1
|
Wang X, Lu Y, Li X, Wang M, Liu X, Huang H, Cao W, Liu Y, Ren L, Xu Y. Integrated metabolomics and transcriptomics analyses for understanding the mechanism underlying amantadine-induced toxicity in Laminaria japonica. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137616. [PMID: 39965339 DOI: 10.1016/j.jhazmat.2025.137616] [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: 10/12/2024] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025]
Abstract
The antiviral agent, amantadine, is widely present in marine ecosystems and poses a significant threat to marine organisms. However, studies on the toxicity of amantadine across the full life cycle of the brown alga Laminaria japonica, particularly during the microscopic gametophyte stage, remain lacking. A comprehensive approach combing biochemical analyses and multi-omics techniques was employed to investigate the mechanisms underlying amantadine-induced toxicity in L. japonica gametophytes. The development rate of algal cells was less than 3 % from 103 to 107 ng/L of amantadine. In total, 1049 differentially expressed genes and 215-231 differential metabolites were detected, with the majority involved in amino acid, lipid, and carbohydrate metabolism. Integrated analysis showed that alginate biosynthesis and glycerophospholipid metabolism were affected, suggesting damage to the cell wall and membrane structure. Key genes (e.g., SOD2) and metabolites (e.g., arachidonic acid and α-linolenic acid), associated with the antioxidant system and arachidonic acid metabolism, were identified, leading to oxidative stress in the algae. Furthermore, the downregulation of genes and metabolites involved in porphyrin metabolism, photosynthesis, carbon fixation, glycolysis, and the pentose phosphate pathway may inhibit ATP supply and NADPH generation, negatively affecting metabolic processes and inhibiting algal cell growth. In contrast to disrupting protein synthesis in juvenile sporophytes, amantadine primarily interferes with photosynthesis and carbohydrate metabolism in gametophytes. These findings offer new insights into the mechanisms by which amantadine impedes the growth and metabolism of algae throughout their life cycle in aquatic ecosystems.
Collapse
Affiliation(s)
- Xiaohan Wang
- Yantai Key Laboratory of Quality and Safety Control and Deep Processing of Marine Food, Shandong Key Laboratory of Marine Ecological Restoration, Observation and Research Station of Laizhou Bay Marine Ecosystem, MNR, Shandong Marine Resource and Environment Research Institute, Yantai 264006, China
| | - Yao Lu
- Yantai Key Laboratory of Quality and Safety Control and Deep Processing of Marine Food, Shandong Key Laboratory of Marine Ecological Restoration, Observation and Research Station of Laizhou Bay Marine Ecosystem, MNR, Shandong Marine Resource and Environment Research Institute, Yantai 264006, China
| | - Xiaojie Li
- Shandong Oriental Ocean Sci-Tech Co., Ltd., Shandong Technology Innovation Center of Algae and Sea Cucumber, Yantai 264003, China
| | - Minglei Wang
- Yantai Key Laboratory of Quality and Safety Control and Deep Processing of Marine Food, Shandong Key Laboratory of Marine Ecological Restoration, Observation and Research Station of Laizhou Bay Marine Ecosystem, MNR, Shandong Marine Resource and Environment Research Institute, Yantai 264006, China
| | - Xiaojing Liu
- Yantai Key Laboratory of Quality and Safety Control and Deep Processing of Marine Food, Shandong Key Laboratory of Marine Ecological Restoration, Observation and Research Station of Laizhou Bay Marine Ecosystem, MNR, Shandong Marine Resource and Environment Research Institute, Yantai 264006, China
| | - Hui Huang
- Yantai Key Laboratory of Quality and Safety Control and Deep Processing of Marine Food, Shandong Key Laboratory of Marine Ecological Restoration, Observation and Research Station of Laizhou Bay Marine Ecosystem, MNR, Shandong Marine Resource and Environment Research Institute, Yantai 264006, China
| | - Wei Cao
- Yantai Key Laboratory of Quality and Safety Control and Deep Processing of Marine Food, Shandong Key Laboratory of Marine Ecological Restoration, Observation and Research Station of Laizhou Bay Marine Ecosystem, MNR, Shandong Marine Resource and Environment Research Institute, Yantai 264006, China
| | - Yongchun Liu
- Yantai Key Laboratory of Quality and Safety Control and Deep Processing of Marine Food, Shandong Key Laboratory of Marine Ecological Restoration, Observation and Research Station of Laizhou Bay Marine Ecosystem, MNR, Shandong Marine Resource and Environment Research Institute, Yantai 264006, China
| | - Lihua Ren
- Yantai Key Laboratory of Quality and Safety Control and Deep Processing of Marine Food, Shandong Key Laboratory of Marine Ecological Restoration, Observation and Research Station of Laizhou Bay Marine Ecosystem, MNR, Shandong Marine Resource and Environment Research Institute, Yantai 264006, China
| | - Yingjiang Xu
- Yantai Key Laboratory of Quality and Safety Control and Deep Processing of Marine Food, Shandong Key Laboratory of Marine Ecological Restoration, Observation and Research Station of Laizhou Bay Marine Ecosystem, MNR, Shandong Marine Resource and Environment Research Institute, Yantai 264006, China.
| |
Collapse
|
2
|
Pruckner F, Morelli L, Patwari P, Fabris M. Remodeling of the terpenoid metabolism during prolonged phosphate depletion in the marine diatom Phaeodactylum tricornutum. JOURNAL OF PHYCOLOGY 2025; 61:512-528. [PMID: 40234016 PMCID: PMC12168105 DOI: 10.1111/jpy.70014] [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] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/03/2025] [Accepted: 03/14/2025] [Indexed: 04/17/2025]
Abstract
Terpenoids are a diverse class of naturally occurring organic compounds, which derive from five-carbon isoprene units and play crucial roles in physiology, ecological interactions such as defense mechanisms, or adaptation to environmental stresses. In Phaeodactylum tricornutum, some of the most important isoprenoids are sterols and pigments, derived from precursors of the cytosolic mevalonate and the plastidial methyl-erythritol 4-phosphate pathway, respectively. However, the regulation of isoprenoid metabolism in P. tricornutum has not yet been characterized, presenting a major gap in our understanding of its ecological functions and adaptations. By leveraging metabolic, photosynthetic, and transcriptomic analyses, we characterized the dynamic remodeling of the isoprenoid pathways during prolonged nutrient stress in wild-type diatoms. We observed the down-regulation of the methylerythritol 4-phosphate and pigment biosynthesis pathways and the upregulation of key genes in the mevalonate and sterol biosynthesis pathways. At the metabolite level, we observed an overall decrease in pigment and no changes in sterol levels. Using a genetically engineered diatom strain to produce a heterologous monoterpenoid to monitor the availability of one of the main terpenoid precursors, geranyl diphosphate (GPP), we suggest that cytosolic GPP pools increase during prolonged phosphate depletion. Our results have demonstrated how the biosynthesis of isoprenoid metabolites and the pools of prenyl phosphate are vastly remodeled during phosphate depletion. We anticipate that the knowledge generated in this study can serve as a foundation for understanding ecological responses and adaptations of diatoms to nutrient stress, contributing to our broader comprehension of marine ecosystem dynamics and design strategies for producing high-value compounds in diatoms.
Collapse
Affiliation(s)
- Florian Pruckner
- SDU Biotechnology, Department of Green TechnologyUniversity of Southern DenmarkOdense MDenmark
| | - Luca Morelli
- SDU Biotechnology, Department of Green TechnologyUniversity of Southern DenmarkOdense MDenmark
| | - Payal Patwari
- SDU Biotechnology, Department of Green TechnologyUniversity of Southern DenmarkOdense MDenmark
| | - Michele Fabris
- SDU Biotechnology, Department of Green TechnologyUniversity of Southern DenmarkOdense MDenmark
- SDU Climate Cluster, Faculty of ScienceUniversity of Southern DenmarkOdense MDenmark
| |
Collapse
|
3
|
Wang C, Feng H, Zhao J, Liu C, Wang X, Zhou H, Mai K, He G. Oxidative stress and metabolic perturbations unravel the molecular basis of high dietary poultry by-product meal-induced growth impairment and inflammation response in Litopenaeusvannamei. FISH & SHELLFISH IMMUNOLOGY 2025; 164:110433. [PMID: 40404030 DOI: 10.1016/j.fsi.2025.110433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 04/26/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
The pacific white shrimp, Litopenaeus vannamei (L. vannamei) is one of the most widely farmed shrimp species in global aquaculture. The scarcity of fishmeal as a component of aquafeeds poses a considerable challenge to the sustainable development of the aquaculture industry. This study aimed to assess the feasibility of poultry by-product meal (PBM) as an alternative to fishmeal of L. vannamei diets by assessing its impact on growth performance, immune response, postprandial metabolism and signaling pathways. Six diets were formulated to be iso-nitrogenous (42 %) and iso-lipidic (7 %), with 0, 8 %, 16 %, 25 %, 33 %, and 50 % fishmeal replaced by poultry by-product meal (marked as CON, PBM8, PBM16, PBM25, PBM33, PBM50). After an 8 weeks of growth trial, results showed that dietary PBM replacement up to 16 % didn't negatively affect growth performance of L. vannamei. However, PBM replacement at 16 % or higher led to significant reductions in lipase and trypsin activity, deteriorated gut morphology, and disrupted the balance of immune-related gene expression and phosphorylation of Nf-κB in the intestine or hepatopancreas. Mechanically, oxidative stress progressively increased in the hepatopancreas with higher dietary PBM levels, inducing ferroptosis and cuproptosis while leaving apoptosis and autophagy unaffected. Postprandial mTORC1 signaling in muscle was significantly suppressed by higher PBM levels, impairing protein synthesis. Furthermore, high PBM replacement disrupted postprandial energy metabolism, affecting glycolysis, gluconeogenesis, and TCA cycle pathways. Taken together, these results shed light on the molecular, immune and metabolic impacts of dietary PBM replacement in L. vannamei, enhancing comprehension of the relationship between dietary protein sources and shrimp physiological health.
Collapse
Affiliation(s)
- Chenxi Wang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Huali Feng
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Jican Zhao
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Chengdong Liu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China.
| | - Xuan Wang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Huihui Zhou
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Kangsen Mai
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Gen He
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China; Key Laboratory of Aquaculture Nutrition and Feeds, Ministry of Agriculture, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
4
|
Li J, Li Y, Fu L, Chen H, Du F, Wang Z, Zhang Y, Huang Y, Miao J, Xiao Y. Targeting ncRNAs to overcome metabolic reprogramming‑mediated drug resistance in cancer (Review). Int J Oncol 2025; 66:35. [PMID: 40116120 PMCID: PMC12002672 DOI: 10.3892/ijo.2025.5741] [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: 01/22/2025] [Accepted: 03/07/2025] [Indexed: 03/23/2025] Open
Abstract
The emergence of resistance to antitumor drugs in cancer cells presents a notable obstacle in cancer therapy. Metabolic reprogramming is characterized by enhanced glycolysis, disrupted lipid metabolism, glutamine dependence and mitochondrial dysfunction. In addition to promoting tumor growth and metastasis, metabolic reprogramming mediates drug resistance through diverse molecular mechanisms, offering novel opportunities for therapeutic intervention. Non‑coding RNAs (ncRNAs), a diverse class of RNA molecules that lack protein‑coding function, represent a notable fraction of the human genome. Due to their distinct expression profiles and multifaceted roles in various cancers, ncRNAs have relevance in cancer pathophysiology. ncRNAs orchestrate metabolic abnormalities associated with drug resistance in cancer cells. The present review provides a comprehensive analysis of the mechanisms by which metabolic reprogramming drives drug resistance, with an emphasis on the regulatory roles of ncRNAs in glycolysis, lipid metabolism, mitochondrial dysfunction and glutamine metabolism. Furthermore, the present review aimed to discuss the potential of ncRNAs as biomarkers for predicting chemotherapy responses, as well as emerging strategies to target ncRNAs that modulate metabolism, particularly in the context of combination therapy with anti‑cancer drugs.
Collapse
Affiliation(s)
- Junxin Li
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yanyu Li
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Lin Fu
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Huiling Chen
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital of Southwest Medical University, Meishan, Sichuan 64200, P.R. China
| | - Zhongshu Wang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yan Zhang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yu Huang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Jidong Miao
- Department of Oncology, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yi Xiao
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| |
Collapse
|
5
|
Saleh RO, Aboqader Al-Aouadi RF, Almuzaini NA, Uthirapathy S, Sanghvi G, Soothwal P, Arya R, Bareja L, Mohamed Abdelgawwad El-Sehrawy AA, Hulail HM. Glucose metabolism is controlled by non-coding RNAs in autoimmune diseases; a glimpse into immune system dysregulation. Hum Immunol 2025; 86:111269. [PMID: 39999745 DOI: 10.1016/j.humimm.2025.111269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025]
Abstract
The immune system accidentally targets the body's tissues, causing inflammation and tissue damage, the root causes of autoimmune illnesses. In recent studies, non-coding RNAs have been shown to significantly control gene expression and metabolic pathways linked to autoimmune diseases. This review investigates the effects of non-coding RNA on glucose metabolism, a route frequently dysregulated in autoimmune illnesses such as multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, and diabetes. We review how non-coding RNA affects immune cell activity modulation, glucose absorption, glycolysis, and other metabolic processes critical to immune function. We also investigate the possibility of using non-coding RNA-mediated metabolic pathway targeting as a new therapeutic approach to treat autoimmune disorders. By clarifying the complex interplay of non-coding RNA, glucose metabolism, and immune dysregulation, this study endeavors to enhance comprehension of autoimmune etiology and facilitate the creation of focused therapies.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al Maarif, Anbar, Iraq
| | | | | | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003, Gujarat, India
| | - Pradeep Soothwal
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Renu Arya
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Lakshay Bareja
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India
| | | | - Hanen Mahmod Hulail
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
6
|
Zhang Y, Jin H, Jia W, Liu Y, Wang Y, Xue S, Liu Y, Hao H. Ermiao San attenuating rheumatoid arthritis via PI3K/AKT/mTOR signaling activate HIF-1α induced glycolysis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119615. [PMID: 40081512 DOI: 10.1016/j.jep.2025.119615] [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: 12/10/2024] [Revised: 01/14/2025] [Accepted: 03/08/2025] [Indexed: 03/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a classic formula, Ermiao San (EMS) characterized by its less medicinal flavor and strong potency had been proven to be effective and safe in the treatment of rheumatoid arthritis (RA) during clinical experience and our previous research. AIM OF THE STUDY The therapeutic characteristics of multi-component and multi-target of traditional Chinese medicine prompted us to further investigated the effective compounds of EMS, and evaluated its potential mechanisms in treating RA. MATERIALS AND METHODS Ultra-high-performance liquid chromatography high-resolution mass spectrometry (UPLC-HRMS) was used to analyze the primary absorption components of EMS in rat serum, with secondary mass spectrometry used to assist in identifying the structures of the compounds. Open field experiments, H&E staining, safranin-O-turquoise staining, ELISA, and other methods were applied to verify the alleviating effects of EMS on exercise capacity, inflammation, and cartilage damage in CIA rats. The RA-FLS model was established using TNF-α, and observed the effects of EMS on cell migration and invasion were observed through wound healing and transwell assays. In addition, immunohistochemistry and western blotting were employed to investigate the PI3K/AKT/mTOR/HIF-1α pathway both in vivo and in vitro. RESULTS Seventeen compounds were identified in rat serum, which were considered as active ingredients involved in the improvement of RA by EMS. Furthermore, EMS demonstrated the outstanding anti-RA ability, as evidenced by the improvement in foot swelling and arthritis scores, alleviation of pathological changes in joint tissue, inhibition of inflammatory factors, and restoration of exercise ability. In vivo data showed that EMS reduced joint injury through the PI3K/AKT/mTOR/HIF-1α signaling pathway. In vitro studies indicated that TNF-α induced the expression of Glut1 and HK2 proteins, accelerated the glycolysis rate, and promoted migration and invasion of RA-FLS cells, leading to adverse outcomes. However, EMS regulated the expression of glycolysis-related molecules, HK2 and Glut1 through the PI3K/AKT/mTOR/HIF-1α pathway, thereby inhibiting inflammation, migration, and invasion of RA-FLS cells. CONCLUSION The beneficial effects of EMS in CIA rats can be attributed to the inhibition of glycolysis in synovial fibroblasts via the PI3K/AKT/mTOR/HIF-1α pathway. This finding further enriches our understanding of the mechanisms by which EMS contributes to the treatment of RA.
Collapse
Affiliation(s)
- Yumeng Zhang
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China; Basic Laboratory of Integrated Traditional Chinese and Western Medicine, Shanxi University of Chinese Medicine, Jinzhong, 030619, China; Engineering Research Center of Cross Innovation for Chinese Traditional Medicine of Shanxi Province, Jinzhong, 030619, China
| | - Haizhu Jin
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Wenyue Jia
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Yuqi Liu
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Yuru Wang
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Shuyan Xue
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Yang Liu
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China; Basic Laboratory of Integrated Traditional Chinese and Western Medicine, Shanxi University of Chinese Medicine, Jinzhong, 030619, China; Engineering Research Center of Cross Innovation for Chinese Traditional Medicine of Shanxi Province, Jinzhong, 030619, China.
| | - Huiqin Hao
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China; Basic Laboratory of Integrated Traditional Chinese and Western Medicine, Shanxi University of Chinese Medicine, Jinzhong, 030619, China; Engineering Research Center of Cross Innovation for Chinese Traditional Medicine of Shanxi Province, Jinzhong, 030619, China.
| |
Collapse
|
7
|
Zhou P, Liu X, Tian Y, Ren S, Liang H. High-throughput metabolomics exploring the pharmacological effects and mechanism of icariin on rheumatoid arthritis rat based on ultrahigh-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry. Front Mol Biosci 2025; 12:1514882. [PMID: 40270592 PMCID: PMC12015166 DOI: 10.3389/fmolb.2025.1514882] [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: 10/21/2024] [Accepted: 03/07/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction Metabolomics could provide insights into the pharmacological effects and action mechanisms of drugs through assessment of the changes in relevant biomarkers and biological pathways. Icariin (ICA) is a promising ffavonoid compound known to have significant anticancer activity; however, the pharmacological mechanisms of ICA in the treatment of rheumatoid arthritis (RA) need to be explored further. Methods The changes in the metabolic profiles of serum samples were revealed using non-targeted metabolomics based on ultrahigh-performance liquid chromatography coupled with quadrupole time-of-fight mass spectrometry. Tissue histopathology, physical parameters, and biochemical indicators were also measured and analyzed to reveal the mechanisms of ICA in the treatment of RA. Results and discussion Thirty-one potential biomarkers were identified to highlight the metabolic disorders in an RA animal model, out of which twenty-three were regulated by ICA treatment. These biomarkers were mainly involved in alanine, aspartate, and glutamate metabolism; arachidonic acid metabolism; citrate cycle; pyruvate metabolism; and glycolysis/gluconeogenesis pathways. The anticancer mechanism of ICA on RA may be attributed to amelioration of the amino acid metabolism, unsaturated fatty acid metabolism, citrate cycle, pyruvate metabolism, and others, which in turn regulate the oxidative stress state and inflammatory effects. Thus, metabolomics is a promising approach for revealing the biomarker distribution and pathways of RA to determine the effects and mechanisms of ICA, which can benefit the development of natural medicines.
Collapse
Affiliation(s)
- Peng Zhou
- School of Continuing Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xixi Liu
- Beijing Mentougou District of Traditional Chinese Medicine, Beijing, China
| | - Yushi Tian
- Department of Chinese Formulae, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shouze Ren
- Department of Chinese Formulae, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hua Liang
- Department of Chinese Formulae, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
8
|
Xu X, Xu P, Shen G, Peng X, Liu Z, Chen C, Yu W, Su Z, Lin J, Zheng G, Ye G, Wang P, Xie Z, Wu Y, Shen H, Li J. Targeting macrophage polarization by inhibiting Pim2 alleviates inflammatory arthritis via metabolic reprogramming. Cell Mol Immunol 2025; 22:418-436. [PMID: 40000906 PMCID: PMC11955556 DOI: 10.1038/s41423-025-01268-9] [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: 07/03/2024] [Revised: 01/25/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Macrophage polarization and energy metabolic reprogramming play pivotal roles in the onset and progression of inflammatory arthritis. Moreover, although previous studies have reported that the proviral integration of Moloney virus 2 (Pim2) kinase is involved in various cancers through the mediation of aerobic glycolysis in cancer cells, its role in inflammatory arthritis remains unclear. In this study, we demonstrated that multiple metabolic enzymes are activated upon Pim2 upregulation during M1 macrophage polarization. Specifically, Pim2 directly phosphorylates PGK1-S203, PDHA1-S300, and PFKFB2-S466, thereby promoting glycolytic reprogramming. Pim2 expression was elevated in macrophages from patients with inflammatory arthritis and collagen-induced arthritis (CIA) model mice. Conditional knockout of Pim2 in macrophages or administration of the Pim2 inhibitor HJ-PI01 attenuated arthritis development by inhibiting M1 macrophage polarization. Through molecular docking and dynamic simulation, bexarotene was identified as an inhibitor of Pim2 that inhibits glycolysis and downstream M1 macrophage polarization, thereby mitigating the progression of inflammatory arthritis. For targeted treatment, neutrophil membrane-coated bexarotene (Bex)-loaded PLGA-based nanoparticles (NM@NP-Bex) were developed to slow the progression of inflammatory arthritis by suppressing the polarization of M1 macrophages, and these nanoparticles (NPs) exhibited superior therapeutic effects with fewer side effects. Taken together, the results of our study demonstrated that targeting Pim2 inhibition could effectively alleviate inflammatory arthritis via glycolysis inhibition and reversal of the M1/M2 macrophage imbalance. NM@NPs loaded with bexarotene could represent a promising targeted strategy for the treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Xiaojun Xu
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
| | - Peitao Xu
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
| | - Guozhen Shen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
| | - Xiaoshuai Peng
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
| | - Zhidong Liu
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
| | - Chaoqiang Chen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
| | - Wenhui Yu
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
| | - Zepeng Su
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
| | - Jiajie Lin
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
| | - Guan Zheng
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
| | - Guiwen Ye
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
| | - Zhongyu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China
| | - Yanfeng Wu
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China.
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China.
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China.
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China.
| | - Jinteng Li
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518033, PR China.
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518033, PR China.
| |
Collapse
|
9
|
Zhu X, Zhou X, Li S, Liu Z, Yu S, Shi H, Zhu L, Song B, Si Z, Sun M, Zhu W. PFKFB3 decreases α-ketoglutarate production while partial PFKFB3 knockdown in macrophages ameliorates arthritis in tumor necrosis factor-transgenic mice. Int Immunopharmacol 2025; 148:114102. [PMID: 39870011 DOI: 10.1016/j.intimp.2025.114102] [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: 11/11/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 01/29/2025]
Abstract
OBJECTIVE Aberrant 6-phosphofructo-2kinase/fructose-2,6-bisphoshatase 3 (PFKFB3) expression is tightly correlated with multiple steps of tumorigenesis; however, the pathological significance of PFKFB3 in macrophages in patients with rheumatoid arthritis (RA) remains obscure. In this study, we examined whether PFKFB3 modulates macrophage activation and promotes RA development. METHOD Peripheral blood mononuclear cells (PBMCs) from patients with RA, THP-1 cells, and bone marrow-derived macrophages from conditional PFKFB3-knockout mice were used to investigate the mechanism underlying PFKFB3-induced macrophage regulation of RA. RESULT We demonstrated that patients with RA have higher PFKFB3 levels than healthy volunteers. PFKFB3 silencing suppressed M1 macrophage polarization and downregulated IL-1β, CD80, IFIT1, CCL8, and CXCL10 in macrophages of patients with RA. PFKFB3 overexpression markedly upregulated IRF5, HIF1α, IL-1β, CD80, IFI27, IFI44, IFIT1, IFIT3, CCL2, CCL8, CXCL10, CXCL11, and MMP13 in phorbol 12-myristate 13-acetate-induced THP-1 cells, although these changes were partially reversed by PFK15, an inhibitor of PFKFB3 enzyme activity. Co-immunoprecipitation assays revealed that PFKFB3 interacted with GLUD1 and decreased glutamate dehydrogenase (GDH) activity and α-ketoglutarate production. PFKFB3, TNFα, IL-6, IFNγ, CXCL9, CXCL10, CXCL11, MMP13, and MMP19 were downregulated in bone marrow-derived macrophages of conditional PFKFB3-knockout mice relative to those of wild-type mice. Partial PFKFB3 knockdown in macrophages ameliorated the clinical signs of arthritis and bone destruction, inhibited proinflammatory factor expression, and promoted GDH activity and α-ketoglutarate production in tumor necrosis factor-transgenic mice. Single-cell sequencing revealed that macrophages were the most abundant cells in the ankles of arthritic mice, and partial PFKFB3 knockdown promoted M2-like polarization and was correlated with TREM2, SPP1, APOE, and C1Q expression. CONCLUSION PFKFB3 is upregulated in macrophages in patients with RA. PFKFB3 aggravates arthritis by modulating macrophage activity, which may be related to decreased α-ketoglutarate production.
Collapse
Affiliation(s)
- Xiaodong Zhu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Xiaohui Zhou
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Shuaiyi Li
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Zenghui Liu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Shidi Yu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Hong Shi
- Department of Rheumatology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - LingLing Zhu
- Department of Rheumatology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Baohui Song
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Zihou Si
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Mingshuang Sun
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Wei Zhu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China.
| |
Collapse
|
10
|
Chowdhury S, Fong SS, Uetz P. The protein interactome of Escherichia coli carbohydrate metabolism. PLoS One 2025; 20:e0315240. [PMID: 39903745 PMCID: PMC11793828 DOI: 10.1371/journal.pone.0315240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 11/21/2024] [Indexed: 02/06/2025] Open
Abstract
We investigate how protein-protein interactions (PPIs) can regulate carbohydrate metabolism in Escherichia coli. We specifically investigated the stoichiometry of 378 PPIs involving carbohydrate metabolic enzymes. In 48 interactions, the interactors were much more abundant than the enzyme and are thus likely to affect enzyme activity and carbohydrate metabolism. Many of these PPIs are conserved across thousands of bacteria including pathogens and microbial species. E. coli adapts to different cellular environments by adjusting the quantities of the interacting proteins (25 PPIs) in a way that the protein-enzyme interaction (PEI) is a likely mechanism to regulate its metabolism in specific environments. We predict 3 PPIs (RpsB-AdhE, DcyD-NanE and MinE-Yccx) previously not known to regulate metabolism.
Collapse
Affiliation(s)
- Shomeek Chowdhury
- Center for Integrative Life Sciences Education, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Stephen S. Fong
- Center for Integrative Life Sciences Education, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Peter Uetz
- Center for Biological Data Science, School of Life Sciences, Virginia Commonwealth University, Richmond, VA, United States of America
| |
Collapse
|
11
|
Rojas-Pirela M, Andrade-Alviárez D, Rojas V, Marcos M, Salete-Granado D, Chacón-Arnaude M, Pérez-Nieto MÁ, Kemmerling U, Concepción JL, Michels PAM, Quiñones W. Exploring glycolytic enzymes in disease: potential biomarkers and therapeutic targets in neurodegeneration, cancer and parasitic infections. Open Biol 2025; 15:240239. [PMID: 39904372 PMCID: PMC11793985 DOI: 10.1098/rsob.240239] [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: 08/26/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 02/06/2025] Open
Abstract
Glycolysis, present in most organisms, is evolutionarily one of the oldest metabolic pathways. It has great relevance at a physiological level because it is responsible for generating ATP in the cell through the conversion of glucose into pyruvate and reducing nicotinamide adenine dinucleotide (NADH) (that may be fed into the electron chain in the mitochondria to produce additional ATP by oxidative phosphorylation), as well as for producing intermediates that can serve as substrates for other metabolic processes. Glycolysis takes place through 10 consecutive chemical reactions, each of which is catalysed by a specific enzyme. Although energy transduction by glucose metabolism is the main function of this pathway, involvement in virulence, growth, pathogen-host interactions, immunomodulation and adaptation to environmental conditions are other functions attributed to this metabolic pathway. In humans, where glycolysis occurs mainly in the cytosol, the mislocalization of some glycolytic enzymes in various other subcellular locations, as well as alterations in their expression and regulation, has been associated with the development and progression of various diseases. In this review, we describe the role of glycolytic enzymes in the pathogenesis of diseases of clinical interest. In addition, the potential role of these enzymes as targets for drug development and their potential for use as diagnostic and prognostic markers of some pathologies are also discussed.
Collapse
Affiliation(s)
- Maura Rojas-Pirela
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Servicio de Medicina Interna, Hospital Universitario de Salamanca, Salamanca37007, Spain
| | - Diego Andrade-Alviárez
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - Verónica Rojas
- Instituto de Biología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso2373223, Chile
| | - Miguel Marcos
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Servicio de Medicina Interna, Hospital Universitario de Salamanca, Salamanca37007, Spain
| | - Daniel Salete-Granado
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
| | - Marirene Chacón-Arnaude
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - María Á. Pérez-Nieto
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León, Soria42002, Spain
| | - Ulrike Kemmerling
- Instituto de Ciencias Biomédicas, Universidad de Chile, Facultad de Medicina, Santiago de Chile8380453, Chile
| | - Juan Luis Concepción
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - Paul A. M. Michels
- School of Biological Sciences, University of Edinburgh, The King’s Buildings, EdinburghEH9 3FL, UK
| | - Wilfredo Quiñones
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| |
Collapse
|
12
|
Wan X, Zhang Y, Zhang K, Mou Y, Jin X, Huang X. The alterations of ocular surface metabolism and the related immunity inflammation in dry eye. ADVANCES IN OPHTHALMOLOGY PRACTICE AND RESEARCH 2025; 5:1-12. [PMID: 39758836 PMCID: PMC11699629 DOI: 10.1016/j.aopr.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/22/2024] [Accepted: 08/09/2024] [Indexed: 01/07/2025]
Abstract
Background Dry eye disease (DED) stands as a prominent ocular condition of global prevalence, emerging as a growing concern within public health. However, the underlying mechanisms involved in its pathogenesis remain largely unknown. In recent years, with the development of metabolomics, numerous studies have reported alterations in ocular surface metabolism in DED and offered fresh perspectives on the development of DED. Main text The metabolic changes of the ocular surface of DED patients are closely intertwined with the cellular metabolism process and immune inflammation changes. This article expounds upon the correlation between ocular surface metabolism and immune inflammation alterations in DED in terms of glycolysis, lipid metabolism, amino acid metabolism, cellular signaling pathways, and immune inflammation regulation. Conclusions The alterations in ocular surface metabolism of patients with dry eye are closely associated with their inflammatory status. Our work contributes novel insights into the pathogenesis of dry eye diseases and offers innovative molecular targets for diagnosing, detecting, and managing DED patients.
Collapse
Affiliation(s)
- Xiaojie Wan
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Yu Zhang
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Kaiye Zhang
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Yujie Mou
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Xiuming Jin
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Xiaodan Huang
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| |
Collapse
|
13
|
Batori RK, Bordan Z, Padgett CA, Huo Y, Chen F, Atawia RT, Lucas R, Ushio-Fukai M, Fukai T, Belin de Chantemele EJ, Stepp DW, Fulton DJR. PFKFB3 Connects Glycolytic Metabolism with Endothelial Dysfunction in Human and Rodent Obesity. Antioxidants (Basel) 2025; 14:172. [PMID: 40002359 PMCID: PMC11851787 DOI: 10.3390/antiox14020172] [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: 01/07/2025] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity and type 2 diabetes (T2D) increase cardiovascular risk, largely due to altered metabolic state. An early consequence of T2D/obesity is the loss of endothelial function and impaired nitric oxide (NO) signaling. In blood vessels, endothelial nitric oxide synthase (eNOS) synthesizes NO to maintain vessel homeostasis. The biological actions of NO are compromised by superoxide that is generated by NADPH oxidases (NOXs). Herein we investigated how altered metabolism affects superoxide/NO balance in obesity. We found that eNOS expression and NO bioavailability are significantly decreased in endothelial cells (ECs) from T2D patients and animal models of obesity. In parallel, PFKFB3, a key glycolytic regulatory enzyme, is significantly increased in ECs of obese animals. EC overexpression of wild-type and a cytosol-restricted mutant PFKFB3 decreased NO production due to increased eNOS-T495 phosphorylation. PFKFB3 also blunted Akt-S473 phosphorylation, reducing stimulus-dependent phosphorylation of S1177 and the activation of eNOS. Furthermore, PFKFB3 enhanced the activities of NOX1 and NOX5, which are major contributors to endothelial dysfunction. Prolonged exposure of ECs to high glucose or TNFα, which are hallmarks of T2D, leads to increased PFKFB3 expression. These results demonstrate a novel functional relationship between endothelial metabolism, ROS, and NO balance that may contribute to endothelial dysfunction in obesity.
Collapse
Affiliation(s)
- Robert K. Batori
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
| | - Zsuzsanna Bordan
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
| | - Caleb A. Padgett
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
| | - Yuqing Huo
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 210029, China;
| | - Reem T. Atawia
- Department of Pharmaceutical Sciences, College of Pharmacy, Southwestern Oklahoma State University, Weatherford, OK 73096, USA;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Rudolf Lucas
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
- Department of Medicine (Cardiology), Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30912, USA
| | - Eric J. Belin de Chantemele
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
- Department of Medicine (Cardiology), Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - David W. Stepp
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - David J. R. Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
14
|
He K, Tao F, Lu Y, Fang M, Huang H, Zhou Y. The Role of HK2 in Tumorigenesis and Development: Potential for Targeted Therapy with Natural Products. Int J Med Sci 2025; 22:790-805. [PMID: 39991762 PMCID: PMC11843137 DOI: 10.7150/ijms.105553] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/11/2025] [Indexed: 02/25/2025] Open
Abstract
Hexokinase 2 (HK2) is widely distributed in various tissues, particularly showing significantly elevated expression levels in tumor tissues. As the initial rate-limiting enzyme in the glycolysis process, HK2 is believed to directly participate in the metabolic reprogramming of tumor cells. This phenomenon, known as the "Warburg effect," provides the energy and substances necessary for the rapid proliferation, growth, and division of tumor cells. Furthermore, by enhancing glycolysis, HK2 exerts its influence on various metabolic pathways in tumor cells, such as pentose phosphate metabolism, glutamine metabolism, serine metabolism, and glycine metabolism, thereby playing a role in the occurrence and development of cancer. Therefore, HK2 represents a promising target for cancer therapy. Simultaneously, natural products with effects on inhibiting the expression or activity of HK2, have already been discovered to exhibit significant anticancer potential. Flavonoids, pentacyclic triterpenoids, phenolic compounds, and lignans constitute the majority of these natural products, directly inhibiting HK2 or indirectly downregulating it through protein kinase B (AKT), hypoxia-inducible factor 1 alpha (HIF-1α), and c-Myc signaling pathways. However, several challenges remain, such as further screening for natural products that directly target and inhibit HK2, optimizing the selection of natural product inhibitors for HK2, and elucidating the molecular mechanisms by which natural products indirectly inhibit HK2. In conclusion, the potential of targeting HK2 for cancer therapy is promising, and with these challenges addressed, natural products inhibiting HK2 will play an even greater role in the fight against cancer.
Collapse
Affiliation(s)
- Keren He
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangfang Tao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yangyuxiao Lu
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengqi Fang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hong Huang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuan Zhou
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
15
|
Chen Z, Yu T, Wang Y, Li J, Zhang B, Zhou L. Mechanistic insights into the role of traditional Chinese medicine in treating gastric cancer. Front Oncol 2025; 14:1443686. [PMID: 39906672 PMCID: PMC11790455 DOI: 10.3389/fonc.2024.1443686] [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: 06/04/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025] Open
Abstract
Gastric cancer remains a leading cause of cancer-related mortality worldwide, with advanced stages presenting significant challenges due to metastasis and drug resistance. Traditional Chinese Medicine (TCM) offers a promising complementary approach characterized by holistic treatment principles and minimal side effects. This review comprehensively explores the multifaceted mechanisms by which TCM addresses gastric cancer. Specifically, we detail how TCM inhibits aerobic glycolysis by downregulating key glycolytic enzymes and metabolic pathways, thereby reducing the energy supply essential for cancer cell proliferation. We examine how TCM suppresses angiogenesis by targeting the vascular endothelial growth factor (VEGF) and cyclooxygenase-2 (COX-2) pathways, effectively starving tumors of nutrients and oxygen required for growth and metastasis. Furthermore, TCM modulates the immune microenvironment by enhancing the activity of effector immune cells such as CD4+ and CD8+ T cells and natural killer (NK) cells while reducing immunosuppressive cells like regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). These actions collectively contribute to slowing tumor progression, inhibiting metastasis, and enhancing the body's antitumor response. The insights presented underscore the significant potential of TCM as an integral component of comprehensive gastric cancer treatment strategies, highlighting avenues for future research and clinical application to improve patient outcomes.
Collapse
Affiliation(s)
- Ziqiang Chen
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Ting Yu
- Department of Rheumatism, Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yunhe Wang
- Department of Endocrinology, Metabolism and Gastroenterology, Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jiaxin Li
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Bo Zhang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Liya Zhou
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
16
|
Suh JY, Sim DY, Ahn CH, Park SY, Shim BS, Kim B, Lee DY, Jeong HB, Lee HE, Kim SH. Crucial Role of c-Myc/Monocarboxylate Transporter 4 Signaling in Capsaicin Induced Apoptotic and Anti-Warburg Effects in Hepatocellular Carcinoma. Phytother Res 2025; 39:536-547. [PMID: 39655472 DOI: 10.1002/ptr.8388] [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: 06/05/2024] [Revised: 10/10/2024] [Accepted: 10/29/2024] [Indexed: 01/30/2025]
Abstract
Though Capsaicin from chili peppers was known to have antitumor effects in several cancers, the underlying antitumor pathogenesis of Capsaicin is not clear to date. Thus, the antitumor mechanism of Capsaicin was explored in Hep3B and Huh7 hepatocellular carcinoma (HCC) cells in relation to c-Myc/monocarboxylate transporter 4 (MCT4) signaling. To elucidate the antitumor mechanism of capsaicin, cytotoxicity assay, cell cycle analysis, Western blotting, RT-qPCR, RNA interference, ELISA, immunoprecipitation, and mouse xenograft model were used in this work. Capsaicin increased the cytotoxicity, subG1 population, and the number of TUNEL-positive bodies in Huh7 and Hep3B cells. Consistently, Capsaicin diminished the expression of pro-PARP, HK2, PKM2, LDHA, glucose transporter type 1 (Glut1), c-Myc, and monocarboxylate transporter 4 (MCT4) in Huh7 and Hep3B cells, along with decreased production of glucose, lactate, and ATP. However, a glycolysis end product pyruvate treatment reversed the capacity of Capsaicin to attenuate the expression of pro-PARP, HK2, c-Myc, and MCT4 in Hep3B cells. Furthermore, Capsaicin reduced c-Myc stability in the presence of cycloheximide and induced c-Myc ubiquitination in Hep3B cells, while c-Myc directly binds to MCT4 as a lactate transporter and downstream of c-Myc in Hep3B cells by immunoprecipitation and correlation factor (Spearman efficient = 0.0027). Furthermore, a preliminary analysis of an animal study reveals that Capsaicin significantly suppressed the growth of Hep3B cells inoculated in BALB/c nude mice without hurting body weight, liver, and spleen. Our findings provide novel evidence that Capsaicin exerts apoptotic and anti-Warburg effect via c-Myc/MCT4 signaling axis as a potent anticancer candidate for liver cancer therapy.
Collapse
Affiliation(s)
- Jin Young Suh
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Deok Yong Sim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chi-Hoon Ahn
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Su-Yeon Park
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bum-Sang Shim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bonglee Kim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Dae Young Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Hyo Bong Jeong
- Department of Horticultural Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Hye Eun Lee
- Department of Horticultural Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Sung-Hoon Kim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
17
|
Bibi M, Baboo I, Majeed H, Kumar S, Lackner M. Molecular Docking of Key Compounds from Acacia Honey and Nigella sativa Oil and Experimental Validation for Colitis Treatment in Albino Mice. BIOLOGY 2024; 13:1035. [PMID: 39765702 PMCID: PMC11673436 DOI: 10.3390/biology13121035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025]
Abstract
Colitis, an inflammatory condition of the colon that encompasses ulcerative colitis (UC) and Crohn's disease, presents significant challenges due to the limitations and side effects of current treatments. This study investigates the potential of natural products, specifically AH and NSO, as organic therapeutic agents for colitis. Molecular docking studies were conducted to identify the binding affinities and interaction mechanisms between the bioactive compounds in AH and NSO and proteins implicated in colitis, such as those involved in inflammation and oxidative stress pathways. An in vivo experiment was performed using an albino mouse model of colitis, with clinical symptoms, histopathological assessments, and biochemical analyses conducted to evaluate the therapeutic effects of the compounds both individually and in combination. Results from the molecular docking studies revealed promising binding interactions between fructose and Prostaglandin G/H synthase 2 (Ptgs2) and between fructose and cellular tumor antigen p53, with docking energy measured at -6.0 kcal/mol and -5.1 kcal/mol, respectively. Meanwhile, the presence of glucose molecule glucokinase chain A (-6.3 kcal/mol) and chain B (-5.8 kcal/mol) indicated potential efficacy in modulating inflammatory pathways. Experimental data demonstrated that treatment with AH and NSO significantly reduced inflammation, improved gut health, and ameliorated colitis symptoms. Histopathological evaluations confirmed reduced mucosal damage and immune cell infiltration, while biochemical analyses showed normalization of inflammatory markers and oxidative stress levels. This study provides compelling evidence for the potential of AH and NSO as natural, complementary treatments for colitis, suggesting their future role in integrative therapeutic strategies. However, further research into long-term safety, optimal dosing, and mechanisms of action is warranted to translate these findings into clinical applications.
Collapse
Affiliation(s)
- Mehwish Bibi
- Department of Zoology, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur 63100, Pakistan; (M.B.); (S.K.)
| | - Irfan Baboo
- Department of Zoology, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur 63100, Pakistan; (M.B.); (S.K.)
| | - Hamid Majeed
- Department of Food Science and Technology, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur 63100, Pakistan;
| | - Santosh Kumar
- Department of Zoology, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur 63100, Pakistan; (M.B.); (S.K.)
| | - Maximilian Lackner
- Department of Industrial Engineering, University of Applied Sciences Technikum Wien, 17 Hoechstaedtplatz 6, 1200 Vienna, Austria
| |
Collapse
|
18
|
Li Z, Li N, Ndzie Noah ML, Shao Q, Zhan X. Pharmacoproteomics reveals energy metabolism pathways as therapeutic targets of ivermectin in ovarian cancer toward 3P medical approaches. EPMA J 2024; 15:711-737. [PMID: 39635022 PMCID: PMC11612093 DOI: 10.1007/s13167-024-00385-1] [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: 10/21/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Objective Ovarian cancer is the malignant tumor with the highest mortality rate in the female reproductive system, enormous socio-economic burden, and limited effective drug therapy. There is an urgent need to find novel effective drugs for ovarian cancer therapy. Our previous in vitro studies demonstrate that ivermectin effectively inhibits ovarian cancer cells and affects energy metabolism pathways. This study aims to clarify in vivo mechanisms and therapeutic targets of ivermectin in the treatment of ovarian cancer to establish predictive biomarkers, guide personalized treatments, and improve preventive strategies in the framework of 3P medicine. Methods A TOV-21G tumor-bearing mouse model was constructed based on histopathological data and biochemical parameters. TMT-based proteomic analysis was performed on tumor tissues from the different treatment groups. All significantly differentially abundant proteins were characterized by hierarchical clustering, Gene Ontology (GO) enrichment analyses, and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. In addition, the data were integrated and analyzed with the proteomic data of clinical ovarian cancer tissues from our previous study and the proteomic data of ivermectin intervention in ovarian cancer cells to identify key regulators of ivermectin. Results Ivermectin (10 mg/kg) had a significant anti-ovarian cancer effect in mice, with a tumor inhibitory rate of 61.5%. Molecular changes in tumor tissue of ivermectin-treated mice were established, and protein-protein interaction (PPI) analysis showed that the main differential pathway networks included the TCA cycle, propanoate metabolism, 2-0xocarboxyacid metabolism, and other pathways. Integrating our previous clinical ovarian cancer tissue and cell experimental data, this study found that ivermectin significantly interfered with the energy metabolic pathways of ovarian cancer, including glycolysis, TCA cycle, oxidative phosphorylation, and other related pathways. Conclusions This study evaluated the anti-ovarian cancer effect in vitro and in vivo, and its specific regulatory effect on energy metabolism. The expressions of drug target molecules in the energy metabolism pathway of ovarian cancer will be used to guide the diagnosis and prevention of ovarian cancer. The significant efficacy of ivermectin will be applied to the treatment of ovarian cancer and personalized medication. This has guiding significance for the clinical diagnosis, treatment, personalized medication, and prognosis evaluation of ovarian cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00385-1.
Collapse
Affiliation(s)
- Zhijun Li
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Na Li
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Marie Louise Ndzie Noah
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Qianwen Shao
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingao Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
19
|
Ge L, Feng T, Cao S, Yin H, Zhang Y, Pan J, Chen W, Cheng K, Wang L. Enhancing Student Comprehension of Glucose Metabolism Visualization Through Virtual Simulation Platform: An Educational Approach. Pharmacol Res Perspect 2024; 12:e70042. [PMID: 39601766 PMCID: PMC11600983 DOI: 10.1002/prp2.70042] [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: 07/09/2024] [Revised: 10/22/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
With the rapid progression of biotechnology, the significance translational research on glycolysis in molecular pharmacology has become increasingly evident. To deepen students' understanding of glycolytic processes and facilitate their comprehension of drug action mechanisms, we have developed a visual virtual simulation platform dedicated glycolysis. The educational approach commenced with theoretical lectures on glycolysis, followed by practical laboratory sessions where students measured glycolysis-related parameters such as hexokinase, pyruvate kinase, and lactate. Students then engaged with the virtual simulation training platform to explore glycolytic stress tests and positron emission tomography/computed tomography (PET/CT) imaging, with their progress tracked through an assessment mode. The study involved 67 s-year undergraduate students majoring in biomedical sciences, all of whom had received instruction in glucose metabolism theories and completed the associated questionnaires. The results showed that the students gained a deeper understanding of glycolysis and the clinical application of PET/CT imaging in the context of glycolysis. The majority also agreed that the integration of scientific and clinical cases in teaching is beneficial and that the project sparked their interest in scientific research. These findings align with existing literature that emphasizes the importance of innovative educational tools in enhancing student engagement and understanding of the underlying theories of the curriculum. This project designed an innovative glycolytic metabolism teaching system encompassing the monitoring of traditional glycolytic indicators, glycolytic stress tests, and PET/CT imaging based on glycolysis. The visual virtual simulation platform for glycolysis can serve as an innovative educational tool in the molecular pharmacology curriculum or other courses involving glycolysis, assisting students in deeply understanding the molecular mechanisms of glycolysis and its significance in disease and drug action.
Collapse
Affiliation(s)
- Luna Ge
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| | - Tingting Feng
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| | - Shuo Cao
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| | - Hang Yin
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| | - Yuang Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| | - Jihong Pan
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| | - Wei Chen
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| | - Kai Cheng
- Department of PET/CT Center, Shandong Cancer Hospital and InstituteShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
| | - Lin Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| |
Collapse
|
20
|
Ao Y, Guo Y, Zhang Y, Xie L, Xia R, Xu J, Shi M, Gao X, Yu X, Chen Z. Hypoxia-Mimicking Mediated Macrophage-Elimination of Erythrocytes Promotes Bone Regeneration via Regulating Integrin α vβ 3/Fe 2+-Glycolysis-Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403921. [PMID: 39352318 PMCID: PMC11615788 DOI: 10.1002/advs.202403921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/20/2024] [Indexed: 12/06/2024]
Abstract
Erythrocytes are the dominant component of a blood clot in terms of volume and number. However, longstanding compacted erythrocytes in blood clots form a physical barrier and make fibrin mesh more anti-fibrinolytic, thus impeding infiltration of mesenchymal stem cells. The necrosis or lysis of erythrocytes that are not removed timely can also lead to the release of pro-inflammatory toxic metabolites, interfering with bone regeneration. Proper bio-elimination of erythrocytes is essential for an undisturbed bone regeneration process. Here, hypoxia-mimicking is applied to enhance macrophage-elimination of erythrocytes. The effect of macrophage-elimination of erythrocytes on the macrophage intracellular reaction, bone regenerative microenvironment, and bone regeneration outcome is investigated. Results show that the hypoxia-mimicking agent dimethyloxalylglycine successfully enhances erythrophagocytosis by macrophages in a dose-dependent manner primarily by up-regulating the expression of integrin αvβ3. Increased phagocytosed erythrocytes then regulate macrophage intracellular Fe2+-glycolysis-inflammation, creating an improved bone regenerative microenvironment characterized by loose fibrin meshes with down-regulated local inflammatory responses in vivo, thus effectively promoting early osteogenesis and ultimate bone generation. Modulating macrophage-elimination of erythrocytes can be a promising strategy for eradicating erythrocyte-caused bone regeneration hindrance and offers a new direction for advanced biomaterial development focusing on the bio-elimination of erythrocytes.
Collapse
Affiliation(s)
- Yong Ao
- Hospital of StomatologyGuanghua School of StomatologySun Yat‐sen UniversityGuangdong Research Center for Dental and Cranial Rehabilitation and Material EngineeringGuangzhou510055China
| | - Yuanlong Guo
- Hospital of StomatologyGuanghua School of StomatologySun Yat‐sen UniversityGuangdong Research Center for Dental and Cranial Rehabilitation and Material EngineeringGuangzhou510055China
| | - Yingye Zhang
- Hospital of StomatologyGuanghua School of StomatologySun Yat‐sen UniversityGuangdong Research Center for Dental and Cranial Rehabilitation and Material EngineeringGuangzhou510055China
| | - Lv Xie
- Hospital of StomatologyGuanghua School of StomatologySun Yat‐sen UniversityGuangdong Research Center for Dental and Cranial Rehabilitation and Material EngineeringGuangzhou510055China
| | - Ruidi Xia
- Hospital of StomatologyGuanghua School of StomatologySun Yat‐sen UniversityGuangdong Research Center for Dental and Cranial Rehabilitation and Material EngineeringGuangzhou510055China
| | - Jieyun Xu
- Hospital of StomatologyGuanghua School of StomatologySun Yat‐sen UniversityGuangdong Research Center for Dental and Cranial Rehabilitation and Material EngineeringGuangzhou510055China
| | - Mengru Shi
- Hospital of StomatologyGuanghua School of StomatologySun Yat‐sen UniversityGuangdong Research Center for Dental and Cranial Rehabilitation and Material EngineeringGuangzhou510055China
| | - Xiaomeng Gao
- Hospital of StomatologyGuanghua School of StomatologySun Yat‐sen UniversityGuangdong Research Center for Dental and Cranial Rehabilitation and Material EngineeringGuangzhou510055China
| | - Xiaoran Yu
- Hospital of StomatologyGuanghua School of StomatologySun Yat‐sen UniversityGuangdong Research Center for Dental and Cranial Rehabilitation and Material EngineeringGuangzhou510055China
| | - Zetao Chen
- Hospital of StomatologyGuanghua School of StomatologySun Yat‐sen UniversityGuangdong Research Center for Dental and Cranial Rehabilitation and Material EngineeringGuangzhou510055China
| |
Collapse
|
21
|
Liu Q, Li J, Li X, Zhang L, Yao S, Wang Y, Tuo B, Jin H. Advances in the understanding of the role and mechanism of action of PFKFB3‑mediated glycolysis in liver fibrosis (Review). Int J Mol Med 2024; 54:105. [PMID: 39301662 PMCID: PMC11448561 DOI: 10.3892/ijmm.2024.5429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
Liver fibrosis is a pathophysiologic manifestation of chronic liver disease and a precursor to cirrhosis and hepatocellular carcinoma. Glycolysis provides intermediate metabolites as well as energy support for cell proliferation and phenotypic transformation in liver fibers. 6‑Phosphofructo‑2‑kinase/fructose‑2,6‑bisphosphatase 3 (PFKFB3) is a key activator of glycolysis and plays an important role in the process of glycolysis. The role of PFKFB3‑mediated glycolysis in myocardial fibrosis, renal fibrosis and pulmonary fibrosis has been demonstrated, and the role of PFKFB3 in the activation of hepatic stellate cells by aerobic glycolysis has been proven by relevant experiments. The present study reviews the research progress on the role and mechanism of action of PFKFB3‑mediated glycolysis in the progression of hepatic fibrosis to discuss the role of PFKFB3‑mediated glycolysis in hepatic fibrosis and to provide new ideas for research on PFKFB3 as a target for the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Qian Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jiajia Li
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine 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
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine 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
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine 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
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine 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
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
22
|
Henriques J, Berenbaum F, Mobasheri A. Obesity-induced fibrosis in osteoarthritis: Pathogenesis, consequences and novel therapeutic opportunities. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100511. [PMID: 39483440 PMCID: PMC11525450 DOI: 10.1016/j.ocarto.2024.100511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/26/2024] [Accepted: 08/12/2024] [Indexed: 11/03/2024] Open
Abstract
Osteoarthritis (OA) is a significant global burden, affecting more than half a billion people across the world. It is characterized by degeneration and loss of articular cartilage, synovial inflammation, and subchondral bone sclerosis, leading to pain and functional impairment. After age, obesity is a major modifiable risk factor for OA, and it has recently been identified as a chronic disease by the World Health Organization (WHO). Obesity is associated with high morbidity and mortality, imposing a significant cost on individuals and society. Obesity increases the risk of knee OA through increased joint loading, altered body composition, and elevated pro-inflammatory adipokines in the systemic circulation. Moreover, obesity triggers fibrotic processes in different organs and tissues, including those involved in OA. Fibrosis in OA refers to the abnormal accumulation of fibrous tissue within and around the joints. It can be driven by increased adiposity, low-grade inflammation, oxidative stress, and metabolic alterations. However, the clinical outcomes of fibrosis in OA are unclear. This review focuses on the link between obesity and OA, explores the mechanism of obesity-driven fibrosis, and examines potential therapeutic opportunities for targeting fibrotic processes in OA.
Collapse
Affiliation(s)
- João Henriques
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Francis Berenbaum
- Sorbonne University, Paris, France
- Department of Rheumatology, Saint-Antoine Hospital, Assistance Publique-Hopitaux de Paris, Paris, France
- INSERM CRSA, Paris, France
| | - Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium
| |
Collapse
|
23
|
Fan Y, Sun J, Fan W, Zhong X, Yin Z, Su B, Yao J, Hong X, Zhai J, Wang Z, Chen H, Guo F, Wen X, Ning C, Chen L, Yu P. Three-Dimensional Semiconductor Network as Regulators of Energy Metabolism Drives Angiogenesis in Bone Regeneration. ACS NANO 2024; 18:32602-32616. [PMID: 39530623 DOI: 10.1021/acsnano.4c09971] [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: 11/16/2024]
Abstract
Insufficient vascularization is a primary cause of bone implantation failure. The management of energy metabolism is crucial for the achievement of vascularized osseointegration. In light of the bone semiconductor property and the electric property of semiconductor heterojunctions, a three-dimensional semiconductor heterojunction network (3D-NTBH) implant has been devised with the objective of regulating cellular energy metabolism, thereby driving angiogenesis for bone regeneration. The three-dimensional heterojunction interfaces facilitate electron transfer and establish internal electric fields at the nanoscale interfaces. The 3D-NTBH was found to noticeably accelerate glycolysis in endothelial cells, thereby rapidly providing energy to support cellular metabolic activities and ultimately driving angiogenesis within the bone tissue. Molecular dynamic simulations have demonstrated that the 3D-NTBH facilitates the exposure of fibronectin's Arg-Gly-Asp peptide binding site, thereby regulating the glycolysis of endothelial cells. Further evidence suggests that 3D-NTBH promotes early vascular network reconstruction and bone regeneration in vivo. The findings of this research offer a promising research perspective for the design of vascularizing implants.
Collapse
Affiliation(s)
- Youzhun Fan
- School of Materials Science and Engineering, Guangdong Engineering Technology Research Center of Metallic Materials Surface Functionalization, National Engineering Research Center for Tissue Restoration and Reconstruction, Medical Devices Research and Testing Center, South China University of Technology, Guangzhou 510641, China
| | - Jiwei Sun
- Department of Stomatology, School of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Wenjie Fan
- Department of Stomatology, School of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xianwei Zhong
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Lab of Green Chemical Product Technology, South China University of Technology, Guangzhou 510641, China
| | - Zhaoyi Yin
- Faculty of Materials Science and Engineering, Kunming University of Science and Technology, Kunming 650093, China
| | - Bin Su
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Yao
- Department of Stomatology, School of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xinyu Hong
- Department of Stomatology, School of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Jinxia Zhai
- School of Materials Science and Engineering, Guangdong Engineering Technology Research Center of Metallic Materials Surface Functionalization, National Engineering Research Center for Tissue Restoration and Reconstruction, Medical Devices Research and Testing Center, South China University of Technology, Guangzhou 510641, China
| | - Zhengao Wang
- School of Materials Science and Engineering, Guangdong Engineering Technology Research Center of Metallic Materials Surface Functionalization, National Engineering Research Center for Tissue Restoration and Reconstruction, Medical Devices Research and Testing Center, South China University of Technology, Guangzhou 510641, China
| | - Haoyan Chen
- School of Materials Science and Engineering, Guangdong Engineering Technology Research Center of Metallic Materials Surface Functionalization, National Engineering Research Center for Tissue Restoration and Reconstruction, Medical Devices Research and Testing Center, South China University of Technology, Guangzhou 510641, China
| | - Fengyuan Guo
- Department of Stomatology, School of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xiufang Wen
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Lab of Green Chemical Product Technology, South China University of Technology, Guangzhou 510641, China
| | - Chengyun Ning
- School of Materials Science and Engineering, Guangdong Engineering Technology Research Center of Metallic Materials Surface Functionalization, National Engineering Research Center for Tissue Restoration and Reconstruction, Medical Devices Research and Testing Center, South China University of Technology, Guangzhou 510641, China
| | - Lili Chen
- Department of Stomatology, School of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Peng Yu
- School of Materials Science and Engineering, Guangdong Engineering Technology Research Center of Metallic Materials Surface Functionalization, National Engineering Research Center for Tissue Restoration and Reconstruction, Medical Devices Research and Testing Center, South China University of Technology, Guangzhou 510641, China
| |
Collapse
|
24
|
Kirdaite G, Denkovskij J, Mieliauskaite D, Pachaleva J, Bernotiene E. The Challenges of Local Intra-Articular Therapy. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1819. [PMID: 39597004 PMCID: PMC11596802 DOI: 10.3390/medicina60111819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Fibroblast-like synoviocytes (FLSs) are among the main disease-driving players in most cases of monoarthritis (MonoA), oligoarthritis, and polyarthritis. In this review, we look at the characteristics and therapeutic challenges at the onset of arthritis and during follow-up management. In some cases, these forms of arthritis develop into autoimmune polyarthritis, such as rheumatoid arthritis (RA), whereas local eradication of the RA synovium could still be combined with systemic treatment using immunosuppressive agents. Currently, the outcomes of local synovectomies are well studied; however, there is still a lack of a comprehensive analysis of current local intra-articular treatments highlighting their advantages and disadvantages. Therefore, the aim of this study is to review local intra-articular therapy strategies. According to publications from the last decade on clinical studies focused on intra-articular treatment with anti-inflammatory molecules, a range of novel slow-acting forms of steroidal drugs for the local treatment of synovitis have been investigated. As pain is an essential symptom, caused by both inflammation and cartilage damage, various molecules acting on pain receptors are being investigated in clinical trials as potential targets for local intra-articular treatment. We also overview the new targets for local treatment, including surface markers and intracellular proteins, non-coding ribonucleic acids (RNAs), etc.
Collapse
Affiliation(s)
- Gailute Kirdaite
- Department of Personalised Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Jaroslav Denkovskij
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania (E.B.)
| | - Diana Mieliauskaite
- Department of Personalised Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Jolita Pachaleva
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania (E.B.)
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania (E.B.)
- Faculty of Fundamental Sciences, Vilnius Gediminas Technical University, VilniusTech, Sauletekio al. 11, LT-10223 Vilnius, Lithuania
| |
Collapse
|
25
|
Lin TY, Gu SY, Lin YH, Shih JH, Lin JH, Chou TY, Lee YC, Chang SF, Lang YD. Paclitaxel-resistance facilitates glycolytic metabolism via Hexokinase-2-regulated ABC and SLC transporter genes in ovarian clear cell carcinoma. Biomed Pharmacother 2024; 180:117452. [PMID: 39341074 DOI: 10.1016/j.biopha.2024.117452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Ovarian clear cell carcinoma (OCCC) frequently develops resistance to platinum-based therapies, which is regarded as an aggressive subtype. However, metabolic changes in paclitaxel resistance remain unclear. Herein, we present the metabolic alternations of paclitaxel resistance in bioenergetic profiling in OCCC. Paclitaxel-resistant OCCC cells were developed and metabolically active with oxygen consumption rates (OCR) compared to parental cells. Metabolite profiling analysis revealed that paclitaxel-resistant OCCC cells reduced intracellular ATP and GTP influx rates, increasing the NADH/NAD+ ratio. We further demonstrated that paclitaxel-resistant OCCC cells led to characteristic alternations of metabolite levels in energy-requiring and energy-releasing steps of glycolysis and their corresponding glycolytic enzymes. Copy number alterations and RNA sequencing analysis demonstrated that ATP-binding cassette (ABC) transporters and solute carrier (SLC) transporter genes involved in glycolysis metabolism and molecular transport were enriched in paclitaxel-resistant OCCC cells. We first identified that Hexokinase 2 (HK2) expression is upregulated in paclitaxel-resistant OCCC cells to determine the quantity of glucose entering glycolysis. Utilizing proteolysis-targeting chimera (PROTAC) HK2 degraders, we also found that paclitaxel sensitivity, viability, and oxygen consumption rates under paclitaxel treatment were restored by HK2 degraders treatment, and decreased downstream expression of the ABC and SLC transporters was shown in OCCC cells. Taken together, these findings highlight the paclitaxel resistance in OCCC elucidates metabolic alternation, including ABC- and SLC- drug transporters, thereby affecting glycolysis metabolism in response to paclitaxel resistance, and HK2 may become a novel potential therapeutic target for paclitaxel resistance.
Collapse
Affiliation(s)
- Tsai-Yu Lin
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Shin-Yuan Gu
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan; Department of Pathology and Precision Medicine Research Center, Taipei Medical University Hospital and Precision Health Center, Taipei Medical University, Taipei, Taiwan
| | - Yi-Hui Lin
- Department of Obstetrics and Gynecology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Jou-Ho Shih
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jiun-Han Lin
- Department of Industrial Technology, Ministry of Economic Affairs, Taipei, Taiwan; Food Industry Research and Development Institute, Hsinchu City, Taiwan
| | - Teh-Ying Chou
- Department of Pathology and Precision Medicine Research Center, Taipei Medical University Hospital and Precision Health Center, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ching Lee
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| | - Shwu-Fen Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yaw-Dong Lang
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan; Department of Pathology and Precision Medicine Research Center, Taipei Medical University Hospital and Precision Health Center, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
26
|
Zhou Y, Peng X, Fang C, Peng X, Tang J, Wang Z, Long Y, Chen J, Peng Y, Zhang Z, Zhou Y, Tang J, Liao J, Xiao D, Tao Y, Shi Y, Liu S. Histones Methyltransferase NSD3 Inhibits Lung Adenocarcinoma Glycolysis Through Interacting with PPP1CB to Decrease STAT3 Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400381. [PMID: 39119928 PMCID: PMC11481231 DOI: 10.1002/advs.202400381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/28/2024] [Indexed: 08/10/2024]
Abstract
Histones methyltransferase NSD3 targeting H3K36 is frequently disordered and mutant in various cancers, while the function of NSD3 during cancer initiation and progression remains unclear. In this study, it is proved that downregulated level of NSD3 is linked to clinical features and poor survival in lung adenocarcinoma. In vivo, NSD3 inhibited the proliferation, immigration, and invasion ability of lung adenocarcinoma. Meanwhile, NSD3 suppressed glycolysis by inhibiting HK2 translation, transcription, glucose uptake, and lactate production in lung adenocarcinoma. Mechanistically, as an intermediary, NSD3 binds to PPP1CB and p-STAT3 in protein levels, thus forming a trimer to dephosphorylate the level of p-STAT3 by PPP1CB, leading to the suppression of HK2 transcription. Interestingly, the phosphorylation function of PPP1CB is related to the concentration of carbon dioxide and pH value in the culture environment. Together, this study revealed the critical non-epigenetic role of NSD3 in the regulation of STAT3-dependent glycolysis, providing a piece of compelling evidence for targeting the NSD3/PPP1CB/p-STAT3 in lung adenocarcinoma.
Collapse
Affiliation(s)
- Yanling Zhou
- Department of OncologyInstitute of Medical SciencesNational Clinical Research Center for Geriatric DisordersInstitue of Medical SciencesXiangya Hospital, Central South UniversityChangshaHunan410008China
- Department of HematologyXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Xintong Peng
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunan410028China
| | - Cheng Fang
- Department of Cardiac SurgeryXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Xin Peng
- Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Jianing Tang
- Department of Liver SurgeryXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Zuli Wang
- Center for Tissue Engineering and Stem Cell ResearchGuizhou Medical UniversityGuiyangGuizhou561113China
| | - Yao Long
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunan410028China
| | - Jielin Chen
- Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Yuanhao Peng
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunan410028China
| | - Zewen Zhang
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunan410028China
| | - Yanmin Zhou
- Department of OncologyInstitute of Medical SciencesNational Clinical Research Center for Geriatric DisordersInstitue of Medical SciencesXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Jun Tang
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunan410028China
| | - Jingzhong Liao
- Department of Laboratory MedicineXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Desheng Xiao
- Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunan410028China
| | - Ying Shi
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunan410028China
| | - Shuang Liu
- Department of OncologyInstitute of Medical SciencesNational Clinical Research Center for Geriatric DisordersInstitue of Medical SciencesXiangya Hospital, Central South UniversityChangshaHunan410008China
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
| |
Collapse
|
27
|
Zhong W, Wang C, Sun Y. LncRNA PCIF1 promotes aerobic glycolysis in A549/DDP cells by competitively binding miR-326 to regulate PKM expression. Mol Cell Probes 2024; 77:101977. [PMID: 39074568 DOI: 10.1016/j.mcp.2024.101977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 07/20/2024] [Accepted: 07/27/2024] [Indexed: 07/31/2024]
Abstract
OBJECTIVE Utilizing transcriptome analysis to investigate the mechanisms and therapeutic approaches for cisplatin resistance in non-small cell lung cancer (NSCLC). METHODS Firstly, the biological characters of A549 cells and A549/DDP cells were detected by RNA sequencing, CCK-8 and hippocampal energy analyzer. Then, the differential Genes were functionally enriched by GO and KEGG and the competitive endogenous RNA network map was constructed. Finally, the effects of the predicted biogenesis pathway on the biological functions of A549/DDP cells were verified by in vitro and in vivo experiments. RESULT The differentially transcribed genes of A549 and A549/DDP cells were analyzed by enrichment analysis and cell biological characteristics detection. The results showed that A549/DDP cells showed significantly increased resistance to cisplatin, glucose metabolism signaling pathway and glycolysis levels compared with A549 cells. Among glycolysis-related transcription genes, PKM had the most significant difference Fold Change is 8. LncRNA PCIF1 is a new marker of A549/DDP cells and can be used as a molecular sponge to regulate the expression of PKM. LncRNA PCIF1 targets miR-326 to induce PKM expression, promote glycolysis level, and enhance the resistance of A549/DDP cells to cisplatin. CONCLUSION LncRNA PCIF1 as biomarkers of A549/DDP cells, higher expression can induce the PKM, promote cell glycolysis, lead to the occurrence of cisplatin resistance. LncRNA PCIF1 can be considered as a potential target for treating cisplatin-resistant NSCLC.
Collapse
Affiliation(s)
- Wan Zhong
- Department of Obstetrics and Gynecology, General Hospital of the Northern Theater Command, 83 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Chun Wang
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, 79 Chongshan Eastern Road, Huang gu District, Shenyang, 110847, China.
| | - Ye Sun
- Department of Pathogenic Biology, Shenyang Medical College, 146 Huanghe North Street, Yuhong District, Shenyang, 110034, China.
| |
Collapse
|
28
|
Hu Y, Tang J, Xu Q, Fang Z, Li R, Yang M, Zhao J, Chen X. Role of pyruvate kinase M2 in regulating sepsis (Review). Mol Med Rep 2024; 30:185. [PMID: 39155878 DOI: 10.3892/mmr.2024.13309] [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: 06/11/2024] [Accepted: 07/29/2024] [Indexed: 08/20/2024] Open
Abstract
Glycolysis occurs in all living organisms as a form of energy supply. Pyruvate kinase M2 (PKM2) is one of the rate‑limiting enzymes in the glycolytic process. PKM2 is considered to serve an important role in several terminal diseases, including sepsis. However, to the best of our knowledge, the specific mechanistic role of PKM2 in sepsis remains to be systematically summarised. Therefore, the present review aims to summarise the roles of PKM2 in sepsis progression. In addition, potential treatment strategies for patients with sepsis are discussed. The present review hopes to lay the groundwork for studying the role of PKM2 and developing therapeutic strategies against metabolic disorders that occur during sepsis.
Collapse
Affiliation(s)
- Yifei Hu
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Jing Tang
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Qiao Xu
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Zenghui Fang
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Rongqing Li
- Department of Clinical Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu 225000, P.R. China
| | - Mengxuan Yang
- Department of Clinical Laboratory, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Jie Zhao
- Department of Clinical Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu 225000, P.R. China
| | - Xin Chen
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| |
Collapse
|
29
|
Rong J, Wang Q, Li T, Qian J, Cheng J. Glucose metabolism in glioma: an emerging sight with ncRNAs. Cancer Cell Int 2024; 24:316. [PMID: 39272133 PMCID: PMC11395608 DOI: 10.1186/s12935-024-03499-8] [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: 03/05/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Glioma is a primary brain tumor that grows quickly, has an unfavorable prognosis, and can spread intracerebrally. Glioma cells rely on glucose as the major energy source, and glycolysis plays a critical role in tumorigenesis and progression. Substrate utilization shifts throughout glioma progression to facilitate energy generation and biomass accumulation. This metabolic reprogramming promotes glioma cell proliferation and metastasis and ultimately decreases the efficacy of conventional treatments. Non-coding RNAs (ncRNAs) are involved in several glucose metabolism pathways during tumor initiation and progression. These RNAs influence cell viability and glucose metabolism by modulating the expression of key genes of the glycolytic pathway. They can directly or indirectly affect glycolysis in glioma cells by influencing the transcription and post-transcriptional regulation of oncogenes and suppressor genes. In this review, we discussed the role of ncRNAs in the metabolic reprogramming of glioma cells and tumor microenvironments and their abnormal expression in the glucometabolic pathway in glioma. In addition, we consolidated the existing theoretical knowledge to facilitate the use of this emerging class of biomarkers as biological indicators and potential therapeutic targets for glioma.
Collapse
Affiliation(s)
- Jun Rong
- Department of Neurosurgery, Xuancheng People's Hospital, The Affiliated Xuancheng Hospital of Wannan Medical College, Xuancheng, People's Republic of China
| | - Qifu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), WuHu, People's Republic of China
| | - Tingzheng Li
- Department of Neurosurgery, Xuancheng Central Hospital, Xuancheng, People's Republic of China
| | - Jin Qian
- Department of Neurosurgery, Xuancheng People's Hospital, The Affiliated Xuancheng Hospital of Wannan Medical College, Xuancheng, People's Republic of China.
| | - Jinchao Cheng
- Department of Neurosurgery, Xuancheng Central Hospital, Xuancheng, People's Republic of China.
| |
Collapse
|
30
|
Gao J, He L, Zhang J, Xi L, Feng H. Development of a diagnostic model based on glycolysis-related genes and immune infiltration in intervertebral disc degeneration. Heliyon 2024; 10:e36158. [PMID: 39247348 PMCID: PMC11379615 DOI: 10.1016/j.heliyon.2024.e36158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 08/03/2024] [Accepted: 08/11/2024] [Indexed: 09/10/2024] Open
Abstract
Background The glycolytic pathway and immune response play pivotal roles in the intervertebral disc degeneration (IDD) progression. This study aimed to develop a glycolysis-related diagnostic model and analyze its relationship with the immune response to IDD. Methods GSE70362, GSE23130, and GSE15227 datasets were collected and merged from the Gene Expression Omnibus, and differential expression analysis was performed. Glycolysis-related differentially expressed genes (GLRDEGs) were identified, and a machine learning-based diagnostic model was constructed and validated, followed by Gene Set Enrichment Analysis (GSEA). Gene Ontology functional enrichment and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed, and mRNA-miRNA and mRNA-transcription factor (TF) interaction networks were constructed. Immune infiltration was analyzed using single-sample GSEA (ssGSEA) and cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) algorithm between high- and low-risk groups. Results In the combined dataset, samples from 31 patients with IDD and 55 normal controls were analyzed, revealing differential expression of 16 GLRDEGs between the two groups. Using advanced machine learning techniques (LASSO, support vector machine, and random forest algorithms), we identified eight common GLRDEGs (PXK, EIF3D, WSB1, ZNF185, IGFBP3, CKAP4, RPL15, and, SSR1) and developed a diagnostic model, which demonstrated high accuracy in distinguishing IDD from control samples (area under the curve, 0.935). We identified 42 mRNA-miRNA and 33 mRNA-TF interaction pairs. Using the RiskScore from the diagnostic model, the combined dataset was stratified into high- and low-risk groups. SsGSEA revealed significant differences in the infiltration abundances of the four immune cell types between the groups. The CIBERSORT algorithm revealed the strongest correlation between resting natural killer (NK) cells and ZNF185 in the low-risk group and between CD8+ T cells and SSR1 in the high-risk group. Conclusions Our study reveals a potential interplay between glycolysis-associated genes and immune infiltration in IDD pathogenesis. These findings contribute to our understanding of IDD and may guide development of novel diagnostic markers and therapeutic interventions.
Collapse
Affiliation(s)
- Jian Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, 030032, Taiyuan, China
| | - Liming He
- Department of Orthopedics, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, 030032, Taiyuan, China
| | - Jianguo Zhang
- Department of Orthopedics, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, 030032, Taiyuan, China
| | - Leimin Xi
- Department of Orthopedics, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, 030032, Taiyuan, China
| | - Haoyu Feng
- Department of Orthopedics, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, 030032, Taiyuan, China
| |
Collapse
|
31
|
Wang RH, Chen PR, Chen YT, Chen YC, Chu YH, Chien CC, Chien PC, Lo SY, Wang ZL, Tsou MC, Chen SY, Chiu GS, Chen WL, Wu YH, Wang LHC, Wang WC, Lin SY, Kung HJ, Wang LH, Cheng HC, Lin KT. Hydrogen sulfide coordinates glucose metabolism switch through destabilizing tetrameric pyruvate kinase M2. Nat Commun 2024; 15:7463. [PMID: 39198443 PMCID: PMC11358145 DOI: 10.1038/s41467-024-51875-9] [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: 09/02/2023] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
Most cancer cells reprogram their glucose metabolic pathway from oxidative phosphorylation to aerobic glycolysis for energy production. By reducing enzyme activity of pyruvate kinase M2 (PKM2), cancer cells attain a greater fraction of glycolytic metabolites for macromolecule synthesis needed for rapid proliferation. Here we demonstrate that hydrogen sulfide (H2S) destabilizes the PKM2 tetramer into monomer/dimer through sulfhydration at cysteines, notably at C326, leading to reduced PKM2 enzyme activity and increased PKM2-mediated transcriptional activation. Blocking PKM2 sulfhydration at C326 through amino acid mutation stabilizes the PKM2 tetramer and crystal structure further revealing the tetramer organization of PKM2-C326S. The PKM2-C326S mutant in cancer cells rewires glucose metabolism to mitochondrial respiration, significantly inhibiting tumor growth. In this work, we demonstrate that PKM2 sulfhydration by H2S inactivates PKM2 activity to promote tumorigenesis and inhibiting this process could be a potential therapeutic approach for targeting cancer metabolism.
Collapse
Grants
- National Science and Technology Council (Taiwan), 108-2314-B-007-003-MY3, 111-2320-B-007-005-MY3; National Tsing Hua University (NTHU), 111Q2713E1, 112Q2511E1, and 112Q2521E1, 113Q2524E1.
- National Science and Technology Council (Taiwan), 110-2320-B-007-004-MY3; National Health Research Institutes (Taiwan), NHRI-EX113-11124BI. National Tsing Hua University (NTHU), 112QI033E1
- National Science and Technology Council (Taiwan),110-2320-B-039-066; Ministry of Education (Taiwan), CMRC-CENTER-0
- National Science and Technology Council (Taiwan), 108-2311-B-007-002-MY3, 111-2311-B-007-009
Collapse
Affiliation(s)
- Rong-Hsuan Wang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Pin-Ru Chen
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yue-Ting Chen
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Chang Chen
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Hsin Chu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Chen Chien
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Po-Chen Chien
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Shao-Yun Lo
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Zhong-Liang Wang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Min-Chen Tsou
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ssu-Yu Chen
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Guang-Shen Chiu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Ling Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Yi-Hsuan Wu
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Lily Hui-Ching Wang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Ching Wang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Shu-Yi Lin
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan
| | - Hsing-Jien Kung
- College of Medical Science and Technology, PhD Program for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Lu-Hai Wang
- Chiese Medicine Research Center, and Institute of Integrated Medicine, China Medical University, Taichung City, Taiwan.
| | - Hui-Chun Cheng
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan.
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan.
| | - Kai-Ti Lin
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan.
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan.
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
32
|
Zhu H, Zhang H, Zhao XJ, Zhang L, Liu X, Zhang ZY, Ren YZ, Feng Y. Tetramerization of PKM2 Alleviates Traumatic Brain Injury by Ameliorating Mitochondrial Damage in Microglia. J Neuroimmune Pharmacol 2024; 19:48. [PMID: 39196455 DOI: 10.1007/s11481-024-10138-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 06/27/2024] [Indexed: 08/29/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. Microglial activation and neuroinflammation are key cellular events that determine the outcome of TBI, especially neuronal and cognitive function. Studies have suggested that the metabolic characteristics of microglia dictate their inflammatory response. The pyruvate kinase isoform M2 (PKM2), a key glycolytic enzyme, is involved in the regulation of various cellular metabolic processes, including mitochondrial metabolism. This suggests that PKM2 may also participate in the regulation of microglial activation during TBI. Therefore, the present study aimed to evaluate the role of PKM2 in regulating microglial activation and neuroinflammation and its effects on cognitive function following TBI. A controlled cortical impact (CCI) mouse model and inflammation-induced primary mouse microglial cells in vitro were used to investigate the potential effects of PKM2 inhibition and regulation. PKM2 was significantly increased during the acute and subacute phases of TBI and was predominantly detected in microglia rather than in neurons. Our results demonstrate that shikonin and TEPP-46 can inhibit microglial inflammation, improving mitochondria, improving mouse behavior, reducing brain defect volume, and alleviating pathological changes after TBI. There is a difference in the intervention of shikonin and TEPP-46 on PKM2. Shikonin directly inhibits General PKM2; TEPP-46 can promote the expression of PKM2 tetramer. In vitro experiments, TEPP-46 can promote the expression of PKM2 tetramer, enhance the interaction between PKM2 and MFN2, improve mitochondria, alleviate neuroinflammation. General inhibition and tetramerization activation of PKM2 attenuated cognitive function caused by TBI, whereas PKM2 tetramerization exhibited a better treatment effect. Our experiments demonstrated the non-metabolic role of PKM2 in the regulation of microglial activation following TBI. Both shikonin and TEPP-46 can inhibit pro-inflammatory factors, but only TEPP-46 can promote PKM2 tetramerization and upregulate the release of anti-inflammatory factors from microglia.
Collapse
Affiliation(s)
- Haiyan Zhu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Huiwen Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Xiao-Jing Zhao
- Department of Pathology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, China
| | - Lingyuan Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Xue Liu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Zhi-Yuan Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Yi-Zhi Ren
- Department of Clinical Genetics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China, 262 North Zhongshan Road.
| | - Yong Feng
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research &, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 210009, China, Baiziting 42.
| |
Collapse
|
33
|
Yang Y, Pu J, Yang Y. Glycolysis and chemoresistance in acute myeloid leukemia. Heliyon 2024; 10:e35721. [PMID: 39170140 PMCID: PMC11336864 DOI: 10.1016/j.heliyon.2024.e35721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
While traditional high-dose chemotherapy can effectively prolong the overall survival of acute myeloid leukemia (AML) patients and contribute to better prognostic outcomes, the advent of chemoresistance is a persistent challenge to effective AML management in the clinic. The therapeutic resistance is thought to emerge owing to the heterogeneous and adaptable nature of tumor cells when exposed to exogenous stimuli. Recent studies have focused on exploring metabolic changes that may afford novel opportunities to treat AML, with a particular focus on glycolytic metabolism. The Warburg effect, a hallmark of cancer, refers to metabolism of glucose through glycolysis under normoxic conditions, which contributes to the development of chemoresistance. Despite the key significance of this metabolic process in the context of malignant transformation, the underlying molecular mechanisms linking glycolysis to chemoresistance in AML remain incompletely understood. This review offers an overview of the current status of research focused on the relationship between glycolytic metabolism and AML resistance to chemotherapy, with a particular focus on the contributions of glucose transporters, key glycolytic enzymes, signaling pathways, non-coding RNAs, and the tumor microenvironment to this relationship. Together, this article will provide a foundation for the selection of novel therapeutic targets and the formulation of new approaches to treating AML.
Collapse
Affiliation(s)
- Yan Yang
- Department of Neonatology, Zigong Maternity and Child Health Care Hospital, Zigong, Sichuan, 643000, China
| | - Jianlin Pu
- Department of Psychiatry, The Zigong Affiliated Hospital of Southwest Medical University, Zigong mental health Center, Zigong Institute of Brain Science, Zigong, Sichuan, 643000, China
| | - You Yang
- Department of Pediatrics (Children Hematological Oncology), Birth Defects and Childhood Hematological Oncology Laboratory, The Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, 646000, China
- The Second Hospital, Centre for Reproductive Medicine, Advanced Medical Research Institute, Key Laboratory for Experimental Teratology of the Ministry of Education, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, China
| |
Collapse
|
34
|
Li S, Huo C, Liu A, Zhu Y. Mitochondria: a breakthrough in combating rheumatoid arthritis. Front Med (Lausanne) 2024; 11:1439182. [PMID: 39161412 PMCID: PMC11330793 DOI: 10.3389/fmed.2024.1439182] [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: 05/27/2024] [Accepted: 07/26/2024] [Indexed: 08/21/2024] Open
Abstract
As a chronic autoimmune disease with complex aetiology, rheumatoid arthritis (RA) has been demonstrated to be associated with mitochondrial dysfunction since mitochondrial dysfunction can affect the survival, activation, and differentiation of immune and non-immune cells involved in the pathogenesis of RA. Nevertheless, the mechanism behind mitochondrial dysfunction in RA remains uncertain. Accordingly, this review addresses the possible role and mechanisms of mitochondrial dysfunction in RA and discusses the potential and challenges of mitochondria as a potential therapeutic strategy for RA, thereby providing a breakthrough point in the prevention and treatment of RA.
Collapse
Affiliation(s)
- Shuang Li
- Graduate School of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Chenlu Huo
- Graduate School of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Anting Liu
- Graduate School of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yan Zhu
- Department of Geriatrics, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
35
|
Sun Y, Su X, Zhao L, Sun T, Liu W. Carbon metabolism of a novel isolate from Lacticaseibacillus rhamnosus Probio-M9 derived through space mutant. J Appl Microbiol 2024; 135:lxae205. [PMID: 39152088 DOI: 10.1093/jambio/lxae205] [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: 10/19/2023] [Revised: 07/07/2024] [Accepted: 08/15/2024] [Indexed: 08/19/2024]
Abstract
AIMS Carbon source is a necessary nutrient for bacterial strain growth. In industrial production, the cost of using different carbon sources varies greatly. Moreover, the complex environment in space may cause metabolic a series of changes in the strain, and this method has been successfully applied in some basic research. To date, space mutagenesis is still limited number of studies, particularly in carbon metabolism of probiotics. METHODS AND RESULTS HG-R7970-41 was isolated from bacterium suspension (Probio-M9) after space flight, which can produce capsular polysaccharide after space mutagenesis. Phenotype Microarray (PM) was used to evaluated the metabolism of HG-R7970-41 in 190 single carbon sources. RNA sequencing and total protein identification of two strains revealed their different carbon metabolism mechanisms. PM results demonstrated the metabolism of 10 carbon sources were different between Probio-M9 and HG-R7970-41. Transcriptomic and proteomic analyses revealed that this change in carbon metabolism of HG-R7970-41 mainly related to changes in phosphorylation and the glycolysis pathway. Based on the metabolic mechanism of different carbon sources and related gene cluster analysis, we found that the final metabolic activities of HG-R7970-41 and Probio-M9 were mainly regulated by PTS-specific membrane embedded permease, carbohydrate kinase and two rate-limiting enzymes (phosphofructokinase and pyruvate kinase) in the glycolysis pathway. The expanded culture test also confirmed that HG-R7970-41 had different metabolic characteristics from original strain. CONCLUSIONS These results suggested that space environment could change carbon metabolism of Probio-M9. The new isolate (HG-R7970-41) showed a different carbon metabolism pattern from the original strain mainly by the regulation of two rate-limiting enzymes.
Collapse
Affiliation(s)
- Yue Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
- College of Food Science and Technology, Wuhan Business University, Wuhan, Hubei province, 430056, China
| | - Xin Su
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Lixia Zhao
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Tiansong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Wenjun Liu
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| |
Collapse
|
36
|
Delgado Dolset MI, Pablo-Torres C, Contreras N, Couto-Rodríguez A, Escolar-Peña A, Graña-Castro O, Izquierdo E, López-Rodríguez JC, Macías-Camero A, Pérez-Gordo M, Villaseñor A, Zubeldia-Varela E, Barber D, Escribese MM. Severe Allergy as a Chronic Inflammatory Condition From a Systems Biology Perspective. Clin Exp Allergy 2024; 54:550-584. [PMID: 38938054 DOI: 10.1111/cea.14517] [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: 01/17/2024] [Revised: 05/14/2024] [Accepted: 05/26/2024] [Indexed: 06/29/2024]
Abstract
Persistent and unresolved inflammation is a common underlying factor observed in several and seemingly unrelated human diseases, including cardiovascular and neurodegenerative diseases. Particularly, in atopic conditions, acute inflammatory responses such as those triggered by insect venom, food or drug allergies possess also a life-threatening potential. However, respiratory allergies predominantly exhibit late immune responses associated with chronic inflammation, that can eventually progress into a severe phenotype displaying similar features as those observed in other chronic inflammatory diseases, as is the case of uncontrolled severe asthma. This review aims to explore the different facets and systems involved in chronic allergic inflammation, including processes such as tissue remodelling and immune cell dysregulation, as well as genetic, metabolic and microbiota alterations, which are common to other inflammatory conditions. Our goal here was to deepen on the understanding of an entangled disease as is chronic allergic inflammation and expose potential avenues for the development of better diagnostic and intervention strategies.
Collapse
Affiliation(s)
- M I Delgado Dolset
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - C Pablo-Torres
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - N Contreras
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Couto-Rodríguez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Escolar-Peña
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - O Graña-Castro
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - E Izquierdo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - J C López-Rodríguez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Macías-Camero
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - M Pérez-Gordo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Villaseñor
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - E Zubeldia-Varela
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - D Barber
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - M M Escribese
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| |
Collapse
|
37
|
Toledano Zur R, Atar O, Barliya T, Hoogi S, Abramovich I, Gottlieb E, Ron-Harel N, Cohen CJ. Genetically engineering glycolysis in T cells increases their antitumor function. J Immunother Cancer 2024; 12:e008434. [PMID: 38964783 PMCID: PMC11227835 DOI: 10.1136/jitc-2023-008434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND T cells play a central role in the antitumor response. However, they often face numerous hurdles in the tumor microenvironment, including the scarcity of available essential metabolites such as glucose and amino acids. Moreover, cancer cells can monopolize these resources to thrive and proliferate by upregulating metabolite transporters and maintaining a high metabolic rate, thereby outcompeting T cells. METHODS Herein, we sought to improve T-cell antitumor function in the tumor vicinity by enhancing their glycolytic capacity to better compete with tumor cells. To achieve this, we engineered human T cells to express a key glycolysis enzyme, phosphofructokinase, in conjunction with Glucose transporter 3, a glucose transporter. We co-expressed these, along with tumor-specific chimeric antigen or T-cell receptors. RESULTS Engineered cells demonstrated an increased cytokine secretion and upregulation of T-cell activation markers compared with control cells. Moreover, they displayed superior glycolytic capacity, which translated into an improved in vivo therapeutic potential in a xenograft model of human tumors. CONCLUSION In summary, these findings support the implementation of T-cell metabolic engineering to enhance the efficacy of cellular immunotherapies for cancer.
Collapse
Affiliation(s)
| | - Orna Atar
- Technion Israel Institute of Technology, Haifa, Haifa, Israel
| | | | | | - Ifat Abramovich
- Technion Israel Institute of Technology, Haifa, Haifa, Israel
| | - Eyal Gottlieb
- Technion Israel Institute of Technology, Haifa, Haifa, Israel
| | - Noga Ron-Harel
- Technion Israel Institute of Technology, Haifa, Haifa, Israel
| | - Cyrille J Cohen
- Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Tel Aviv, Israel
| |
Collapse
|
38
|
Fang Y, Li Z, Yang L, Li W, Wang Y, Kong Z, Miao J, Chen Y, Bian Y, Zeng L. Emerging roles of lactate in acute and chronic inflammation. Cell Commun Signal 2024; 22:276. [PMID: 38755659 PMCID: PMC11097486 DOI: 10.1186/s12964-024-01624-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 04/20/2024] [Indexed: 05/18/2024] Open
Abstract
Traditionally, lactate has been considered a 'waste product' of cellular metabolism. Recent findings have shown that lactate is a substance that plays an indispensable role in various physiological cellular functions and contributes to energy metabolism and signal transduction during immune and inflammatory responses. The discovery of lactylation further revealed the role of lactate in regulating inflammatory processes. In this review, we comprehensively summarize the paradoxical characteristics of lactate metabolism in the inflammatory microenvironment and highlight the pivotal roles of lactate homeostasis, the lactate shuttle, and lactylation ('lactate clock') in acute and chronic inflammatory responses from a molecular perspective. We especially focused on lactate and lactate receptors with either proinflammatory or anti-inflammatory effects on complex molecular biological signalling pathways and investigated the dynamic changes in inflammatory immune cells in the lactate-related inflammatory microenvironment. Moreover, we reviewed progress on the use of lactate as a therapeutic target for regulating the inflammatory response, which may provide a new perspective for treating inflammation-related diseases.
Collapse
Affiliation(s)
- Yunda Fang
- School of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhengjun Li
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- College of Health Economics Management, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lili Yang
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jingwen Library, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wen Li
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Acupuncture-Moxibustion and Tuina, ·School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yutong Wang
- School of Acupuncture-Moxibustion and Tuina, ·School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ziyang Kong
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Acupuncture-Moxibustion and Tuina, ·School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jia Miao
- School of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yanqi Chen
- School of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yaoyao Bian
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- School of Acupuncture-Moxibustion and Tuina, ·School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- TCM Rehabilitation Center, Jiangsu Second Chinese Medicine Hospital, Nanjing, 210023, China.
| | - Li Zeng
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, 999078, China.
| |
Collapse
|
39
|
Liang H, Fan X, Cheng H, Ma X, Sun Y, Nan F, Zhou J, Shu P, Zhang W, Zuo F, Nakatsukasa H, Zhang D. CPT-11 mitigates autoimmune diseases by suppressing effector T cells without affecting long-term anti-tumor immunity. Cell Death Discov 2024; 10:218. [PMID: 38704362 PMCID: PMC11069576 DOI: 10.1038/s41420-024-01983-8] [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: 11/08/2023] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/06/2024] Open
Abstract
The incidence of autoimmune diseases has significantly increased over the past 20 years. Excessive host immunoreactions and disordered immunoregulation are at the core of the pathogenesis of autoimmune diseases. The traditional anti-tumor chemotherapy drug CPT-11 is associated with leukopenia. Considering that CPT-11 induces leukopenia, we believe that it is a promising drug for the control of autoimmune diseases. Here, we show that CPT-11 suppresses T cell proliferation and pro-inflammatory cytokine production in healthy C57BL/6 mice and in complete Freund's adjuvant-challenged mice. We found that CPT-11 effectively inhibited T cell proliferation and Th1 and Th17 cell differentiation by inhibiting glycolysis in T cells. We also assessed CPT-11 efficacy in treating autoimmune diseases in models of experimental autoimmune encephalomyelitis and psoriasis. Finally, we proved that treatment of autoimmune diseases with CPT-11 did not suppress long-term immune surveillance for cancer. Taken together, these results show that CPT-11 is a promising immunosuppressive drug for autoimmune disease treatment.
Collapse
Affiliation(s)
- Hantian Liang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xinzou Fan
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hao Cheng
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiao Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yutong Sun
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fang Nan
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jingyang Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Panyin Shu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wei Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fengqiong Zuo
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Hiroko Nakatsukasa
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Dunfang Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
40
|
Escal J, Neel T, Hodin S, Boussoualim K, Amouzougan A, Coassy A, Locrelle H, Thomas T, Delavenne X, Marotte H. Proteomics analyses of human plasma reveal triosephosphate isomerase as a potential blood marker of methotrexate resistance in rheumatoid arthritis. Rheumatology (Oxford) 2024; 63:1368-1376. [PMID: 37527020 DOI: 10.1093/rheumatology/kead390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/02/2023] [Accepted: 07/09/2023] [Indexed: 08/03/2023] Open
Abstract
OBJECTIVE The objective of this study was to assess differentially expressed blood proteins between patients with active RA and patients in remission after MTX treatment, with the aim of identifying a biomarker of MTX resistance (MTXR). METHODS Two populations of RA patients treated with a stable dose of s.c. MTX for at least 3 months were constituted according to the DAS28: remission (DAS28 < 2.6; n = 24) and active disease (DAS28 > 3.2; n = 32). The two groups of RA patients were homogeneous regarding their epidemiological characteristics, except for the duration of treatment, which was longer in the remission group. After collection of a blood sample, plasma protein digestion was performed, followed by untargeted proteomics analysis. Then, a targeted analysis was performed to confirm the results of the untargeted approach. RESULTS Untargeted proteomics analysis revealed eight plasma proteins that were differentially expressed between the two groups of patients. Among them, triosephosphate isomerase (TPI-1) and glucose-6-phosphate isomerase (GPI), which are main actors in glycolysis, were found down-regulated in the active group. This result was confirmed for TPI-1 in the targeted proteomics analysis. CONCLUSION A first step was achieved in the search for biomarkers of MTXR, with the identification of two actors in glycolysis (TPI-1 and GPI). The next step will be to confirm these results in a larger cohort, including samples from treatment-naive patients, to assess the predictive potential of these protein markers.
Collapse
Affiliation(s)
- Jean Escal
- Université Jean Monnet Saint-Étienne, CHU Saint-Etienne, Laboratoire de Pharmacologie et Toxicologie, INSERM, SAINBIOSE U1059, Saint-Etienne, France
| | - Tiphany Neel
- CHU Saint-Étienne, Service de Rhumatologie, Saint-Etienne, France
| | - Sophie Hodin
- Université Jean Monnet Saint-Étienne, INSERM, SAINBIOSE U1059, Saint-Etienne, France
| | | | | | - Astrid Coassy
- CHU Saint-Étienne, Service de Rhumatologie, Saint-Etienne, France
| | - Hervé Locrelle
- CHU Saint-Étienne, Service de Rhumatologie, Saint-Etienne, France
| | - Thierry Thomas
- Université Jean Monnet Saint-Étienne, CHU Saint-Étienne, Service de Rhumatologie, Mines Saint-Etienne, INSERM, SAINBIOSE U1059, Saint-Etienne, France
| | - Xavier Delavenne
- Université Jean Monnet Saint-Étienne, CHU Saint-Etienne, Laboratoire de Pharmacologie et Toxicologie, INSERM, SAINBIOSE U1059, Saint-Etienne, France
| | - Hubert Marotte
- Université Jean Monnet Saint-Étienne, CHU Saint-Étienne, Service de Rhumatologie, Mines Saint-Etienne, INSERM, SAINBIOSE U1059, Saint-Etienne, France
| |
Collapse
|
41
|
Chen X, Hong L, Diao L, Yin T, Liu S. Hyperandrogenic environment regulates the function of ovarian granulosa cells by modulating macrophage polarization in PCOS. Am J Reprod Immunol 2024; 91:e13854. [PMID: 38716832 DOI: 10.1111/aji.13854] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/21/2024] [Accepted: 04/15/2024] [Indexed: 01/03/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common endocrine-metabolic disorder characterized by oligo-anovulation, hyperandrogenism, and polycystic ovaries, with hyperandrogenism being the most prominent feature of PCOS patients. However, whether excessive androgens also exist in the ovarian microenvironment of patients with PCOS, and their modulatory role on ovarian immune homeostasis and ovarian function, is not clear. METHODS Follicular fluid samples from patients participating in their first in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) treatment were collected. Androgen concentration of follicular fluid was assayed by chemiluminescence, and the macrophage M1:M2 ratio was detected by flow cytometry. In an in vitro model, we examined the regulatory effects of different concentrations of androgen on macrophage differentiation and glucose metabolism levels using qRT-PCR, Simple Western and multi-factor flow cytometry assay. In a co-culture model, we assessed the effect of a hyperandrogenic environment in the presence or absence of macrophages on the function of granulosa cells using qRT-PCR, Simple Western, EdU assay, cell cycle assay, and multi-factor flow cytometry assay. RESULTS The results showed that a significantly higher androgen level and M1:M2 ratio in the follicular fluid of PCOS patients with hyperandrogenism. The hyperandrogenic environment promoted the expression of pro-inflammatory and glycolysis-related molecules and inhibited the expression of anti-inflammatory and oxidative phosphorylation-related molecules in macrophages. In the presence of macrophages, a hyperandrogenic environment significantly downregulated the function of granulosa cells. CONCLUSION There is a hyperandrogenic microenvironment in the ovary of PCOS patients with hyperandrogenism. Hyperandrogenic microenvironment can promote the activation of ovarian macrophages to M1, which may be associated with the reprogramming of macrophage glucose metabolism. The increased secretion of pro-inflammatory cytokines by macrophages in the hyperandrogenic microenvironment would impair the normal function of granulosa cells and interfere with normal ovarian follicle growth and development.
Collapse
Affiliation(s)
- Xi Chen
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ling Hong
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
| | - Tailang Yin
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, Wuhan, China
| | - Su Liu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
| |
Collapse
|
42
|
Li F, Liu X, Li M, Wu S, Le Y, Tan J, Zhu C, Wan Q. Inhibition of PKM2 suppresses osteoclastogenesis and alleviates bone loss in mouse periodontitis. Int Immunopharmacol 2024; 129:111658. [PMID: 38359663 DOI: 10.1016/j.intimp.2024.111658] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/29/2024] [Accepted: 02/04/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND Chronic periodontitis triggers an increase in osteoclastogenesis, with glycolysis playing a crucial role in this process. Pyruvate kinase M2 (PKM2) is a critical enzyme involved in glycolysis and pyruvate metabolism. Yet, the precise function of PKM2 in osteoclasts and their formation remains unclear and requires further investigation. METHODS Bioinformatics was used to investigate critical biological processes in osteoclastogenesis. In vitro, osteoclastogenesis was analyzed using tartrate-resistant acid phosphatase (TRAP) staining, phalloidin staining, quantitative real‑time PCR (RT-qPCR), and Western blotting. Small interfering RNA (siRNA) of PKM2 and Shikonin, a specific inhibitor of PKM2, were used to verify the role of PKM2 in osteoclastogenesis. The mouse model of periodontitis was used to assess the effect of shikonin on bone loss. Analyses included micro computed tomography, immunohistochemistry, flow cytometry, TRAP staining and HE staining. RESULTS Bioinformatic analysis revealed a significant impact of glycolysis and pyruvate metabolism on osteoclastogenesis. Inhibition of PKM2 leads to a significant reduction in osteoclastogenesis. In vitro, co-culture of the heat-killed Porphyromonas gingivalis significantly promoted osteoclastogenesis, concomitant with an increased PKM2 expression in osteoclasts. Shikonin weakened the promoting effect of porphyromonas gingivalis on osteoclastogenesis. In vivo experiments demonstrated that inhibition of PKM2 by shikonin alleviated bone loss induced by periodontitis, suppressed excessive osteoclastogenesis in alveolar bone, and reduced tissue inflammation to some extent. CONCLUSION PKM2 inhibition by shikonin, a specific inhibitor of this enzyme, attenuated osteoclastogenesis and bone resorption in periodontitis. Shikonin appears to be a promising therapeutic agent for treating periodontitis.
Collapse
Affiliation(s)
- Feng Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China.
| | - Xinyuan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China.
| | - Mingjuan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China.
| | - Shuxuan Wu
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.
| | - Yushi Le
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China.
| | - Jingjing Tan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China.
| | - Chongjie Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China.
| | - Qilong Wan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China.
| |
Collapse
|
43
|
Dou Q, Grant AK, Coutinto de Souza P, Moussa M, Nasser I, Ahmed M, Tsai LL. Characterizing Metabolic Heterogeneity of Hepatocellular Carcinoma with Hyperpolarized 13C Pyruvate MRI and Mass Spectrometry. Radiol Imaging Cancer 2024; 6:e230056. [PMID: 38426887 PMCID: PMC10988335 DOI: 10.1148/rycan.230056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 12/16/2023] [Accepted: 01/11/2024] [Indexed: 03/02/2024]
Abstract
Purpose To characterize the metabolomic profiles of two hepatocellular carcinoma (HCC) rat models, track evolution of these profiles to a stimulated tumor state, and assess their effect on lactate flux with hyperpolarized (HP) carbon 13 (13C) MRI. Materials and Methods Forty-three female adult Fischer rats were implanted with N1S1 or McA-RH7777 HCC tumors. In vivo lactate-to-pyruvate ratio (LPR) was measured with HP 13C MRI at 9.4 T. Ex vivo mass spectrometry was used to measure intratumoral metabolites, and Ki67 labeling was used to quantify proliferation. Tumors were first compared with three normal liver controls. The tumors were then compared with stimulated variants via off-target hepatic thermal ablation treatment. All comparisons were made using the Mann-Whitney test. Results HP 13C pyruvate MRI showed greater LPR in N1S1 tumors compared with normal liver (mean [SD], 0.564 ± 0.194 vs 0.311 ± 0.057; P < .001 [n = 9]), but not for McA-RH7777 (P = .44 [n = 8]). Mass spectrometry confirmed that the glycolysis pathway was increased in N1S1 tumors and decreased in McA-RH7777 tumors. The pentose phosphate pathway was also decreased only in McA-RH7777 tumors. Increased proliferation in stimulated N1S1 tumors corresponded to a net increase in LPR (six stimulated vs six nonstimulated, 0.269 ± 0.148 vs 0.027 ± 0.08; P = .009), but not in McA-RH7777 (eight stimulated vs six nonstimulated, P = .13), despite increased proliferation and metastases. Mass spectrometry demonstrated relatively increased lactate production with stimulation in N1S1 tumors only. Conclusion Two HCC subtypes showed divergent glycolytic dependency at baseline and during transformation to a high proliferation state. This metabolic heterogeneity in HCC should be considered with use of HP 13C MRI for diagnosis and tracking. Keywords: Molecular Imaging-Probe Development, Liver, Abdomen/GI, Oncology, Hepatocellular Carcinoma © RSNA, 2024 See also commentary by Ohliger in this issue.
Collapse
Affiliation(s)
- Qianhui Dou
- From the Departments of Radiology (Q.D., A.K.G., M.M., M.A., L.L.T.)
and Pathology (I.N.), Beth Israel Deaconess Medical Center, Harvard Medical
School, 330 Brookline Ave, ANSIN 230, Boston, MA 02215; and Merck & Co,
Rahway, NJ (P.C.d.S.)
| | - Aaron K. Grant
- From the Departments of Radiology (Q.D., A.K.G., M.M., M.A., L.L.T.)
and Pathology (I.N.), Beth Israel Deaconess Medical Center, Harvard Medical
School, 330 Brookline Ave, ANSIN 230, Boston, MA 02215; and Merck & Co,
Rahway, NJ (P.C.d.S.)
| | - Patricia Coutinto de Souza
- From the Departments of Radiology (Q.D., A.K.G., M.M., M.A., L.L.T.)
and Pathology (I.N.), Beth Israel Deaconess Medical Center, Harvard Medical
School, 330 Brookline Ave, ANSIN 230, Boston, MA 02215; and Merck & Co,
Rahway, NJ (P.C.d.S.)
| | - Marwan Moussa
- From the Departments of Radiology (Q.D., A.K.G., M.M., M.A., L.L.T.)
and Pathology (I.N.), Beth Israel Deaconess Medical Center, Harvard Medical
School, 330 Brookline Ave, ANSIN 230, Boston, MA 02215; and Merck & Co,
Rahway, NJ (P.C.d.S.)
| | - Imad Nasser
- From the Departments of Radiology (Q.D., A.K.G., M.M., M.A., L.L.T.)
and Pathology (I.N.), Beth Israel Deaconess Medical Center, Harvard Medical
School, 330 Brookline Ave, ANSIN 230, Boston, MA 02215; and Merck & Co,
Rahway, NJ (P.C.d.S.)
| | - Muneeb Ahmed
- From the Departments of Radiology (Q.D., A.K.G., M.M., M.A., L.L.T.)
and Pathology (I.N.), Beth Israel Deaconess Medical Center, Harvard Medical
School, 330 Brookline Ave, ANSIN 230, Boston, MA 02215; and Merck & Co,
Rahway, NJ (P.C.d.S.)
| | - Leo L. Tsai
- From the Departments of Radiology (Q.D., A.K.G., M.M., M.A., L.L.T.)
and Pathology (I.N.), Beth Israel Deaconess Medical Center, Harvard Medical
School, 330 Brookline Ave, ANSIN 230, Boston, MA 02215; and Merck & Co,
Rahway, NJ (P.C.d.S.)
| |
Collapse
|
44
|
Jiang G, Hong J, Sun L, Wei H, Gong W, Wang S, Zhu J. Glycolysis regulation in tumor-associated macrophages: Its role in tumor development and cancer treatment. Int J Cancer 2024; 154:412-424. [PMID: 37688376 DOI: 10.1002/ijc.34711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/27/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023]
Abstract
Tumor-associated macrophages constitute the main cell population in the tumor microenvironment and play a crucial role in regulating the microenvironment composition. Emerging evidence has revealed that the metabolic profile determines the tumor-associated macrophage phenotype. Tumor-associated macrophage function is highly dependent on glucose metabolism, with glycolysis being the major metabolic pathway. Recent reports have demonstrated diversity in glucose flux of tumor-associated macrophages and complex substance communication with cancer cells. However, how the glucose flux in tumor-associated macrophages connects with glycolysis to influence tumor progression and the tumor microenvironment is still obscure. Moreover, while the development of single-cell sequencing technology allows a clearer and more accurate classification of tumor-associated macrophages, the metabolic profiles of tumor-associated macrophages from the perspective of single-cell omics has not been well summarized. Here, we review the current state of knowledge on glucose metabolism in tumor-associated macrophages and summarize the metabolic profiles of different tumor-associated macrophage subtypes from the perspective of single-cell omics. Additionally, we describe the current strategies targeting glycolysis in tumor-associated macrophages for cancer therapy.
Collapse
Affiliation(s)
- Guangyi Jiang
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| | - Junjie Hong
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| | - Lu Sun
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| | - Haibin Wei
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| | - Wangang Gong
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| | - Shu Wang
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, Hangzhou, China
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Jianqing Zhu
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| |
Collapse
|
45
|
Jiang D, Guo J, Liu Y, Li W, Lu D. Glycolysis: an emerging regulator of osteoarthritis. Front Immunol 2024; 14:1327852. [PMID: 38264652 PMCID: PMC10803532 DOI: 10.3389/fimmu.2023.1327852] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024] Open
Abstract
Osteoarthritis (OA) has been a leading cause of disability in the elderly and there remains a lack of effective therapeutic approaches as the mechanisms of pathogenesis and progression have yet to be elucidated. As OA progresses, cellular metabolic profiles and energy production are altered, and emerging metabolic reprogramming highlights the importance of specific metabolic pathways in disease progression. As a crucial part of glucose metabolism, glycolysis bridges metabolic and inflammatory dysfunctions. Moreover, the glycolytic pathway is involved in different areas of metabolism and inflammation, and is associated with a variety of transcription factors. To date, it has not been fully elucidated whether the changes in the glycolytic pathway and its associated key enzymes are associated with the onset or progression of OA. This review summarizes the important role of glycolysis in mediating cellular metabolic reprogramming in OA and its role in inducing tissue inflammation and injury, with the aim of providing further insights into its pathological functions and proposing new targets for the treatment of OA.
Collapse
Affiliation(s)
- Dingming Jiang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianan Guo
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yingquan Liu
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenxin Li
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Hangzhou Linping District Nanyuan Street Community Health Center, Hangzhou, China
| | - Dezhao Lu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
46
|
Kapnick SM, Martin CA, Jewell CM. Engineering metabolism to modulate immunity. Adv Drug Deliv Rev 2024; 204:115122. [PMID: 37935318 PMCID: PMC10843796 DOI: 10.1016/j.addr.2023.115122] [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: 01/14/2023] [Revised: 07/19/2023] [Accepted: 10/25/2023] [Indexed: 11/09/2023]
Abstract
Metabolic programming and reprogramming have emerged as pivotal mechanisms for altering immune cell function. Thus, immunometabolism has become an attractive target area for treatment of immune-mediated disorders. Nonetheless, many hurdles to delivering metabolic cues persist. In this review, we consider how biomaterials are poised to transform manipulation of immune cell metabolism through integrated control of metabolic configurations to affect outcomes in autoimmunity, regeneration, transplant, and cancer. We emphasize the features of nanoparticles and other biomaterials that permit delivery of metabolic cues to the intracellular compartment of immune cells, or strategies for altering signals in the extracellular space. We then provide perspectives on the potential for reciprocal regulation of immunometabolism by the physical properties of materials themselves. Lastly, opportunities for clinical translation are highlighted. This discussion contributes to our understanding of immunometabolism, biomaterials-based strategies for altering metabolic configurations in immune cells, and emerging concepts in this evolving field.
Collapse
Affiliation(s)
- Senta M Kapnick
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, USA; Department of Veterans Affairs, VA Maryland Health Care System, 10 N Green Street, Baltimore, MD, USA
| | - Corinne A Martin
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, USA; Department of Veterans Affairs, VA Maryland Health Care System, 10 N Green Street, Baltimore, MD, USA; Robert E. Fischell Institute for Biomedical Devices, 8278 Paint Branch Drive, College Park, MD, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, 22 S Greene Street, Suite N9E17, Baltimore, MD, USA.
| |
Collapse
|
47
|
Qin C, Xie T, Yeh WW, Savas AC, Feng P. Metabolic Enzymes in Viral Infection and Host Innate Immunity. Viruses 2023; 16:35. [PMID: 38257735 PMCID: PMC10820379 DOI: 10.3390/v16010035] [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: 11/21/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Metabolic enzymes are central players for cell metabolism and cell proliferation. These enzymes perform distinct functions in various cellular processes, such as cell metabolism and immune defense. Because viral infections inevitably trigger host immune activation, viruses have evolved diverse strategies to blunt or exploit the host immune response to enable viral replication. Meanwhile, viruses hijack key cellular metabolic enzymes to reprogram metabolism, which generates the necessary biomolecules for viral replication. An emerging theme arising from the metabolic studies of viral infection is that metabolic enzymes are key players of immune response and, conversely, immune components regulate cellular metabolism, revealing unexpected communication between these two fundamental processes that are otherwise disjointed. This review aims to summarize our present comprehension of the involvement of metabolic enzymes in viral infections and host immunity and to provide insights for potential antiviral therapy targeting metabolic enzymes.
Collapse
Affiliation(s)
- Chao Qin
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| | | | | | | | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
48
|
Meng F, Tao X, Li L, Jia W, Yang X, Yang Y. Network Pharmacology and Molecular Docking Explore the Mechanism of Mubiezi-Yinyanghuo Herb Pair in the Treatment of Rheumatoid Arthritis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2023; 2023:4502994. [PMID: 38106514 PMCID: PMC10723923 DOI: 10.1155/2023/4502994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/06/2023] [Accepted: 09/08/2023] [Indexed: 12/19/2023]
Abstract
Objective Our previous studies have shown that the Mubiezi-Yinyanghuo (MBZ-YYH) herb pair inhibits rheumatoid arthritis (RA) cell proliferation and glycolysis, promising results with an obscure mechanism of action. Methods Therefore, it is necessary to explore the main components of MBZ-YYH and unravel the potential mechanism in RA based on network pharmacology and molecular docking methods. Components and targets of MBZ-YYH were retrieved from the TCMSP. Relevant targets of RA were searched in GeneCards, therapeutic target database (TTD), and DisGeNET databases; the common targets of the MBZ-YYH compounds and RA were obtained by comparison; and a component-target interaction network was established by Cytoscape 3.9.1. Gene ontology (GO) analysis and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway enrichment analysis were performed through the David database. Molecular docking was performed by PyMoL2.3.0 and AutoDock Vina1.1.2 software. Results 7 active ingredients and 58 putatively identified target genes were screened from MBZ, and 16 effective components of YYH and 230 potential targets were identified. There were 29 mutual targets between the two herbs and RA. Through the PPI network, 9 hub targets which contain JUN, CASP3, PPARG, PTGS2, GSK3B, CASP8, HMOX1, ICAM1, and HK2 were screened out. GO term enrichment analysis indicated that positive regulation of the apoptotic process, response to drugs, and response to hypoxia were significantly enriched. Based on KEGG analysis, it was mainly associated with the IL-17 signaling pathway, the TNF signaling pathway, and the p53 signaling pathway. The docking analysis revealed that the effective components showed strong binding activity with the receptors. Conclusion The effects of the MBZ-YYH herb pair on RA were coordinated by the interaction of diverse components, which may be through the IL-17 signaling pathway and the TNF signaling pathway, which target GSK3B, HK2, caspase 3, and caspase 8, inhibiting the proliferation and glycolysis of rheumatoid arthritis fibroblast-like synovial cells (RA-FLS) and tending towards an increasing efficacy and decreasing toxicity effect on RA.
Collapse
Affiliation(s)
- Fuxue Meng
- The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou, China
| | - Xiaomai Tao
- The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou, China
- Guizhou Medical University, Guiyang, Guizhou, China
| | - Longkuan Li
- The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou, China
| | - Wei Jia
- The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou, China
| | - Xin Yang
- The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou, China
| | - Yuchen Yang
- The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou, China
| |
Collapse
|
49
|
He Y, Jiang S, Zhong Y, Wang X, Cui Y, Liang J, Sun Y, Zhu Z, Huang Z, Mao X. USP7 promotes non-small-cell lung cancer cell glycolysis and survival by stabilizing and activating c-Abl. Clin Transl Med 2023; 13:e1509. [PMID: 38082439 PMCID: PMC10713873 DOI: 10.1002/ctm2.1509] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Abelson tyrosine kinase (c-Abl) is frequently mutated and highly expressed, and promotes non-small-cell lung cancer (NSCLC) survival, metastasis and tumorigenesis. c-Abl could also be modified through ubiquitination, but the underlying mechanism is not well understood. METHODS Mass spectrometry assays were performed to search c-Abl deubiquitination enzymes. The molecular mechanism was determined using Co-IP assays, pull-down assays, Western blotting upon gene knockdown or overexpression. Cell lines and animal models were used to investigate the role of c-Abl and USP7 in NSCLC. EdU staining assay and Transwell assay were performed to evaluate the proliferation and migration ability of NSCLC cells, respectively. RESULTS Ubiquitin-specific protease 7 (USP7) is found to upregulate c-Abl via the deubiquitinase screen. USP7 interacts with c-Abl and decreases its K48-linked polyubiquitination, thereby increasing the stability of c-Abl. In addition to the wild-type one, c-Abl mutants can also be deubiquitinated and stabilized by USP7. Moreover, USP7 promotes c-Abl accumulation in cytoplasm by increasing its binding to 14-3-3α/β and activates the oncogenic c-Abl signalling pathway. Furthermore, the USP7/c-Abl axis promotes NSCLC cell glycolysis by direct phosphorylating and stabilizing hexokinase-2 (HK2). Knockdown of USP7 or c-Abl suppresses NSCLC cell glycolysis and reduces lactate production. Further studies revealed that overexpression of USP7 facilitates NSCLC cell growth and metastasis as well as xenograft growth in nude mice, while these activities are suppressed with USP7 or c-Abl being knocked down. CONCLUSIONS USP7 is a deubiquitinase of c-Abl and upregulates its oncogenic activity. USP7 promotes NSCLC cell metabolism by activating c-Abl and HK2. Targeting the USP7/c-Abl/HK2 axis might be a potential strategy to the precision therapy of NSCLC.
Collapse
Affiliation(s)
- Yuanming He
- Department of Hematology, The Key Laboratory of Advanced Interdisciplinary StudiesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouP. R. China
- Guangdong and Guangzhou Key Laboratory of Protein Modification and Degradation, School of Basic Medical SciencesGuangzhou Medical UniversityGuangzhouP. R. China
| | - Shuoyi Jiang
- Guangdong and Guangzhou Key Laboratory of Protein Modification and Degradation, School of Basic Medical SciencesGuangzhou Medical UniversityGuangzhouP. R. China
| | - Yueya Zhong
- Guangdong and Guangzhou Key Laboratory of Protein Modification and Degradation, School of Basic Medical SciencesGuangzhou Medical UniversityGuangzhouP. R. China
| | - Xiaoge Wang
- Guangdong and Guangzhou Key Laboratory of Protein Modification and Degradation, School of Basic Medical SciencesGuangzhou Medical UniversityGuangzhouP. R. China
| | - Yaoli Cui
- Guangdong and Guangzhou Key Laboratory of Protein Modification and Degradation, School of Basic Medical SciencesGuangzhou Medical UniversityGuangzhouP. R. China
| | - Jingpei Liang
- Guangdong and Guangzhou Key Laboratory of Protein Modification and Degradation, School of Basic Medical SciencesGuangzhou Medical UniversityGuangzhouP. R. China
| | - Yuening Sun
- Department of Hematology, The Key Laboratory of Advanced Interdisciplinary StudiesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouP. R. China
- Guangdong and Guangzhou Key Laboratory of Protein Modification and Degradation, School of Basic Medical SciencesGuangzhou Medical UniversityGuangzhouP. R. China
| | - Zhigang Zhu
- Division of Hematology & Oncology, Department of GeriatricsGuangzhou First People's Hospital, College of Medicine, South China University of TechnologyGuangzhouGuangdongP. R. China
| | - Zhenqian Huang
- Department of Hematology, The Key Laboratory of Advanced Interdisciplinary StudiesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouP. R. China
| | - Xinliang Mao
- Department of Hematology, The Key Laboratory of Advanced Interdisciplinary StudiesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouP. R. China
- GMU‐GIBH Joint School of Life Sciences, The Guangdong‐Hong Kong‐Macau Joint Laboratory for Cell Fate Regulation and DiseasesGuangzhou Medical UniversityGuangzhouP. R. China
| |
Collapse
|
50
|
Liu Y, Xing L, Zhang Y, Liu X, Li T, Zhang S, Wei H, Li J. Mild Intermittent Cold Stimulation Affects Cardiac Substance Metabolism via the Neuroendocrine Pathway in Broilers. Animals (Basel) 2023; 13:3577. [PMID: 38003194 PMCID: PMC10668735 DOI: 10.3390/ani13223577] [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: 09/04/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
This study aimed to investigate the impact of cold adaptation on the neuroendocrine and cardiac substance metabolism pathways in broilers. The broilers were divided into the control group (CC), cold adaptation group (C3), and cold-stressed group (C9), and experimental period was divided into the training period (d 1-35), recovery period (d 36-43), and cold stress period (d 43-44). During the training period, the CC group was reared at ambient temperature, while C3 and C9 groups were reared at 3 °C and 9 °C lower than the ambient temperature, respectively, for 5 h/d at 1 d intervals. During the recovery period, all the groups were maintained at 20 °C. Lastly, during the cold stress period, the groups were divided into two sub-groups, and each sub-group was placed at 10 °C for 12 h (Y12) or 24 h (Y24) for acute cold stimulation. The blood, hypothalamic, and cardiac tissues samples were obtained from all the groups during the training, recovery, and acute stress periods. The results revealed that the transcription of calcium voltage-gated channel subunit alpha 1 C (CACNAIC) was increased in the hypothalamic tissues of the C3 group (p < 0.05). Moreover, compared to the CC group, the serum norepinephrine (NE) was increased in the C9 group (p < 0.05), but insulin (INS) was decreased in the C9 group (p < 0.05). In addition, the transcription of the phosphoinositide-3 kinase (PI3K), protein kinase B (Akt), mammalian target of rapamycin (mTOR), SREBP1c, FASN, ACC1, and SCD genes was down-regulated in the C3 and C9 groups (p < 0.05); however, their expression increased in the C3 and C9 groups after acute cold stimulation (p < 0.05). Compared to the CC group, the transcription of forkhead box O1 (FoxO1), PEPCK, G6Pase, GLUT1, HK1, PFK, and LDHB genes was up-regulated in the C3 and C9 groups (p < 0.05. Furthermore, compared to the CC and C9 groups, the protein and mRNA expressions of heat shock protein (HSP) 70 and HSP90 were significantly increased in the C3 group (p < 0.05). These results indicate that intermittent cold training can enhance cold stress tolerance in broilers by regulating their neuroendocrine and cardiac substance metabolism pathways.
Collapse
Affiliation(s)
- Yuanyuan Liu
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Lu Xing
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Yong Zhang
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Xiaotao Liu
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Tingting Li
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Shijie Zhang
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Haidong Wei
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Jianhong Li
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China
| |
Collapse
|