Huang T, Zhou MY, Zou GL, Hu RH, Han L, Zhang QX, Zhao XK. Focal adhesion kinase promotes aerobic glycolysis in hepatic stellate cells via the cyclin D1/c-Myc/MCT-1 pathway to induce liver fibrosis. Sci Rep 2025; 15:4552. [PMID: 39915293 PMCID: PMC11802747 DOI: 10.1038/s41598-025-88538-8] [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: 09/11/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
Hepatic stellate cells (HSCs) transdifferentiate into myofibroblasts during liver fibrosis and exhibit increased glycolysis. Phosphorylated focal adhesion kinase (FAK) (pY397-FAK) promotes monocarboxylate transporter 1 (MCT-1) expression in HSCs to increase aerobic glycolysis and cause liver fibrosis. A combined multiomics analysis of C57BL/6 mice with tetrachloromethane (CCl4)-induced liver fibrosis was performed to identify the downstream FAK signaling pathway. The effect of the FAK inhibitor PF562271 on CCl4-induced liver fibrosis was explored by immunofluorescence of liver tissues. The migration, proliferation and aerobic glycolysis of LX-2 cells after stimulation and activation by transforming growth factor beta-1 (TGF-β1) or suppression by PF562271 was assessed in vitro. Multiomics analysis of a successfully generated CCl4-induced liver fibrosis mouse model was performed. FAK and cyclin D1 were significantly enriched in mice with CCl4-induced liver fibrosis. In vivo, the MCT-1 and alpha smooth muscle actin (α-SMA) levels were increased in mice with CCl4-induced liver fibrosis, and MCT-1 and α-SMA expression decreased after PF562271 treatment. In vitro, PF562271 alleviated TGF-β1-induced LX-2 activation. LX-2 cells showed diminished migration, proliferation, and aerobic glycolysis after PF562271 intervention. FAK promotes aerobic glycolysis in LX-2 cells through the cyclin D1/c-Myc/MCT-1 pathway, thereby increasing liver fibrosis.
Collapse
Affiliation(s)
- Tao Huang
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Ming-Yu Zhou
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Gao-Liang Zou
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Rui-Han Hu
- Department of Cardiology, Guiqian International General Hospital, Guiyang, Guizhou Province, China
| | - Lu Han
- Department of Comprehensive Ward, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Qing-Xiu Zhang
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Xue-Ke Zhao
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China.
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, No. 9 Beijing Road, Guiyang, 550004, Guizhou Province, China.
| |
Collapse
|
17
|
Zhang X, Zeng Y, Ying H, Hong Y, Xu J, Lin R, Chen Y, Wu X, Cai W, Xia Z, Zhao Q, Wang Y, Zhou R, Zhu D, Yu F. AdipoRon mitigates liver fibrosis by suppressing serine/glycine biosynthesis through ATF4-dependent glutaminolysis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117511. [PMID: 39662457 DOI: 10.1016/j.ecoenv.2024.117511] [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: 05/19/2024] [Revised: 11/22/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
AdipoRon has been validated for its ability to reverse liver fibrosis, yet the underlying mechanisms remain to be thoroughly investigated. Collagen, predominantly synthesized and secreted in hepatic stellate cells (HSCs), relies on glycine as a crucial constituent. Activating transcription factor 4 (ATF4) serves as a pivotal transcriptional regulator in amino acid metabolism. Therefore, our objective is to explore the impact of AdipoRon on ATF4-mediated endoplasmic reticulum stress and amino acid metabolism in HSCs. We induced liver fibrosis in mice through intraperitoneal injection of CCl4 and administered AdipoRon (50 mg/kg) via gavage. In vitro studies were predominantly conducted using LX-2 cells. Our findings demonstrated that AdipoRon effectively suppressed ATF4-mediated endoplasmic reticulum stress in HSCs and assumed a crucial role in hindering serine/glycine biosynthesis. Interestingly, this inhibitory effect of AdipoRon on serine/glycine biosynthesis is regulated by PSAT1-mediated glutaminolysis, resulting in a subsequent decrease in collagen synthesis within HSCs. This study provides potential mechanistic insights into the treatment of liver fibrosis with AdipoRon.
Collapse
Affiliation(s)
- Xiangting Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuan Zeng
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huiya Ying
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiwen Hong
- Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jun Xu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Rong Lin
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuhao Chen
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiao Wu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weimin Cai
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ziqiang Xia
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qian Zhao
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yixiao Wang
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ruoru Zhou
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dandan Zhu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fujun Yu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
18
|
Gilgenkrantz H, Paradis V, Lotersztajn S. Cell metabolism-based therapy for liver fibrosis, repair, and hepatocellular carcinoma. Hepatology 2025; 81:269-287. [PMID: 37212145 PMCID: PMC11643143 DOI: 10.1097/hep.0000000000000479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/21/2023] [Indexed: 05/23/2023]
Abstract
Progression of chronic liver injury to fibrosis, abnormal liver regeneration, and HCC is driven by a dysregulated dialog between epithelial cells and their microenvironment, in particular immune, fibroblasts, and endothelial cells. There is currently no antifibrogenic therapy, and drug treatment of HCC is limited to tyrosine kinase inhibitors and immunotherapy targeting the tumor microenvironment. Metabolic reprogramming of epithelial and nonparenchymal cells is critical at each stage of disease progression, suggesting that targeting specific metabolic pathways could constitute an interesting therapeutic approach. In this review, we discuss how modulating intrinsic metabolism of key effector liver cells might disrupt the pathogenic sequence from chronic liver injury to fibrosis/cirrhosis, regeneration, and HCC.
Collapse
Affiliation(s)
- Hélène Gilgenkrantz
- Paris-Cité University, INSERM, Center for Research on Inflammation, Paris, France
| | - Valérie Paradis
- Paris-Cité University, INSERM, Center for Research on Inflammation, Paris, France
- Pathology Department, Beaujon Hospital APHP, Paris-Cité University, Clichy, France
| | - Sophie Lotersztajn
- Paris-Cité University, INSERM, Center for Research on Inflammation, Paris, France
| |
Collapse
|
19
|
Li L, Lei Q, Zhen Y, Cao L, Dong Y, Liu X, Wang M. Lactate Dehydrogenase Inhibition Protects against Hepatic Fibrosis by Regulating Metabolic Reprogramming of Hepatic Stellate Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27953-27964. [PMID: 39632278 DOI: 10.1021/acs.jafc.4c08211] [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: 12/07/2024]
Abstract
Hepatic stellate cells (HSCs) activation results in liver fibrosis. When HSCs are activated, metabolism is reprogrammed. However, metabolic alteration in HSCs activation has not been sufficiently addressed. This study aims to investigate the role of lactate dehydrogenase (LDH) inhibition in HSCs activation with an emphasis on the metabolic reprogramming. Mice were subjected to carbon tetrachloride (CCl4) to induce liver injury. In addition, the primary HSCs were isolated for mechanism investigation. Our study demonstrated that LDH inhibition impaired HSCs activation through suppressing the enhanced glycolysis by blocking nicotinamide adenine dinucleotide (NAD+) regeneration. Meanwhile, LDH inhibition also impeded the glutamine metabolism through the lactic acid/histone deacetylase (HDAC)/histone acetylation/cellular-myelocytomatosis viral oncogene (c-Myc) signaling pathway. Our findings demonstrated that LDH inhibition is a potential target for liver fibrosis treatment, which provides new insight into the pathogenesis of liver fibrosis from the aspect of metabolic reprogramming, contributing to the design of a novel therapeutic strategy in the management of liver fibrosis.
Collapse
Affiliation(s)
- Lisi Li
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Provincial Technology Innovation Center for Anti-tumor Molecular Targeting New Drugs Development, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
- Department of Biology and Medical Technology, Shijiazhuang Information Engineering Vocational College, Shijiazhuang 052161, China
| | - Qi Lei
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Provincial Technology Innovation Center for Anti-tumor Molecular Targeting New Drugs Development, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
| | - Yifan Zhen
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Provincial Technology Innovation Center for Anti-tumor Molecular Targeting New Drugs Development, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
| | - Lixue Cao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Provincial Technology Innovation Center for Anti-tumor Molecular Targeting New Drugs Development, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
| | - Yujia Dong
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Provincial Technology Innovation Center for Anti-tumor Molecular Targeting New Drugs Development, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
| | - Xifu Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Provincial Technology Innovation Center for Anti-tumor Molecular Targeting New Drugs Development, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
| | - Meng Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Provincial Technology Innovation Center for Anti-tumor Molecular Targeting New Drugs Development, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
20
|
Zhang Q, Ran T, Li S, Han L, Chen S, Lin G, Wu H, Wu H, Feng S, Chen J, Zhang Q, Zhao X. Catalpol ameliorates liver fibrosis via inhibiting aerobic glycolysis by EphA2/FAK/Src signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156047. [PMID: 39321687 DOI: 10.1016/j.phymed.2024.156047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/03/2024] [Accepted: 09/12/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Hepatic fibrosis is a pathological process in a variety of acute or chronic liver injuries. Catalpol (CAT), an iridoid glycoside found in Rehmannia glutinosa, has several pharmacological properties, including anti-inflammatory, antidiabetic and anti-fibrotic effects. Nevertheless, there is currently no report on whether CAT regulates the aerobic glycolysis of hepatic stellate cells (HSCs) to inhibit liver fibrosis. OBJECTIVE This study aimed to investigate the protective effects of CAT on hepatic fibrosis and elucidate its underlying mechanisms. METHODS To explore whether CAT improved liver fibrosis in vivo and in vitro, hepatic fibrosis was induced to mice by intraperitoneally injecting carbon tetrachloride (CCl4). Additionally, LX-2 cells were stimulated with transforming growth factor-β (TGF-β) to simulate fibrosis in vitro. Serum markers of liver injury were examined by using an automatic biochemical analyzer. Histopathological staining, Immunofluorescence (IF) staining, Western blot (WB) analysis, co-immunoprecipitation (Co-IP), drug affinity responsive target stability (DARTS), cellular thermal shift assay (CETSA), etc. were employed to identify the targeting between CAT and EphA2 and detect the expression of aerobic glycolysis related proteins, fiber markers and signaling pathways that are responsible for CAT's anti-fibrotic effects of CAT. RESULTS Results showed that CAT significantly inhibited hepatic injury, fibrogenesis and inflammation in mice treated with CCl4. This was demonstrated by the enhancement of fibrosis markers, liver function indices, and histopathology. In addition, CAT significantly inhibited the activation of HSCs in TGF-β-induced LX-2 cells, as indicated by decreased proliferation, migration, and expression of collagen I and a-SMA. The study results also suggested that CAT may exert anti-fibrotic effects by inhibiting glycolysis in activated HSCs and in CCl4-treated mice. Mechanistically, CAT directly targets Ephrin type-A receptor 2 (EphA2) to reduce binding with focal adhesion kinases (FAK) and significantly inhibits the FAK/Src pathway. In addition, the pharmacological inhibition of EphA2 cannot further increase the therapeutic effects of CAT on liver fibrosis in vivo and in vitro. CONCLUSION The study findings generally demonstrated that CAT presented a novel therapeutic method to treat hepatic fibrosis; this method which inhibits the aerobic glycolysis of activated HSCs through the EphA2/FAK/Src signaling pathway.
Collapse
Affiliation(s)
- Qingxiu Zhang
- Department of Infectious Disease, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China.
| | - Tao Ran
- Department of Infectious Disease, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China.
| | - Shiliang Li
- Department of Vascular Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China.
| | - Lu Han
- Department of Infectious Disease, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China.
| | - Shaojie Chen
- Guizhou Medical University, Guiyang 550000, China.
| | - Guoyuan Lin
- Department of Infectious Disease, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China.
| | - Huayue Wu
- Department of Infectious Disease, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China.
| | - Huan Wu
- Department of Infectious Disease, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China.
| | - Shu Feng
- Department of Infectious Disease, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China.
| | - Jiyu Chen
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China.
| | - Qian Zhang
- Department of Nephrology, The Guizhou provincial people's Hospital, Guiyang 550000, China.
| | - Xueke Zhao
- Department of Infectious Disease, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China.
| |
Collapse
|
21
|
Liu Y, Zhang W, Lin N, Yang Z, Liu Y, Chen H. SPARC activates p38γ signaling to promote PFKFB3 protein stabilization and contributes to keloid fibroblast glycolysis. Inflamm Regen 2024; 44:44. [PMID: 39482755 PMCID: PMC11529245 DOI: 10.1186/s41232-024-00357-y] [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: 08/01/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Keloids are currently challenging to treat because they recur after resection which may affect patients' quality of life. At present, no universal consensus on treatment regimen has been established. Thus, finding new molecular mechanisms underlying keloid formation is imminent. This study aimed to explore the function of secreted protein acidic and cysteine rich (SPARC) on keloids and its behind exact mechanisms. METHODS The expression of SPARC, p38γ, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), α-SMA, and Ki67 in patients with keloid and bleomycin (BLM)-induced fibrosis mice was assessed utilizing western blot, qRT-PCR, and immunohistochemical staining. After transfected with pcDNA-SPARC, si-SPARC-1#, si-SPARC-2#, and si-p38γ, and treated with glycolytic inhibitor (2-DG) or p38 inhibitor (SB203580), CCK-8, EdU, transwell, and western blot were utilized for assessing the proliferation, migration, and collagen production of keloid fibroblasts (KFs). RESULTS SPARC, p38γ, and PFKFB3 were highly expressed in patients with keloid and BLM-induced fibrosis mice. SPARC promoted the proliferation, migration, and collagen production of KFs via inducing glycolysis. Moreover, SPARC could activate p38γ signaling to stabilize PFKFB3 protein expression in KFs. Next, we demonstrated that SPARC promoted the proliferation, migration, collagen production, and glycolysis of KFs via regulating p38γ signaling. In addition, in BLM-induced fibrosis mice, inhibition of p38γ and PFKFB3 relieved skin fibrosis. CONCLUSIONS Our findings indicated that SPARC could activate p38γ pathway to stabilize the expression of PFKFB3, and thus promote the glycolysis of KFs and the progression of keloid.
Collapse
Affiliation(s)
- Yining Liu
- Department of Burn and Plastic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, 266100, Shandong, People's Republic of China
| | - Wei Zhang
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 JingWu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Nan Lin
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 JingWu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Zelei Yang
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 JingWu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Yanxin Liu
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 JingWu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Huaxia Chen
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 JingWu Road, Jinan, 250021, Shandong, People's Republic of China.
| |
Collapse
|
22
|
Guo X, Zheng B, Wang J, Zhao T, Zheng Y. Exploring the mechanism of action of Chinese medicine in regulating liver fibrosis based on the alteration of glucose metabolic pathways. Phytother Res 2024; 38:4865-4876. [PMID: 36433866 DOI: 10.1002/ptr.7667] [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: 05/01/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 11/26/2022]
Abstract
In recent years, metabolic reprogramming in liver fibrosis has become a research hotspot in the field of liver fibrosis at home and abroad. Liver fibrosis is a pathological change caused by chronic liver injury from a variety of causes. Liver fibrosis is a common pathological feature of many chronic liver diseases such as chronic hepatitis B, non-alcoholic steatohepatitis, and autoimmune hepatitis, as well as the pathogenesis of the disease. The development of chronic liver disease into cirrhosis must go through the pathological process of liver fibrosis, in which hepatic stellate cells (HSC) play an important role. Following liver injury, HSC are activated and transdifferentiated into scar-forming myofibroblasts, which drive the trauma healing response and which rely on the deposition of collagen-rich extracellular matrix to maintain tissue integrity. This reaction will continue without strict control, which will lead to excessive accumulation of matrix and liver fibrosis. The mechanisms and clinical studies of liver fibrosis have been the focus of research in liver diseases. In recent years, several studies have revealed the mechanism of HSC metabolic reprogramming and the impact of this process on liver fibrosis, in which glucose metabolic reprogramming plays an important role in the activation of HSC, and it mainly meets the energy demand of HSC activation by upregulating glycolysis. Glycolysis is the process by which one molecule of glucose is broken down into two molecules of pyruvate and produces energy and lactate under anaerobic conditions. Various factors have been found to be involved in regulating the glycolytic process of HSC, including glucose transport, intracellular processing of glucose, exosome secretion, and lactate production, etc. Inhibition of the glycolytic process of HSC can be an effective strategy against liver fibrosis. Currently, the combined action of multiple targets and links of Chinese medicine such as turmeric, comfrey, rhubarb and scutellaria baicalensis against the mechanism of liver fibrosis can effectively improve or even reverse liver fibrosis. This paper summarizes that turmeric extract curcumin, comfrey extract comfreyin, rhubarb, Subtle yang yu yin granules, Scutellaria baicalensis extract oroxylin A and cardamom extract cardamomin affect liver fibrosis by regulating gluconeogenic reprogramming. Therefore, studying the mechanism of action of TCM in regulating liver fibrosis through reprogramming of glucose metabolism is promising to explore new methods and approaches for Chinese Medicine modernization research.
Collapse
Affiliation(s)
- Xinhua Guo
- Department of Physiology, College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Bowen Zheng
- Department of Physiology, College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Jiahui Wang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, China
| | - Tiejian Zhao
- Department of Physiology, College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Yang Zheng
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
23
|
Pan M, Li H, Shi X. A New Target for Hepatic Fibrosis Prevention and Treatment: The Warburg Effect. FRONT BIOSCI-LANDMRK 2024; 29:321. [PMID: 39344326 DOI: 10.31083/j.fbl2909321] [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: 05/13/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 10/01/2024]
Abstract
Hepatic fibrosis is a major public health problem that endangers human wellbeing. In recent years, a number of studies have revealed the important impact of metabolic reprogramming on the occurrence and development of hepatic fibrosis. Among them, the Warburg effect, as an intracellular glucose metabolism reprogramming, can promote the occurrence and development of hepatic fibrosis by promoting the activation of hepatic stellate cells (HSCs) and inducing the polarization of liver macrophages (KC). Understanding the Warburg effect and its important role in the progression of hepatic fibrosis will assist in developing new strategies for the prevention and treatment of hepatic fibrosis. This review focuses on the Warburg effect and the specific mechanism by which it affects the progression of hepatic fibrosis by regulating HSCs activation and KC polarization. In addition, we also summarize and discuss the related experimental drugs and their mechanisms that inhibit the Warburg effect by targeting key proteins of glycolysis in order to improve hepatic fibrosis in the hope of providing more effective strategies for the clinical treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Meng Pan
- College of Basic Medical Sciences, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| | - Huanyu Li
- Second Clinical Medical College, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| | - Xiaoyan Shi
- College of Basic Medical Sciences, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| |
Collapse
|
24
|
Zhong Z, Cui XL, Tan KJ, Wu XY, Zhu XJ, Zhang JY, Zhang WJ, Wang HY, Zhang PL. Apoptotic vesicles (apoVs) derived from fibroblast-converted hepatocyte-like cells effectively ameliorate liver fibrosis. J Nanobiotechnology 2024; 22:541. [PMID: 39238002 PMCID: PMC11375929 DOI: 10.1186/s12951-024-02824-7] [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: 06/24/2024] [Accepted: 08/31/2024] [Indexed: 09/07/2024] Open
Abstract
Liver fibrosis is a serious global health issue for which effective treatment remains elusive. Chemical-induced hepatocyte-like cells (ciHeps) have emerged as an appealing source for cell transplantation therapy, although they present several challenges such as the risk of lung thromboembolism or hemorrhage. Apoptotic vesicles (apoVs), small membrane vesicles generated during the apoptosis process, have gained attention for their role in regulating various physiological and pathological processes. In this study, we generated ciHep-derived apoVs (ciHep-apoVs) and investigated their therapeutic potential in alleviating liver fibrosis. Our findings revealed that ciHep-apoVs induced the transformation of macrophages into an anti-inflammatory phenotype, effectively suppressed the activity of activated hepatic stellate cells (aHSCs), and enhanced the survival of hepatocytes. When intravenously administered to mice with liver fibrosis, ciHep-apoVs were primarily engulfed by macrophages and myofibroblasts, leading to a reduction in liver inflammation and fibrosis. Proteomic and miRNA analyses showed that ciHep-apoVs were enriched in various functional molecules that modulate crucial cellular processes, including metabolism, signaling transduction, and ECM-receptor interactions. ciHep-apoVs effectively suppressed aHSCs activity through the synergistic inhibition of glycolysis, the PI3K/AKT/mTOR pathway, and epithelial-to-mesenchymal transition (EMT) cascades. These findings highlight the potential of ciHep-apoVs as multifunctional nanotherapeutics for liver fibrosis and provide insights into the treatment of other liver diseases and fibrosis in other organs.
Collapse
Affiliation(s)
- Zhi Zhong
- National Center for Liver Cancer, Naval Medical University, 366 Qianju Road, Shanghai, 201805, China
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiu-Liang Cui
- National Center for Liver Cancer, Naval Medical University, 366 Qianju Road, Shanghai, 201805, China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University, Shanghai, 200438, China
| | - Kun-Jiang Tan
- National Center for Liver Cancer, Naval Medical University, 366 Qianju Road, Shanghai, 201805, China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University, Shanghai, 200438, China
| | - Xiang-Yu Wu
- National Center for Liver Cancer, Naval Medical University, 366 Qianju Road, Shanghai, 201805, China
| | - Xiang-Jie Zhu
- National Center for Liver Cancer, Naval Medical University, 366 Qianju Road, Shanghai, 201805, China
- Institute of Metabolism & Integrative Biology, Fudan University, Shanghai, 200438, China
| | - Jiu-Yu Zhang
- National Center for Liver Cancer, Naval Medical University, 366 Qianju Road, Shanghai, 201805, China
- Institute of Metabolism & Integrative Biology, Fudan University, Shanghai, 200438, China
| | - Wei-Jia Zhang
- National Center for Liver Cancer, Naval Medical University, 366 Qianju Road, Shanghai, 201805, China
| | - Hong-Yang Wang
- National Center for Liver Cancer, Naval Medical University, 366 Qianju Road, Shanghai, 201805, China.
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University, Shanghai, 200438, China.
| | - Pei-Lin Zhang
- National Center for Liver Cancer, Naval Medical University, 366 Qianju Road, Shanghai, 201805, China.
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University, Shanghai, 200438, China.
| |
Collapse
|
25
|
Miguel V, Alcalde-Estévez E, Sirera B, Rodríguez-Pascual F, Lamas S. Metabolism and bioenergetics in the pathophysiology of organ fibrosis. Free Radic Biol Med 2024; 222:85-105. [PMID: 38838921 DOI: 10.1016/j.freeradbiomed.2024.06.001] [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: 04/07/2024] [Revised: 05/15/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
Fibrosis is the tissue scarring characterized by excess deposition of extracellular matrix (ECM) proteins, mainly collagens. A fibrotic response can take place in any tissue of the body and is the result of an imbalanced reaction to inflammation and wound healing. Metabolism has emerged as a major driver of fibrotic diseases. While glycolytic shifts appear to be a key metabolic switch in activated stromal ECM-producing cells, several other cell types such as immune cells, whose functions are intricately connected to their metabolic characteristics, form a complex network of pro-fibrotic cellular crosstalk. This review purports to clarify shared and particular cellular responses and mechanisms across organs and etiologies. We discuss the impact of the cell-type specific metabolic reprogramming in fibrotic diseases in both experimental and human pathology settings, providing a rationale for new therapeutic interventions based on metabolism-targeted antifibrotic agents.
Collapse
Affiliation(s)
- Verónica Miguel
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
| | - Elena Alcalde-Estévez
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain; Department of Systems Biology, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Belén Sirera
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Fernando Rodríguez-Pascual
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Santiago Lamas
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain.
| |
Collapse
|
26
|
Wang Y, Li H, Jiang S, Fu D, Lu X, Lu M, Li Y, Luo D, Wu K, Xu Y, Li G, Zhou Y, Zhou Y, Chen W, Liu Q, Mao H. The glycolytic enzyme PFKFB3 drives kidney fibrosis through promoting histone lactylation-mediated NF-κB family activation. Kidney Int 2024; 106:226-240. [PMID: 38789037 DOI: 10.1016/j.kint.2024.04.016] [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: 08/18/2023] [Revised: 03/27/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024]
Abstract
Persistently elevated glycolysis in kidney has been demonstrated to promote chronic kidney disease (CKD). However, the underlying mechanism remains largely unclear. Here, we observed that 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), a key glycolytic enzyme, was remarkably induced in kidney proximal tubular cells (PTCs) following ischemia-reperfusion injury (IRI) in mice, as well as in multiple etiologies of patients with CKD. PFKFB3 expression was positively correlated with the severity of kidney fibrosis. Moreover, patients with CKD and mice exhibited increased urinary lactate/creatine levels and kidney lactate, respectively. PTC-specific deletion of PFKFB3 significantly reduced kidney lactate levels, mitigated inflammation and fibrosis, and preserved kidney function in the IRI mouse model. Similar protective effects were observed in mice with heterozygous deficiency of PFKFB3 or those treated with a PFKFB3 inhibitor. Mechanistically, lactate derived from PFKFB3-mediated tubular glycolytic reprogramming markedly enhanced histone lactylation, particularly H4K12la, which was enriched at the promoter of NF-κB signaling genes like Ikbkb, Rela, and Relb, activating their transcription and facilitating the inflammatory response. Further, PTC-specific deletion of PFKFB3 inhibited the activation of IKKβ, I κ B α, and p65 in the IRI kidneys. Moreover, increased H4K12la levels were positively correlated with kidney inflammation and fibrosis in patients with CKD. These findings suggest that tubular PFKFB3 may play a dual role in enhancing NF-κB signaling by promoting both H4K12la-mediated gene transcription and its activation. Thus, targeting the PFKFB3-mediated NF-κB signaling pathway in kidney tubular cells could be a novel strategy for CKD therapy.
Collapse
Affiliation(s)
- Yating Wang
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Hongyu Li
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Simin Jiang
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Dongying Fu
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Xiaohui Lu
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Miaoqing Lu
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China; Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Li
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Dan Luo
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Kefei Wu
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Yiping Xu
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Guanglan Li
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Yi Zhou
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Yiming Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Basic and Translational Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Chen
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.
| | - Qinghua Liu
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.
| | - Haiping Mao
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.
| |
Collapse
|
27
|
Huang X, Wang X, Wang Y, Shen S, Chen W, Liu T, Wang P, Fan X, Liu L, Jia J, Cong M. TIMP-1 Promotes Expression of MCP-1 and Macrophage Migration by Inducing Fli-1 in Experimental Liver Fibrosis. J Clin Transl Hepatol 2024; 12:634-645. [PMID: 38993513 PMCID: PMC11233975 DOI: 10.14218/jcth.2023.00514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/08/2024] [Accepted: 05/11/2024] [Indexed: 07/13/2024] Open
Abstract
Background and Aims Tissue inhibitor of metalloproteinase-1 (TIMP-1) plays a role in the excessive generation of extracellular matrix in liver fibrosis. This study aimed to explore the pathways through which TIMP-1 controls monocyte chemoattractant protein-1 (MCP-1) expression and promotes hepatic macrophage recruitment. Methods Liver fibrosis was triggered through carbon tetrachloride, and an adeno-associated virus containing small interfering RNA targeting TIMP-1 (siRNA-TIMP-1) was administered to both rats and mice. We assessed the extent of fibrosis and macrophage recruitment. The molecular mechanisms regulating macrophage recruitment by TIMP-1 were investigated through transwell migration assays, luciferase reporter assays, the use of pharmacological modulators, and an analysis of extracellular vesicles (EVs). Results siRNA-TIMP-1 alleviated carbon tetrachloride-induced liver fibrosis, reducing macrophage migration and MCP-1 expression. Co-culturing macrophages with hepatic stellate cells (HSCs) post-TIMP-1 downregulation inhibited macrophage migration. In siRNA-TIMP-1-treated HSCs, microRNA-145 (miRNA-145) expression increased, while the expression of Friend leukemia virus integration-1 (Fli-1) and MCP-1 was inhibited. Downregulation of Fli-1 led to decreased MCP-1 expression, whereas Fli-1 overexpression increased MCP-1 expression within HSCs. Transfection with miRNA-145 mimics reduced the expression of both Fli-1 and MCP-1, while miRNA-145 inhibitors elevated the expression of both Fli-1 and MCP-1 in HSCs. miRNA-145 bound directly to the 3'-UTR of Fli-1, and miRNA-145-enriched EVs secreted by HSCs after TIMP-1 downregulation influenced macrophage recruitment. Conclusions TIMP-1 induces Fli-1 expression through miRNA-145, subsequently increasing MCP-1 expression and macrophage recruitment. MiRNA-145-enriched EVs from HSCs can transmit biological information and magnify the function of TIMP-1.
Collapse
Affiliation(s)
- Xiaoli Huang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Xiaofan Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Yanhong Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
- Dongying People's Hospital, Dongying, Shandong, China
| | - Shuangjun Shen
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Wei Chen
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tianhui Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Ping Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Xu Fan
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Lin Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Min Cong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| |
Collapse
|
28
|
Horn P, Tacke F. Metabolic reprogramming in liver fibrosis. Cell Metab 2024; 36:1439-1455. [PMID: 38823393 DOI: 10.1016/j.cmet.2024.05.003] [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: 04/02/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024]
Abstract
Chronic liver diseases, primarily metabolic dysfunction-associated steatotic liver disease (MASLD), harmful use of alcohol, or viral hepatitis, may result in liver fibrosis, cirrhosis, and cancer. Hepatic fibrogenesis is a complex process with interactions between different resident and non-resident heterogeneous liver cell populations, ultimately leading to deposition of extracellular matrix and organ failure. Shifts in cell phenotypes and functions involve pronounced transcriptional and protein synthesis changes that require metabolic adaptations in cellular substrate metabolism, including glucose and lipid metabolism, resembling changes associated with the Warburg effect in cancer cells. Cell activation and metabolic changes are regulated by metabolic stress responses, including the unfolded protein response, endoplasmic reticulum stress, autophagy, ferroptosis, and nuclear receptor signaling. These metabolic adaptations are crucial for inflammatory and fibrogenic activation of macrophages, lymphoid cells, and hepatic stellate cells. Modulation of these pathways, therefore, offers opportunities for novel therapeutic approaches to halt or even reverse liver fibrosis progression.
Collapse
Affiliation(s)
- Paul Horn
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Digital Clinician Scientist Program, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany.
| |
Collapse
|
29
|
Yao S, Chai H, Tao T, Zhang L, Yang X, Li X, Yi Z, Wang Y, An J, Wen G, Jin H, Tuo B. Role of lactate and lactate metabolism in liver diseases (Review). Int J Mol Med 2024; 54:59. [PMID: 38785162 PMCID: PMC11188982 DOI: 10.3892/ijmm.2024.5383] [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: 01/31/2024] [Accepted: 03/22/2024] [Indexed: 05/25/2024] Open
Abstract
Lactate is a byproduct of glycolysis, and before the Warburg effect was revealed (in which glucose can be fermented in the presence of oxygen to produce lactate) it was considered a metabolic waste product. At present, lactate is not only recognized as a metabolic substrate that provides energy, but also as a signaling molecule that regulates cellular functions under pathophysiological conditions. Lactylation, a post‑translational modification, is involved in the development of various diseases, including inflammation and tumors. Liver disease is a major health challenge worldwide. In normal liver, there is a net lactate uptake caused by gluconeogenesis, exhibiting a higher net lactate clearance rate compared with any other organ. Therefore, abnormalities of lactate and lactate metabolism lead to the development of liver disease, and lactate and lactate metabolism‑related genes can be used for predicting the prognosis of liver disease. Targeting lactate production, regulating lactate transport and modulating lactylation may be potential treatment approaches for liver disease. However, currently there is not a systematic review that summarizes the role of lactate and lactate metabolism in liver diseases. In the present review, the role of lactate and lactate metabolism in liver diseases including liver fibrosis, non‑alcoholic fatty liver disease, acute liver failure and hepatocellular carcinoma was summarized with the aim to provide insights for future research.
Collapse
Affiliation(s)
- Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Hongyu Chai
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Ting Tao
- Department of Burns and Plastic Surgery, Fuling Hospital, Chongqing University, Chongqing 408099, P.R. China
| | - Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Xingyue Yang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Xin Li
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Zhiqiang Yi
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yongfeng Wang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jiaxin An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Guorong Wen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
30
|
Khanal S, Liu Y, Bamidele AO, Wixom AQ, Washington AM, Jalan-Sakrikar N, Cooper SA, Vuckovic I, Zhang S, Zhong J, Johnson KL, Charlesworth MC, Kim I, Yeon Y, Yoon S, Noh YK, Meroueh C, Timbilla AA, Yaqoob U, Gao J, Kim Y, Lucien F, Huebert RC, Hay N, Simons M, Shah VH, Kostallari E. Glycolysis in hepatic stellate cells coordinates fibrogenic extracellular vesicle release spatially to amplify liver fibrosis. SCIENCE ADVANCES 2024; 10:eadn5228. [PMID: 38941469 PMCID: PMC11212729 DOI: 10.1126/sciadv.adn5228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/24/2024] [Indexed: 06/30/2024]
Abstract
Liver fibrosis is characterized by the activation of perivascular hepatic stellate cells (HSCs), the release of fibrogenic nanosized extracellular vesicles (EVs), and increased HSC glycolysis. Nevertheless, how glycolysis in HSCs coordinates fibrosis amplification through tissue zone-specific pathways remains elusive. Here, we demonstrate that HSC-specific genetic inhibition of glycolysis reduced liver fibrosis. Moreover, spatial transcriptomics revealed a fibrosis-mediated up-regulation of EV-related pathways in the liver pericentral zone, which was abrogated by glycolysis genetic inhibition. Mechanistically, glycolysis in HSCs up-regulated the expression of EV-related genes such as Ras-related protein Rab-31 (RAB31) by enhancing histone 3 lysine 9 acetylation on the promoter region, which increased EV release. Functionally, these glycolysis-dependent EVs increased fibrotic gene expression in recipient HSC. Furthermore, EVs derived from glycolysis-deficient mice abrogated liver fibrosis amplification in contrast to glycolysis-competent mouse EVs. In summary, glycolysis in HSCs amplifies liver fibrosis by promoting fibrogenic EV release in the hepatic pericentral zone, which represents a potential therapeutic target.
Collapse
Affiliation(s)
- Shalil Khanal
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Yuanhang Liu
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Alexander Q. Wixom
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Alexander M. Washington
- Biochemistry and Molecular Biology Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Nidhi Jalan-Sakrikar
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Shawna A. Cooper
- Biochemistry and Molecular Biology Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Ivan Vuckovic
- Metabolomics Core, Mayo Clinic, Rochester, MN 55905, USA
| | - Song Zhang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jun Zhong
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Iljung Kim
- Department of Computer Science, Hanyang University, Seoul 04763, Republic of South Korea
| | - Yubin Yeon
- Department of Computer Science, Hanyang University, Seoul 04763, Republic of South Korea
| | - Sangwoong Yoon
- School of Computational Sciences, Korea Institute for Advanced Study, Seoul 02455, Republic of South Korea
| | - Yung-Kyun Noh
- Department of Computer Science, Hanyang University, Seoul 04763, Republic of South Korea
- School of Computational Sciences, Korea Institute for Advanced Study, Seoul 02455, Republic of South Korea
| | - Chady Meroueh
- Department of Pathology, Division of Anatomic Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Abdul Aziz Timbilla
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Medical Biochemistry, Faculty of Medicine, Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Usman Yaqoob
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jinhang Gao
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
- Lab of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy; Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Yohan Kim
- Department of Urology, Mayo Clinic, Rochester, MN 55905, USA
| | - Fabrice Lucien
- Department of Urology, Mayo Clinic, Rochester, MN 55905, USA
| | - Robert C. Huebert
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Nissim Hay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Michael Simons
- Cardiovascular Research Center, Yale University, New Haven, CI 06510, USA
| | - Vijay H. Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Enis Kostallari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
31
|
Cao Y, Wang S, Zhang M, Lai B, Liang Y. PFKFB3-mediated glycolysis in hepatic stellate cells promotes liver regeneration. Biochem Biophys Res Commun 2024; 712-713:149958. [PMID: 38640731 DOI: 10.1016/j.bbrc.2024.149958] [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: 03/15/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
Hepatic stellate cells (HSCs) perform a significant function in liver regeneration (LR) by becoming active. We propose to investigate if activated HSCs enhance glycolysis via PFKFB3, an essential glycolytic regulator, and whether targeting this pathway could be beneficial for LR. The liver and isolated HSCs of mice subjected to 2/3 partial hepatectomy (PHx) exhibited a significant rise in PFKFB3 expression, as indicated by quantitative RT-PCR analyses and Western blotting. Also, the primary HSCs of mice subjected to PHx have a significant elevation of the glycolysis level. Knocking down PFKFB3 significantly diminished the enhancement of glycolysis by PDGF in human LX2 cells. The hepatocyte proliferation in mice treated with PHx was almost completely prevented when the PFKFB3 inhibitor 3PO was administered, emerging that PFKFB3 is essential in LR. Furthermore, there was a decline in mRNA expression of immediate early genes and proinflammatory cytokines. In terms of mechanism, both the p38 MAP kinase and ERK1/2 phosphorylation in LO2 cells and LO2 proliferation were significantly reduced by the conditioned medium (CM) obtained from LX2 cells with either PFKFB3 knockdown or inhibition. Compared to the control group, isolated hepatocytes from 3PO-treated mice showed decreased p38 MAP kinase and ERK1/2 phosphorylation and proliferation. Thus, LR after PHx involves the activation of PFKFB3 in HSCs, which enhances glycolysis and promotes lactate production, thereby facilitating hepatocyte proliferation via the p38/ERK MAPK signaling pathway.
Collapse
Affiliation(s)
- Yapeng Cao
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Siyu Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Min Zhang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Baochang Lai
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yanni Liang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xian Yang, 712046, China.
| |
Collapse
|
32
|
Guo Z, Li H, Yu W, Ren Y, Zhu Z. Insights into the effect of benzotriazoles in liver using integrated metabolomic and transcriptomic analysis. ENVIRONMENT INTERNATIONAL 2024; 187:108716. [PMID: 38723456 DOI: 10.1016/j.envint.2024.108716] [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: 02/12/2024] [Revised: 04/03/2024] [Accepted: 05/02/2024] [Indexed: 05/19/2024]
Abstract
Benzotriazoles (BTRs) are a class of benzoheterocyclic chemicals that are frequently used as metal-corrosive inhibitors, both in industry and daily use. However, the exposure effect information on BTRs remains relatively limited. In this study, an integrated metabolomic and transcriptomic approach was utilized to evaluate the effect of three BTRs, benzotriazole, 6-chloro-1-hydroxi-benzotriazole, and 1-hydroxy-benzotriazole, in the mouse liver with results showing disrupted basal metabolic processes and vitamin and cofactor metabolism after 28 days. The expression of several genes that are related to the inflammatory response and aryl hydrocarbon receptor pathways, such as Gstt2 and Arntl, was altered by the exposure to BTRs. Exposure to BTRs also affected metabolites and genes that are involved in the immune system and xenobiotic responses. The altered expression of several cytochrome P450 family genes reveal a potential detoxification mechanism in the mouse liver. Taken together, our findings provide new insights into the multilayer response of the mouse liver to BTRs exposure as well as a resource for further exploration of the molecular mechanisms by which the response occurs.
Collapse
Affiliation(s)
- Zeqin Guo
- Medical College, Jiujiang University, Jiujiang, Jiangxi, 332000, China; Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, Jiangxi, 332000, China.
| | - Huimin Li
- Medical College, Jiujiang University, Jiujiang, Jiangxi, 332000, China; Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, Jiangxi, 332000, China
| | - Wenmin Yu
- Medical College, Jiujiang University, Jiujiang, Jiangxi, 332000, China; Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, Jiangxi, 332000, China
| | - Yaguang Ren
- Medical College, Jiujiang University, Jiujiang, Jiangxi, 332000, China; Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, Jiangxi, 332000, China
| | - Zhiguo Zhu
- Medical College, Jiujiang University, Jiujiang, Jiangxi, 332000, China; College of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, Jiangxi, 332000, China.
| |
Collapse
|
33
|
Peng W, Yang Y, Lu H, Shi H, Jiang L, Liao X, Zhao H, Wang W, Liu J. Network pharmacology combines machine learning, molecular simulation dynamics and experimental validation to explore the mechanism of acetylbinankadsurin A in the treatment of liver fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117682. [PMID: 38169205 DOI: 10.1016/j.jep.2023.117682] [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: 10/03/2023] [Revised: 12/10/2023] [Accepted: 12/26/2023] [Indexed: 01/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Kadsura coccinea (Lem.) A. C. Smith is known as "Heilaohu" of the Tujia ethnomedicine in China. It has anti-tumor, anti-oxidation, anti-HIV, anti-inflammatory and liver protective effects, used to treat diseases such as rheumatoid arthritis, cancer, gastritis and hepatitis. In this research, we investigated the anti-fibrotic effect and possible mechanisms of acetylbinankadsurin A (ACBA) in vitro and in vivo, which is a natural compound derived from roots of K. coccinea. AIM OF THE STUDY We try to evaluate the efficacy of ACBA in the treatment of liver fibrosis and to explore the underlying mechanisms of ACBA by network pharmacology, machine learning, molecular docking, molecular dynamics simulations, and experimental assessment. MATERIALS AND METHODS ACBA was isolated from the CH2Cl2 layer of the roots of K. coccinea through column chromatographic techniques. The structure of ACBA was determined by using 1D and 2D NMR. CCl4-induced C57BL/6 mouse liver fibrosis models were established to evaluate the anti-fibrosis effects of ACBA in vivo. The molecular targets of ACBA and liver fibrosis were obtained from various databases, then constructed a protein-protein interaction (PPI) networks through the STRING database. Gene ontology (GO) enrichment and kyoto encyclopedia of genes and genomes (KEGG) analysis were applied using the "clusterProfiler" R package. Furthermore, the key genes for ACBA treatment of liver fibrosis were identified by the least absolute shrinkage and selection operator (LASSO). Molecular docking and molecular dynamics simulations were also carried out. Finally, the target and pathway of ACBA were verified by immunofluorescence staining, RT-PCR and Western blot. RESULT First, ACBA attenuated CCl4-induced liver injury and fibrosis in vivo. These findings were accompanied by decreased expression of α-SMA and collagen I. Second, ACBA significantly decreased serum levels of ALT, AST, TNF-α and IL-6. Then, we identified 133 potential targets of ACBA and 7987 targets of liver fibrosis. KEGG analysis showed that ACBA could regulate the drug metabolism - cytochrome P450, fructose and mannose metabolism, IL-17 and NF-κB signaling pathways. Next, six core targets was screened by LASSO analysis including AKR1B1, PFKFB3, EPHA3, CDK1, CCR1 and CYP3A4. Molecular docking showed that ACBA has a good binding affinity for CCR1. Furthermore, compared with CCR1 inhibitor BX-471, The results of molecular simulation dynamics showed that ACBA was stable in binding with CCR1. Consistently, ACBA remarkably downregulated the expression of CCR1, p-NF-κBp65, p-IκBα, p-STAT1 and TNF-α proteins, which were upregulated in CCl4-induced hepatic fibrosis and LPS-THP-1 cells. CONCLUSION Our results suggest that ACBA significantly attenuated CCl4-induced liver fibrosis in histopathological and in serum level. This effect may be mediated by CCR1, NF-κB and STAT1 signalling.
Collapse
Affiliation(s)
- Wangxia Peng
- Science and Technology Innovation Center of Hunan University of Traditional Chinese Medicine, Innovation Base of Hunan State Key Laboratory of Innovative Medicine and Traditional Chinese Medicine, Changsha, 410208, China
| | - Yupei Yang
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Huaguan Lu
- Science and Technology Innovation Center of Hunan University of Traditional Chinese Medicine, Innovation Base of Hunan State Key Laboratory of Innovative Medicine and Traditional Chinese Medicine, Changsha, 410208, China
| | - Huan Shi
- Science and Technology Innovation Center of Hunan University of Traditional Chinese Medicine, Innovation Base of Hunan State Key Laboratory of Innovative Medicine and Traditional Chinese Medicine, Changsha, 410208, China
| | - Lihong Jiang
- Science and Technology Innovation Center of Hunan University of Traditional Chinese Medicine, Innovation Base of Hunan State Key Laboratory of Innovative Medicine and Traditional Chinese Medicine, Changsha, 410208, China
| | - Xiaolin Liao
- Science and Technology Innovation Center of Hunan University of Traditional Chinese Medicine, Innovation Base of Hunan State Key Laboratory of Innovative Medicine and Traditional Chinese Medicine, Changsha, 410208, China
| | - Hongqing Zhao
- Science and Technology Innovation Center of Hunan University of Traditional Chinese Medicine, Innovation Base of Hunan State Key Laboratory of Innovative Medicine and Traditional Chinese Medicine, Changsha, 410208, China
| | - Wei Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Jianjun Liu
- Science and Technology Innovation Center of Hunan University of Traditional Chinese Medicine, Innovation Base of Hunan State Key Laboratory of Innovative Medicine and Traditional Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
34
|
Luo L, Zhang W, You S, Cui X, Tu H, Yi Q, Wu J, Liu O. The role of epithelial cells in fibrosis: Mechanisms and treatment. Pharmacol Res 2024; 202:107144. [PMID: 38484858 DOI: 10.1016/j.phrs.2024.107144] [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: 11/02/2023] [Revised: 02/19/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Fibrosis is a pathological process that affects multiple organs and is considered one of the major causes of morbidity and mortality in multiple diseases, resulting in an enormous disease burden. Current studies have focused on fibroblasts and myofibroblasts, which directly lead to imbalance in generation and degradation of extracellular matrix (ECM). In recent years, an increasing number of studies have focused on the role of epithelial cells in fibrosis. In some cases, epithelial cells are first exposed to external physicochemical stimuli that may directly drive collagen accumulation in the mesenchyme. In other cases, the source of stimulation is mainly immune cells and some cytokines, and epithelial cells are similarly altered in the process. In this review, we will focus on the multiple dynamic alterations involved in epithelial cells after injury and during fibrogenesis, discuss the association among them, and summarize some therapies targeting changed epithelial cells. Especially, epithelial mesenchymal transition (EMT) is the key central step, which is closely linked to other biological behaviors. Meanwhile, we think studies on disruption of epithelial barrier, epithelial cell death and altered basal stem cell populations and stemness in fibrosis are not appreciated. We believe that therapies targeted epithelial cells can prevent the progress of fibrosis, but not reverse it. The epithelial cell targeting therapies will provide a wonderful preventive and delaying action.
Collapse
Affiliation(s)
- Liuyi Luo
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Wei Zhang
- Department of Oral Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Siyao You
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Xinyan Cui
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Hua Tu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Qiao Yi
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Jianjun Wu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| | - Ousheng Liu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
35
|
Zhou Y, Yan J, Huang H, Liu L, Ren L, Hu J, Jiang X, Zheng Y, Xu L, Zhong F, Li X. The m 6A reader IGF2BP2 regulates glycolytic metabolism and mediates histone lactylation to enhance hepatic stellate cell activation and liver fibrosis. Cell Death Dis 2024; 15:189. [PMID: 38443347 PMCID: PMC10914723 DOI: 10.1038/s41419-024-06509-9] [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: 11/01/2023] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 03/07/2024]
Abstract
Evidence for the involvement of N6-Methyladenosine (m6A) modification in the etiology and progression of liver fibrosis has emerged and holds promise as a therapeutic target. Insulin-like growth factor 2 (IGF2) mRNA-binding protein 2 (IGF2BP2) is a newly identified m6A-binding protein that functions to enhance mRNA stability and translation. However, its role as an m6A-binding protein in liver fibrosis remains elusive. Here, we observed that IGF2BP2 is highly expressed in liver fibrosis and activated hepatic stellate cells (HSCs), and inhibition of IGF2BP2 protects against HSCs activation and liver fibrogenesis. Mechanistically, as an m6A-binding protein, IGF2BP2 regulates the expression of Aldolase A (ALDOA), a key target in the glycolytic metabolic pathway, which in turn regulates HSCs activation. Furthermore, we observed that active glycolytic metabolism in activated HSCs generates large amounts of lactate as a substrate for histone lactylation. Importantly, histone lactylation transforms the activation phenotype of HSCs. In conclusion, our findings reveal the essential role of IGF2BP2 in liver fibrosis by regulating glycolytic metabolism and highlight the potential of targeting IGF2BP2 as a therapeutic for liver fibrosis.
Collapse
Affiliation(s)
- Yongqiang Zhou
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Jiexi Yan
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Precision Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China
| | - He Huang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Lu Liu
- Department of Pediatrics, The First Hospital of Lanzhou University, Lanzhou, China
| | - Longfei Ren
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Jinjing Hu
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China
| | - Xiaoxu Jiang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yan Zheng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Lingcong Xu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Fupeng Zhong
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Xun Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.
- Precision Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China.
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China.
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China.
| |
Collapse
|
36
|
Shu G, Lei X, Li G, Zhang T, Wang C, Song A, Yu H, Wang X, Deng X. Ergothioneine suppresses hepatic stellate cell activation via promoting Foxa3-dependent potentiation of the Hint1/Smad7 cascade and improves CCl 4-induced liver fibrosis in mice. Food Funct 2023; 14:10591-10604. [PMID: 37955610 DOI: 10.1039/d3fo03643j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Ergothioneine (EGT) is a bioactive compound derived from certain edible mushrooms. The activation of hepatic stellate cells (HSCs) is critically involved in the etiology of liver fibrosis (LF). Here, we report that in LX-2 HSCs, EGT upregulates the expression of Hint1 and Smad7 and suppresses their activation provoked by TGFβ1. The EGT-triggered inhibition of HSC activation is abolished by knocking down the expression of Hint1. Overexpression of Hint1 increases Smad7 and represses TGFβ1-provoked activation of LX-2 HSCs. In silico predictions unveiled that in the promoter region of the human Hint1 gene, there are two conserved cis-acting elements that have the potential to interact with the transcription factor Foxa3 termed hFBS1 and hFBS2, respectively. The knockdown of Foxa3 obviously declined Hint1 expression at both mRNA and protein levels. Transfection of Foxa3 or EGT treatment increased the activity of the luciferase reporter driven by the Hint1 promoter in an hFBS2-dependent manner. The knockdown of Foxa3 eliminated EGT-mediated upregulation of Hint1 promoter activity. Additionally, EGT triggered the nuclear translocation of Foxa3 without obviously affecting its expression level. Molecular docking analysis showed that EGT has the potential to directly interact with the Foxa3 protein. Moreover, Foxa3 played a critical role in EGT-mediated hepatoprotection. EGT modulated the Foxa3/Hint1/Smad7 signaling in mouse primary HSCs and inhibited their activation. The gavage of EGT considerably relieved CCl4-induced LF in mice. Our data provide new insights into the anti-LF activity of EGT. Mechanistically, EGT triggers the nuclear translocation of Foxa3 in HSCs, which promotes Hint1 transcription and subsequently elevates Smad7.
Collapse
Affiliation(s)
- Guangwen Shu
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Hubei, China.
| | - Xiao Lei
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Hubei, China.
| | - Guangqiong Li
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Hubei, China.
| | - Tiantian Zhang
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Hubei, China.
| | - Chuo Wang
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Hubei, China.
| | - Anning Song
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Hubei, China.
| | - Huifan Yu
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiaoming Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Xukun Deng
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Hubei, China.
| |
Collapse
|
37
|
Liu TY, Liao CC, Chang YS, Chen YC, Chen HD, Lai IL, Peng CY, Chung CC, Chou YP, Tsai FJ, Jeng LB, Chang JG. Identification of 13 Novel Loci in a Genome-Wide Association Study on Taiwanese with Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:16417. [PMID: 38003606 PMCID: PMC10671380 DOI: 10.3390/ijms242216417] [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: 10/02/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Liver cancer is caused by complex interactions among genetic factors, viral infection, alcohol abuse, and metabolic diseases. We conducted a genome-wide association study and polygenic risk score (PRS) model in Taiwan, employing a nonspecific etiology approach, to identify genetic risk factors for hepatocellular carcinoma (HCC). Our analysis of 2836 HCC cases and 134,549 controls revealed 13 novel associated loci such as the FAM66C gene, noncoding genes, liver-fibrosis-related genes, metabolism-related genes, and HCC-related pathway genes. We incorporated the results from the UK Biobank and Japanese database into our study for meta-analysis to validate our findings. We also identified specific subtypes of the major histocompatibility complex that influence both viral infection and HCC progression. Using this data, we developed a PRS to predict HCC risk in the general population, patients with HCC, and HCC-affected families. The PRS demonstrated higher risk scores in families with multiple HCCs and other cancer cases. This study presents a novel approach to HCC risk analysis, identifies seven new genes associated with HCC development, and introduces a reproducible PRS model for risk assessment.
Collapse
Affiliation(s)
- Ting-Yuan Liu
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan; (T.-Y.L.); (C.-C.L.); (Y.-S.C.); (Y.-C.C.); (H.-D.C.); (I.-L.L.); (C.-C.C.); (Y.-P.C.)
- Million-Person Precision Medicine Initiative, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| | - Chi-Chou Liao
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan; (T.-Y.L.); (C.-C.L.); (Y.-S.C.); (Y.-C.C.); (H.-D.C.); (I.-L.L.); (C.-C.C.); (Y.-P.C.)
| | - Ya-Sian Chang
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan; (T.-Y.L.); (C.-C.L.); (Y.-S.C.); (Y.-C.C.); (H.-D.C.); (I.-L.L.); (C.-C.C.); (Y.-P.C.)
| | - Yu-Chia Chen
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan; (T.-Y.L.); (C.-C.L.); (Y.-S.C.); (Y.-C.C.); (H.-D.C.); (I.-L.L.); (C.-C.C.); (Y.-P.C.)
- Million-Person Precision Medicine Initiative, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| | - Hong-Da Chen
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan; (T.-Y.L.); (C.-C.L.); (Y.-S.C.); (Y.-C.C.); (H.-D.C.); (I.-L.L.); (C.-C.C.); (Y.-P.C.)
- Department of Laboratory Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - I-Lu Lai
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan; (T.-Y.L.); (C.-C.L.); (Y.-S.C.); (Y.-C.C.); (H.-D.C.); (I.-L.L.); (C.-C.C.); (Y.-P.C.)
| | - Cheng-Yuan Peng
- Department of Internal Medicine, Section of Hepatobiliary Tract, China Medical University Hospital, Taichung 40447, Taiwan;
| | - Chin-Chun Chung
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan; (T.-Y.L.); (C.-C.L.); (Y.-S.C.); (Y.-C.C.); (H.-D.C.); (I.-L.L.); (C.-C.C.); (Y.-P.C.)
| | - Yu-Pao Chou
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan; (T.-Y.L.); (C.-C.L.); (Y.-S.C.); (Y.-C.C.); (H.-D.C.); (I.-L.L.); (C.-C.C.); (Y.-P.C.)
| | - Fuu-Jen Tsai
- Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
- School of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
- Division of Pediatric Genetics, Children’s Hospital of China Medical University, Taichung 40447, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41354, Taiwan
| | - Long-Bin Jeng
- Department of Surgery, Section of Hepatobiliary Tract, China Medical University Hospital, Taichung 40447, Taiwan;
| | - Jan-Gowth Chang
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
38
|
Jiang A, Liu J, Wang Y, Zhang C. cGAS-STING signaling pathway promotes hypoxia-induced renal fibrosis by regulating PFKFB3-mediated glycolysis. Free Radic Biol Med 2023; 208:516-529. [PMID: 37714438 DOI: 10.1016/j.freeradbiomed.2023.09.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 09/17/2023]
Abstract
Hypoxia has long been considered to play an active role in the progression of fibrosis in chronic kidney disease, but its specific mechanism is not fully understood. The stimulator of interferon genes (STING) has been a research hotspot in the fields of tumor, immunity, and infection in recent years, and its role in immune and inflammatory responses related to kidney disease has gradually attracted attention. This study mainly explores the role and mechanism of STING in hypoxia-related renal fibrosis. To address this issue, we stimulated human proximal tubular epithelial (HK-2) cells with hypoxia for 48 h to construct cell models. Meanwhile, C57BL/6J male mice were used to establish a renal fibrosis model induced by renal ischemia-reperfusion injury (IRI). In our present study, we found that the GMP-AMP synthase (cGAS)-STING signaling pathway can promote the progression of renal fibrosis after hypoxic exposure, and this effect is closely related to 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase 3 (PFKFB3)-mediated glycolysis. Furthermore, inhibition of both STING and its downstream interferon regulatory factor 3 (IRF3) reversed elevated PFKFB3 expression, thereby attenuating hypoxia-induced renal fibrosis. Taken together, our data suggest that the cGAS-STING-IRF3-PFKFB3 signaling pathway activated under hypoxia may provide new ideas and targets for the treatment of early renal fibrosis.
Collapse
Affiliation(s)
- Anni Jiang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Liu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yumei Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
39
|
Gui R, Li W, Li Z, Wang H, Wu Y, Jiao W, Zhao G, Shen Y, Wang L, Zhang J, Chen S, Hao L, Cheng Y. Effects and potential mechanisms of IGF1/IGF1R in the liver fibrosis: A review. Int J Biol Macromol 2023; 251:126263. [PMID: 37567540 DOI: 10.1016/j.ijbiomac.2023.126263] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Liver fibrosis is a wound-healing response due to persistent liver damage and it may progress to cirrhosis and even liver cancer if no intervention is given. In the current cognition, liver fibrosis is reversible. So, it is of great significance to explore the related gene targets or biomarker for anti-fibrosis of liver. Insulin like growth factor 1 (IGF1) and IGF1 receptor (IGF1R) are mainly expressed in the liver tissues and play critical roles in the liver function. The present review summarized the role of IGF1/IGF1R and its signaling system in liver fibrosis and illustrated the potential mechanisms including DNA damage repair, cell senescence, lipid metabolism and oxidative stress that may be involved in this process according to the studies on the fibrosis of liver or other organs. In particular, the roles of IGF1 and IGF1R in DNA damage repair were elaborated, including membrane-localized and nucleus-localized IGF1R. In addition, for each of the potential mechanism in anti-fibrosis of liver, the signaling pathways of the IGF1/IGF1R mediated and the cell species in liver acted by IGF1 and IGF1R under different conditions were included. The data in this review will support for the study about the effect of IGF1/IGF1R on liver fibrosis induced by various factors, meanwhile, provide a basis for the study of liver fibrosis to focus on the communications between the different kinds of liver cells.
Collapse
Affiliation(s)
- Ruirui Gui
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Wanqiao Li
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Zhipeng Li
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Hongbin Wang
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Yuchen Wu
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Wenlin Jiao
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Gang Zhao
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Yannan Shen
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Luping Wang
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Jialu Zhang
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Sihan Chen
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Linlin Hao
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China.
| | - Yunyun Cheng
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China.
| |
Collapse
|
40
|
Chen L, Liang B, Xia S, Wang F, Li Z, Shao J, Zhang Z, Chen A, Zheng S, Zhang F. Emodin promotes hepatic stellate cell senescence and alleviates liver fibrosis via a nuclear receptor (Nur77)-mediated epigenetic regulation of glutaminase 1. Br J Pharmacol 2023; 180:2577-2598. [PMID: 37263753 DOI: 10.1111/bph.16156] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 02/13/2023] [Accepted: 05/23/2023] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND AND PURPOSE Senescence in hepatic stellate cells (HSCs) limits liver fibrosis. Glutaminolysis promotes HSC activation. Here, we investigated how emodin affected HSC senescence involving glutaminolysis. EXPERIMENTAL APPROACH Senescence, glutaminolysis metabolites, Nur77 nuclear translocation, glutaminase 1 (GLS1) promoter methylation and related signalling pathways were examined in human HSC-LX2 cells using multiple cellular and molecular approaches. Fibrotic mice with shRNA-mediated knockdown of Nur77 were treated with emodin-vitamin A liposome for investigating the mechanisms in vivo. Human fibrotic liver samples were examined to verify the clinical relevance. KEY RESULTS Emodin upregulated several key markers of senescence and inhibited glutaminolysis cascade in HSCs. Emodin promoted Nur77 nuclear translocation, and knockdown of Nur77 abolished emodin blockade of glutaminolysis and induction of HSC senescence. Mechanistically, emodin facilitated Nur77/DNMT3b interaction and increased GLS1 promoter methylation, leading to inhibited GLS1 expression and blockade of glutaminolysis. Moreover, the glutaminolysis intermediate α-ketoglutarate promoted extracellular signal-regulated kinase (ERK) phosphorylation, which in turn phosphorylated Nur77 and reduced its interaction with DNMT3b. This led to decreased GLS1 promoter methylation and increased GLS1 expression, forming an ERK/Nur77/glutaminolysis positive feedback loop. However, emodin repressed ERK phosphorylation and interrupted the feedback cascade, stimulating senescence in HSCs. Studies in mice showed that emodin-vitamin A liposome inhibited glutaminolysis and induced senescence in HSCs, and consequently alleviated liver fibrosis; but knockdown of Nur77 abrogated these beneficial effects. Similar alterations were validated in human fibrotic liver tissues. CONCLUSIONS AND IMPLICATIONS Emodin stimulated HSC senescence through interruption of glutaminolysis. HSC-targeted delivery of emodin represented a therapeutic option for liver fibrosis.
Collapse
Affiliation(s)
- Li Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Baoyu Liang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Siwei Xia
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feixia Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhanghao Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Anping Chen
- Department of Pathology, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
41
|
Zhao H, Zhu H, Zhang Y, Ding Y, Feng R, Li J, Ma T, Huang C. Lymphocyte-Specific Protein Tyrosine Kinase Contributes to Spontaneous Regression of Liver Fibrosis may by Interacting with Suppressor of Cytokine Signaling 1. Inflammation 2023; 46:1653-1669. [PMID: 37233920 DOI: 10.1007/s10753-023-01831-4] [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: 03/04/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/27/2023]
Abstract
Quiescent hepatic stellate cells (qHSCs), converted to myofibroblasts, produce fibrous scars, which is an essential event during liver fibrogenesis. Clinical and experimental fibrosis undergo remarkable regression when the underlying etiological agent is removed. Some myofibroblasts revert to an inactive phenotype (iHSCs) during the regression of fibrosis. However, the mechanisms underlying HSC activation and reversal remain unclear. The present study demonstrated that the expression of lymphocyte-specific protein tyrosine kinase (LCK) was increased in fibrotic livers but decreased after spontaneous recovery in vivo and in vitro, which was correlated with the expression of α-smooth muscle actin (α-SMA) and type I collagen (COL-1). Further investigation indicated that specific knockdown of LCK by a recombination adeno-associated virus 9 (rAAV9) in C57BL/6 mice ameliorated liver fibrosis. Co-incubation of TGF-β1-induced HSC-T6 cells with LCK-siRNA inhibited cell proliferation and activation. Overexpression of LCK inhibited activated HSCs going to inactivated phenotype. Interestingly, we found that LCK may interact with suppressor of cytokine signaling 1 (SOCS1) and may influence the expression of p-JAK1 and p-STAT1/3. These data suggest that LCK may play a regulatory role in liver fibrosis by inhibiting SOCS1, indicating that LCK is a potential therapeutic target for liver fibrosis treatment.
Collapse
Affiliation(s)
- Huizi Zhao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Hong Zhu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Yuan Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Yuhao Ding
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Rui Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Taotao Ma
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
42
|
He J, Fang B, Shan S, Li Q. Mechanical stiffness promotes skin fibrosis through Piezo1-mediated arginine and proline metabolism. Cell Death Discov 2023; 9:354. [PMID: 37752116 PMCID: PMC10522626 DOI: 10.1038/s41420-023-01656-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/08/2023] [Accepted: 09/18/2023] [Indexed: 09/28/2023] Open
Abstract
The increased mechanics of fibrotic skin tissue continuously regulate fibroblast functions such as survival and differentiation. Although all these processes consume metabolites, it is unclear whether and how cells adapt their metabolic activity to increased matrix stiffness. Here, we show that transferring mouse dermal fibroblasts from soft to stiff substrates causes an up-regulation of arginine and proline metabolism. Increased matrix stiffness stimulates the expression and activity of key metabolic enzymes, leading to the synthesis of L-proline, a major source of collagen. In addition, the novel mechanosensitive channel Piezo1 was identified as a key regulator of arginine and proline metabolism in fibroblasts under increased stiffness. Consistently, targeting Piezo1 to dermal fibroblasts in vivo effectively reduces fibrosis and arginine-proline metabolism in mouse skin. Therefore, mechanical stiffness is a critical environmental cue for fibroblast metabolism and skin fibrosis progression.
Collapse
Affiliation(s)
- Jiahao He
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China
| | - Bin Fang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China.
| | - Shengzhou Shan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China.
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China.
| |
Collapse
|
43
|
Zhang X, Zhao L, Ying K, Xu J, Huang Y, Zhu R, Ding Y, Cai W, Wu X, Miao D, Xu Q, Zeng Y, Yu F. TUG1 protects against ferroptosis of hepatic stellate cells by upregulating PDK4-mediated glycolysis. Chem Biol Interact 2023; 383:110673. [PMID: 37582412 DOI: 10.1016/j.cbi.2023.110673] [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: 05/14/2023] [Revised: 07/21/2023] [Accepted: 08/12/2023] [Indexed: 08/17/2023]
Abstract
The induction of ferroptosis in hepatic stellate cells (HSCs) has shown promise in reversing liver fibrosis. And ferroptosis has been confirmed to be associated with glycolysis. The objective of this study is to determine whether ferroptosis inhibition in HSCs, induced by elevation of recombinant pyruvate dehydrogenase kinase isozyme 4 (PDK4)-mediated glycolysis, could mediate the pathogenesis of liver fibrosis. Liver fibrosis was induced using CCl4, the level of which was assessed through histochemical staining. Lentivirus was used to modulate the expression of specific genes. And underlying mechanisms were explored using primary HSCs extracted from normal mice. The results confirmed that Taurine up-regulated gene 1 (TUG1) expression was upregulated in liver fibrotic tissues and HSCs, showing a positive correlation with fibrosis. In addition, TUG1 attenuated ferroptosis in HSCs by promoting PDK4-mediated glycolysis, thereby promoting the progression of liver fibrosis. Moreover, TUG1 was observed to impact HSCs activation, exacerbating liver fibrosis to some extent. In conclusion, our study revealed that TUG1 expression was elevated in mouse models of liver fibrosis and activated HSCs, which inhibited ferroptosis in HSCs through PDK4-mediated glycolysis. This finding may open up a new therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Xiangting Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Luying Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kanglei Ying
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jun Xu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yangjin Huang
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ruhuang Zhu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yinrong Ding
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weimin Cai
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiao Wu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dan Miao
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qian Xu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuan Zeng
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Fujun Yu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
44
|
Wu F, Wu F, Zhou Q, Liu X, Fei J, Zhang D, Wang W, Tao Y, Lin Y, Lin Q, Pan X, Sun K, Xie F, Bai L. A CCL2 +DPP4 + subset of mesenchymal stem cells expedites aberrant formation of creeping fat in humans. Nat Commun 2023; 14:5830. [PMID: 37730641 PMCID: PMC10511504 DOI: 10.1038/s41467-023-41418-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 09/04/2023] [Indexed: 09/22/2023] Open
Abstract
Creeping fat is a typical feature of Crohn's disease. It refers to the expansion of mesenteric adipose tissue around inflamed and fibrotic intestines and is associated with stricture formation and intestinal obstruction. In this study, we characterize creeping fat as pro-adipogenic and pro-fibrotic. Lipidomics analysis of Crohn's disease patients (sixteen males, six females) and healthy controls (five males, ten females) reveals abnormal lipid metabolism in creeping fat. Through scRNA-seq analysis on mesenteric adipose tissue from patients (five males, one female) and healthy controls (two females), we identify a CCL2+DPP4+ subset of mesenchymal stem cells that expands in creeping fat and expedites adipogenic differentiation into dystrophic adipocytes in response to CCL20+CD14+ monocytes and IL-6, leading to the formation of creeping fat. Ex vivo experiments (tissues from five males, one female) confirm that both CCL20+CD14+ monocytes and IL-6 activate DPP4+ mesenchymal stem cells towards a pro-adipogenic phenotype. This study provides a comprehensive investigation of creeping fat formation and offers a conceptual framework for discovering therapeutic targets for treatment of Crohn's disease.
Collapse
Affiliation(s)
- Fengfei Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fangting Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Zhou
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xi Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jieying Fei
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Da Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weidong Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Tao
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yubing Lin
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiaoqiao Lin
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Kai Sun
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fang Xie
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Lan Bai
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
45
|
Rho H, Terry AR, Chronis C, Hay N. Hexokinase 2-mediated gene expression via histone lactylation is required for hepatic stellate cell activation and liver fibrosis. Cell Metab 2023; 35:1406-1423.e8. [PMID: 37463576 PMCID: PMC11748916 DOI: 10.1016/j.cmet.2023.06.013] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/03/2023] [Accepted: 06/20/2023] [Indexed: 07/20/2023]
Abstract
Lactate was implicated in the activation of hepatic stellate cells (HSCs). However, the mechanism by which lactate exerts its effect remains elusive. Using RNA-seq and CUT&Tag chromatin profiling, we found that induction of hexokinase 2 (HK2) expression in activated HSCs is required for induced gene expression by histone lactylation but not histone acetylation. Inhibiting histone lactylation by Hk2 deletion or pharmacological inhibition of lactate production diminishes HSC activation, whereas exogenous lactate but not acetate supplementation rescues the activation phenotype. Thus, lactate produced by activated HSCs determines the HSC fate via histone lactylation. We found that histone acetylation competes with histone lactylation, which could explain why class I HDAC (histone deacetylase) inhibitors impede HSC activation. Finally, HSC-specific or systemic deletion of HK2 inhibits HSC activation and liver fibrosis in vivo. Therefore, we provide evidence that HK2 may be an effective therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Hyunsoo Rho
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Alexander R Terry
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Constantinos Chronis
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Nissim Hay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA; Research and Development Section, Jesse Brown VA Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
46
|
Chu YD, Chen CW, Lai MW, Lim SN, Lin WR. Bioenergetic alteration in gastrointestinal cancers: The good, the bad and the ugly. World J Gastroenterol 2023; 29:4499-4527. [PMID: 37621758 PMCID: PMC10445009 DOI: 10.3748/wjg.v29.i29.4499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/23/2023] [Accepted: 07/03/2023] [Indexed: 08/02/2023] Open
Abstract
Cancer cells exhibit metabolic reprogramming and bioenergetic alteration, utilizing glucose fermentation for energy production, known as the Warburg effect. However, there are a lack of comprehensive reviews summarizing the metabolic reprogramming, bioenergetic alteration, and their oncogenetic links in gastrointestinal (GI) cancers. Furthermore, the efficacy and treatment potential of emerging anticancer drugs targeting these alterations in GI cancers require further evaluation. This review highlights the interplay between aerobic glycolysis, the tricarboxylic acid (TCA) cycle, and oxidative phosphorylation (OXPHOS) in cancer cells, as well as hypotheses on the molecular mechanisms that trigger this alteration. The role of hypoxia-inducible transcription factors, tumor suppressors, and the oncogenetic link between hypoxia-related enzymes, bioenergetic changes, and GI cancer are also discussed. This review emphasizes the potential of targeting bioenergetic regulators for anti-cancer therapy, particularly for GI cancers. Emphasizing the potential of targeting bioenergetic regulators for GI cancer therapy, the review categorizes these regulators into aerobic glycolysis/ lactate biosynthesis/transportation and TCA cycle/coupled OXPHOS. We also detail various anti-cancer drugs and strategies that have produced pre-clinical and/or clinical evidence in treating GI cancers, as well as the challenges posed by these drugs. Here we highlight that understanding dysregulated cancer cell bioenergetics is critical for effective treatments, although the diverse metabolic patterns present challenges for targeted therapies. Further research is needed to comprehend the specific mechanisms of inhibiting bioenergetic enzymes, address side effects, and leverage high-throughput multi-omics and spatial omics to gain insights into cancer cell heterogeneity for targeted bioenergetic therapies.
Collapse
Affiliation(s)
- Yu-De Chu
- Liver Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Chun-Wei Chen
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Ming-Wei Lai
- Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Siew-Na Lim
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Wey-Ran Lin
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Department of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
47
|
Zeng W, Zhang W, Tse EHY, Liu J, Dong A, Lam KSW, Luan S, Kung WH, Chan TC, Cheung TH. Restoration of CPEB4 prevents muscle stem cell senescence during aging. Dev Cell 2023; 58:1383-1398.e6. [PMID: 37321216 DOI: 10.1016/j.devcel.2023.05.012] [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: 12/27/2021] [Revised: 03/24/2023] [Accepted: 05/19/2023] [Indexed: 06/17/2023]
Abstract
Age-associated impairments in adult stem cell functions correlate with a decline in somatic tissue regeneration capacity. However, the mechanisms underlying the molecular regulation of adult stem cell aging remain elusive. Here, we provide a proteomic analysis of physiologically aged murine muscle stem cells (MuSCs), illustrating a pre-senescent proteomic signature. During aging, the mitochondrial proteome and activity are impaired in MuSCs. In addition, the inhibition of mitochondrial function results in cellular senescence. We identified an RNA-binding protein, CPEB4, downregulated in various aged tissues, which is required for MuSC functions. CPEB4 regulates the mitochondrial proteome and activity through mitochondrial translational control. MuSCs devoid of CPEB4 induced cellular senescence. Importantly, restoring CPEB4 expression rescued impaired mitochondrial metabolism, improved geriatric MuSC functions, and prevented cellular senescence in various human cell lines. Our findings provide the basis for the possibility that CPEB4 regulates mitochondrial metabolism to govern cellular senescence, with an implication of therapeutic intervention for age-related senescence.
Collapse
Affiliation(s)
- Wenshu Zeng
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Wenxin Zhang
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Erin H Y Tse
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Jing Liu
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Anqi Dong
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Kim S W Lam
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Shaoyuan Luan
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Wai Hing Kung
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Tsz Ching Chan
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, Shenzhen-Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
48
|
Zhang Y, Zhang Y, Chen T, Lin Y, Gong J, Xu Q, Wang J, Li J, Meng Y, Li Y, Li X. Caveolin-1 depletion attenuates hepatic fibrosis via promoting SQSTM1-mediated PFKL degradation in HSCs. Free Radic Biol Med 2023; 204:95-107. [PMID: 37116593 DOI: 10.1016/j.freeradbiomed.2023.04.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 04/30/2023]
Abstract
The key glycolytic enzyme phosphofructokinase (PFK) is responsible for maintaining glycolytic stability and an important energy source for activating hepatic stellate cells (HSCs). However, its regulation in activated HSCs remains unclear. Caveolin-1 (Cav1), a major constituent of caveolae, has emerged as a key target for triggering glycolysis. However, the relationship between Cav1 and glycolysis during HSC activation is not well established. In this study, Cav1 was upregulated in mouse and human fibrotic liver tissues. We concluded that HSC-specific Cav1 knockdown markedly alleviates liver injury and fibrosis. Mechanistically, Cav1 was elevated during primary mouse HSC activation, competing with SQSTM1 for the regulatory subunit of PFK liver type and inhibiting the SQSTM1-mediated autophagy-independent lysosomal degradation pathway to sustain HSC activation. We also identified the heptapeptide alamandine as a promising therapeutic agent that downregulates Cav1 protein levels via proteasomal degradation and may impair glycolysis. Our study provides evidence of the crucial role and mechanism of Cav1 in the glucose metabolic network in HSCs and highlights Cav1 as a critical therapeutic target for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Yijie Zhang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Tingting Chen
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Ying Lin
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Jiacheng Gong
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Qihan Xu
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Jun Wang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Jierui Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Ying Meng
- Department of Respiratory Diseases, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| | - Yang Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| |
Collapse
|
49
|
Jerez S, Gao J, Kostallari E. Editorial: Chronic Liver Disease: New Targets and New Mechanisms, Volume II. Front Mol Biosci 2023; 10:1237824. [PMID: 37533679 PMCID: PMC10392929 DOI: 10.3389/fmolb.2023.1237824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 07/11/2023] [Indexed: 08/04/2023] Open
Affiliation(s)
- Sofia Jerez
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, China
| | - Jinhang Gao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Enis Kostallari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, China
| |
Collapse
|
50
|
Yang Q, Zong X, Zhuang L, Pan R, Tudi X, Fan Q, Tao R. PFKFB3 Inhibitor 3PO Reduces Cardiac Remodeling after Myocardial Infarction by Regulating the TGF-β1/SMAD2/3 Pathway. Biomolecules 2023; 13:1072. [PMID: 37509108 PMCID: PMC10377206 DOI: 10.3390/biom13071072] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Adverse cardiac remodeling, including cardiac fibrosis, after myocardial infarction (MI) is a major cause of long-term heart failure. 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), an enzyme that regulates glucose metabolism, also plays an important role in various fibrotic and cardiovascular diseases. However, its effects on MI remain unknown. Here, PFKFB3 inhibitor 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) and a permanent left anterior descending ligation mouse model were used to explore the functional role of PFKFB3 in MI. We showed that PFKFB3 expression increased significantly in the area of cardiac infarction during the early phase after MI, peaking on day 3. 3PO treatment markedly improved cardiac function, accompanied by decreased infarction size and collagen density in the infarct area. Meanwhile, 3PO attenuated cardiac fibrosis after MI by reducing the expression of collagen and fibronectin in murine hearts. Notably, 3PO reduced PFKFB3 expression and inhibited the transforming growth factor-beta 1/mothers against the decapentaplegic homolog 2/3 (TGF-β1/SMAD2/3) signaling pathway to inhibit cardiac fibrosis after MI. Moreover, PFKFB3 expression in neonatal rat cardiac fibroblasts (NRCFs) increased significantly after MI and under hypoxia, whereas 3PO alleviated the migratory capacity and activation of NRCFs induced by TGF-β1. In conclusion, 3PO effectively reduced fibrosis and improved adverse cardiac remodeling after MI, suggesting PFKFB3 inhibition as a novel therapeutic strategy to reduce the incidence of chronic heart failure following MI.
Collapse
Affiliation(s)
- Qian Yang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institution of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiao Zong
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institution of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lingfang Zhuang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institution of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Roubai Pan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xierenayi Tudi
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institution of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qin Fan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rong Tao
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|