1
|
Park SY, Song KH, Kang JH, Oh SH. Glucose transporter 2‑transported glucosamine inhibits glycolysis in cancer cell lines through competition with glucose for hexokinase II. Oncol Rep 2025; 53:73. [PMID: 40314081 PMCID: PMC12062862 DOI: 10.3892/or.2025.8906] [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: 11/19/2024] [Accepted: 03/17/2025] [Indexed: 05/03/2025] Open
Abstract
Antiproliferative effects of glucosamine, a glucose derivative with a similar structure to glucose, have been discovered, but the molecular mechanisms are not yet fully understood. Since glucosamine and glucose not only have similar structures but also are catalyzed by the same enzyme, hexokinase (HK), the present study delved into determining whether the antiproliferative effect of glucosamine involved the inhibition of glycolysis by competition with glucose. Whole‑genome screening analysis showed that a number of the gene pathways controlled by glucosamine were directly and indirectly involved in glycolysis. In vitro experiments revealed that as more glucose was added, the antiproliferative effect of glucosamine decreased. Also, it was found that glucosamine was transported into cells mainly through glucose transporter (GLUT) 2 which was responsible for the antiproliferative effects of glucosamine. In addition, the present study found that cancer cell lines with low expression level of HKII show high sensitivity to glucosamine and a HK inhibitor, 3‑bromopyruvate, enhanced the antiproliferative effect of glucosamine. Under hypoxic conditions, activated hypoxia‑inducible factor 1α (HIF‑1α) inducing glucose uptake and glycolysis hampered glucosamine‑induced cell death and HIF1A knockdown or HK inhibitors restored the antiproliferative effects of glucosamine. These findings demonstrated that glucosamine is an efficient glycolysis inhibitor and that GLUT2 and HKII play important roles as biomarkers for determining sensitivity to glucosamine. Moreover, the results suggested that the antiproliferative effect of glucosamine may be more efficient when administered in combination with other glycolytic agents or inhibitors targeting HIF‑1α.
Collapse
Affiliation(s)
- Se Yong Park
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Ki-Hoon Song
- ViroCure Inc., Guro, Seoul 08381, Republic of Korea
| | - Ju-Hee Kang
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Seung Hyun Oh
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
2
|
Akomolafe SF, Ajayi OO, Agboola OE, Adewale OO. Comparative evaluation of the antidiabetic potential of three varieties of Ipomoea batatas L.. Toxicol Rep 2025; 14:102015. [PMID: 40230512 PMCID: PMC11995110 DOI: 10.1016/j.toxrep.2025.102015] [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: 02/24/2025] [Revised: 03/13/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
Sweet potato (Ipomoea batatas L.) is a nutrient-dense tuber often used in traditional diabetic treatment. This research compares the antidiabetic potential of three sweet potato varieties: orange-fleshed (OFSP), purple-peel white-fleshed (PPWSP), and white-peel white-fleshed (WPWSP), utilising in vitro and in vivo techniques. Sweet potatoes (OFSP, PPWSP, and WPWSP) boiled at 100°C for 20 minutes were incorporated into formulated diets and administered to streptozotocin-induced diabetic rats for 14 days. Aqueous extracts of the diets were tested in vitro for antioxidants and phytochemicals. Glycaemic control parameters, lipid profiles, oxidative stress indicators, and pancreatic histology were investigated. Gene expression analysis was performed on critical diabetes-related pathways. OFSP showed significant strong anti-diabetic benefits, including better glycemic control, weight maintenance, lower HOMA-IR scores, and lowered α-amylase and α-glucosidase activity. OFSP-fed rats had higher insulin, glycogen, and hexokinase activity than those given PPWSP and WPWSP. OFSP decreased mRNA expression of DPP-4 while increasing GLP-1 expression. OFSP also improved lipid profiles, increasing HDLc while decreasing LDLc and triglycerides more than other varieties. Histopathological examination revealed restorative effects in pancreatic beta cells. OFSP demonstrated more pronounced antidiabetic effects compared to PPWSP and WPWSP, particularly in terms of glycemic control, insulin regulation, and lipid profile improvement. These findings suggest that OFSP may offer significant potential for diabetes management. However, further clinical studies are needed to validate these results and explore the practical dietary applications of OFSP in diabetes control.
Collapse
Affiliation(s)
- Seun F. Akomolafe
- Department of Biochemistry, Ekiti State University, Ado Ekiti, Ekiti State, PMB 5363, Nigeria
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Medical University of Warsaw, ul. Banacha 1, Warsaw 02-097, Poland
| | - Oluwadamilare O. Ajayi
- Department of Biochemistry, Ekiti State University, Ado Ekiti, Ekiti State, PMB 5363, Nigeria
| | - Oluwaseun E. Agboola
- Institute for Drug Research and Development, Afe Babalola University, Ado Ekiti, Nigeria
- DamSem Scientific Laboratory and Research, Oke-Ila, Ado Ekiti, Nigeria
| | | |
Collapse
|
3
|
Gao B, Lu Y, Lai X, Xu X, Gou S, Yang Z, Gong Y, Yang H. Metabolic reprogramming in hepatocellular carcinoma: mechanisms of immune evasion and therapeutic implications. Front Immunol 2025; 16:1592837. [PMID: 40370433 PMCID: PMC12075234 DOI: 10.3389/fimmu.2025.1592837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide, with limited treatment options for advanced stages. Metabolic reprogramming is a hallmark of cancer, enabling tumor cells to adapt to the harsh tumor microenvironment (TME) and evade immune surveillance. This review involves the role of metabolic reprogramming in HCC, focusing on the dysregulation of glucose, lipid, and amino acid metabolism, and its impact on immune evasion. Key metabolic pathways, such as the Warburg effect, fatty acid synthesis, and glutaminolysis, are discussed, along with their influence on tumor-associated macrophages (TAMs) and immune cell function. Targeting these metabolic alterations presents a promising therapeutic approach to enhance immunotherapy efficacy and improve HCC patient outcomes.
Collapse
Affiliation(s)
- Bocheng Gao
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Lu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xingyue Lai
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xi Xu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuhua Gou
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhida Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanju Gong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Yang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Chen W, Wang X, Li X, Wang W, Huang Y, Qin Y, Liu P, Wu K, Li B, He Y, Zeng S, Yi L, Wang L, Zhao M, Ding H, Fan S, Li Z, Chen J. Foot-and-mouth disease virus activates glycolysis and hijacks HK2 to inhibit innate immunity and promote viral replication. Vet Res 2025; 56:71. [PMID: 40170113 PMCID: PMC11963632 DOI: 10.1186/s13567-025-01497-w] [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: 10/22/2024] [Accepted: 01/23/2025] [Indexed: 04/03/2025] Open
Abstract
Foot-and-mouth disease (FMD) severely restricts the healthy development of global animal husbandry, and the unclear pathogenic mechanism of FMD virus (FMDV) leads to difficulty in preventing and purifying FMD. Glycolytic remodelling is considered one of the hallmarks of viral infection, providing energy and precursors for viral assembly and replication. In this work, the interaction and mechanism between FMDV and glycolysis were explored from the perspective of immune metabolism. We found that FMDV infection increased the extracellular acidification rate, lactic acid accumulation, and HK2 level. In addition, during FMDV infection, HK2 enhances glycolytic activity and mediates autophagic degradation of IRF3/7 to antagonize the innate immune response, thereby promoting viral replication. Our findings provide evidence that FMDV is closely correlated with host metabolism, increasing the understanding that glycolysis and HK2 facilitate virus infection, and provide new ideas for further elucidating the pathogenic mechanism of FMDV.
Collapse
Affiliation(s)
- Wenxian Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xinyan Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaowen Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Weijun Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yaoyao Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yuwei Qin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Pengfei Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Keke Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Bingke Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yintao He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Sen Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Lin Yi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | | | - Mingqiu Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hongxing Ding
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Shuangqi Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhaoyao Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
- Wen's Foodstuffs Group Co., Ltd., Yunfu, Xinxing, China.
| | - Jinding Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
5
|
Jiang Y, Wang Z, Zhang K, Hu Y, Shang D, Jiang L, Huang M, Wang B, He X, Wu Z, Yan X, Zhang X. Dynamin-Related Protein 1 Orchestrates Inflammatory Responses in Periodontal Macrophages via Interaction With Hexokinase 1. J Clin Periodontol 2025; 52:622-636. [PMID: 39762202 DOI: 10.1111/jcpe.14111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 11/09/2024] [Accepted: 12/15/2024] [Indexed: 03/29/2025]
Abstract
AIM To explore the potential roles of mitochondrial dysfunction in the initiation of inflammation in periodontal macrophages and to determine the mechanism underlying the involvement of dynamin-related protein 1 (Drp1) in macrophage inflammatory responses through its interaction with hexokinase 1 (HK1). MATERIALS AND METHODS Gingival tissues were collected from patients diagnosed with periodontitis or from healthy volunteers. Drp1 tetramer formation and phosphorylation were analysed using western blot. THP-1 macrophages and RAW264.7 cells were stimulated with Porphyromonas gingivalis (Pg) or Pg lipopolysaccharide (Pg LPS), respectively. Alterations in proteins associated with mitochondrial dynamics were scrutinized via western blot. Immunofluorescence was used to evaluate mitochondrial damage and mitochondrial permeability transition pore (mPTP) opening. Western blot was used to examine the inflammatory markers NLRP3, caspase-1, IL-1β and GSDMD. Protein interactions involving Drp1 were verified through immunoprecipitation. RESULTS In periodontitis patient samples, Pg LPS-treated RAW264.7 cells, and Pg-stimulated THP-1 macrophages, over-activated Drp1 was able to drive NLRP3 inflammasome activation and the subsequent release of inflammatory factors. A direct interaction between Drp1 and HK1 was observed, facilitating excessive mPTP opening and subsequent mitochondrial dysfunction. CONCLUSION In the inflammatory milieu of periodontal tissues, Drp1 hyperactivation in the macrophages is implicated in inflammation induction. Modulation of the inflammatory response in periodontal macrophages by Drp1 appears to facilitate mPTP opening.
Collapse
Affiliation(s)
- Yiming Jiang
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Zihan Wang
- Department of Oral Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Kaige Zhang
- Department of Oral Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Yue Hu
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Dehao Shang
- Department of Oral Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Lulu Jiang
- Department of Oral Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Minghao Huang
- Department of Oral Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Biyao Wang
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xiaomin He
- Department of Oral Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Zhou Wu
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Xu Yan
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xinwen Zhang
- Department of Oral Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
- Laboratory Animal Centre, School and Hospital of Stomatology, China Medical University, Shenyang, China
| |
Collapse
|
6
|
Bian W, Jiang X, Li J, Tillman L, Wang C, Zhen W, Weichselbaum RR, Fromme T, Lin W. Metal-organic layer delivers 3-bromopyruvate to mitochondria for metabolic regulation and cancer radio-immunotherapy. Chem Sci 2025; 16:5234-5240. [PMID: 39991561 PMCID: PMC11843481 DOI: 10.1039/d4sc08563a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/14/2025] [Indexed: 02/25/2025] Open
Abstract
Abnormal cancer metabolism causes hypoxia and immunosuppression, limiting the anti-tumor efficacy of radiotherapy. Herein, we report a positively charged, mitochondria-targeted nanoscale metal-organic layer conjugated with 3-bromopyruvate (BP), BP/Hf12-Ir, for metabolic reprogramming and radiosensitization. BP/Hf12-Ir disrupts oxidative phosphorylation and glycolysis, reducing energy production and alleviating hypoxia to enhance radiotherapy and anti-tumor immunity. BP/Hf12-Ir in combination with X-ray irradiation inhibits tumor growth by 95% and prevents lung metastasis in mouse models. When further combined with immune checkpoint blockade, this treatment induces robust anti-tumor immunity, achieving 98% tumor growth inhibition.
Collapse
Affiliation(s)
- Wangqing Bian
- Department of Chemistry, The University of Chicago Chicago Illinois 60637 USA
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich Freising Germany
| | - Xiaomin Jiang
- Department of Chemistry, The University of Chicago Chicago Illinois 60637 USA
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago Chicago Illinois 60637 USA
| | - Jinhong Li
- Department of Chemistry, The University of Chicago Chicago Illinois 60637 USA
| | - Langston Tillman
- Pritzker School of Molecular Engineering, The University of Chicago Chicago Illinois 60637 USA
| | - Chaoyu Wang
- Department of Chemistry, The University of Chicago Chicago Illinois 60637 USA
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago Chicago Illinois 60637 USA
| | - Wenyao Zhen
- Department of Chemistry, The University of Chicago Chicago Illinois 60637 USA
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago Chicago Illinois 60637 USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago Chicago Illinois 60637 USA
| | - Tobias Fromme
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich Freising Germany
- EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich Freising Germany
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago Chicago Illinois 60637 USA
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago Chicago Illinois 60637 USA
| |
Collapse
|
7
|
Bennett JJ, Saint-Martin C, Neumann B, Männistö JME, Houghton JAL, Empting S, Johnson MB, Laver TW, Locke JM, Spurrier B, Wakeling MN, Banerjee I, Dastamani A, Demirbilek H, Mitchell J, Stange M, Mohnike K, Arnoux JB, Owens NDL, Zenker M, Bellanné-Chantelot C, Flanagan SE. Non-coding cis-regulatory variants in HK1 cause congenital hyperinsulinism with variable disease severity. Genome Med 2025; 17:17. [PMID: 40033430 PMCID: PMC11874398 DOI: 10.1186/s13073-025-01440-w] [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: 09/03/2024] [Accepted: 02/14/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND We recently reported non-coding variants in a cis-regulatory element of the beta-cell disallowed gene hexokinase 1 (HK1) as a novel cause of congenital hyperinsulinism. These variants lead to a loss of repression of HK1 in pancreatic beta-cells, causing insulin secretion during hypoglycaemia. In this study, we aimed to determine the prevalence, genetics, and phenotype of HK1-hyperinsulinism by screening a large international cohort of patients living with the condition. METHODS We screened the HK1 cis-regulatory region in 1761 probands with hyperinsulinism of unknown aetiology who had been referred to one of three large European genomics laboratories. RESULTS We identified a HK1 variant in 89/1761 probands (5%) and 63 family members. Within the Exeter HI cohort, these variants accounted for 2.8% of all positive genetic diagnoses (n = 54/1913) establishing this as an important cause of HI. Individuals with a disease-causing variant were diagnosed with hyperinsulinism between birth and 26 years (median: 7 days) with variable response to treatment; 80% were medically managed and 20% underwent pancreatic surgery due to poor response to medical therapy. Glycaemic outcomes varied from spontaneous remission to hypoglycaemia persisting into adulthood. Eight probands had inherited the variant from a parent not reported to have hyperinsulinism (median current age: 39 years), confirming variable penetrance. Two of the 23 novel HK1 variants allowed us to extend the minimal cis-regulatory region from 42 to 46 bp. CONCLUSIONS Non-coding variants within the HK1 cis-regulatory region cause hyperinsulinism of variable severity ranging from neonatal-onset, treatment-resistant disease to being asymptomatic into adulthood. Discovering variants in 89 families confirms HK1 as a major cause of hyperinsulinism and highlights the important role of the non-coding genome in human monogenic disease.
Collapse
Affiliation(s)
- Jasmin J Bennett
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX2 5DW, UK
| | - Cécile Saint-Martin
- Department of Medical Genetics, AP-HP Sorbonne University, Pitié-Salpêtrière Hospital, 75013, Paris, France
| | - Bianca Neumann
- Institute of Human Genetics, University Hospital, Otto-Von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg, 39120, Germany
| | - Jonna M E Männistö
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX2 5DW, UK
- Kuopio Pediatric Research Unit (KuPRU), University of Eastern Finland, Kuopio, 70029, Finland
| | - Jayne A L Houghton
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX2 5DW, UK
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, EX2 5DW, UK
| | - Susann Empting
- Children's University Hospital, Otto-Von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg, 39120, Germany
| | - Matthew B Johnson
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX2 5DW, UK
| | - Thomas W Laver
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX2 5DW, UK
| | - Jonathan M Locke
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX2 5DW, UK
| | - Benjamin Spurrier
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX2 5DW, UK
| | - Matthew N Wakeling
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX2 5DW, UK
| | - Indraneel Banerjee
- Department of Paediatric Endocrinology, Royal Manchester Children's Hospital, Manchester, M13 9WL, UK
| | - Antonia Dastamani
- Endocrinology Department, Great Ormond Street Hospital for Children, London, WC1N 3JH, UK
| | - Hüseyin Demirbilek
- Faculty of Medicine, Department of Pediatric Endocrinology, Hacettepe University, Ankara, Turkey
| | - John Mitchell
- Pediatric Endocrinology and Biochemical Genetics, Human Genetics and Pediatrics, Montreal Children's Hospital-McGill University, McGill University, Montreal, Canada
| | - Markus Stange
- Department of Pediatrics, University Hospital Halle, Ernst Grube Str. 40, Halle, 06120, Germany
| | - Klaus Mohnike
- Children's University Hospital, Otto-Von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg, 39120, Germany
| | - Jean-Baptiste Arnoux
- Reference Center for Inherited Metabolic Diseases, Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, G2M, MetabERN, Paris Cité University, Paris, 75015, France
| | - Nick D L Owens
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX2 5DW, UK
| | - Martin Zenker
- Institute of Human Genetics, University Hospital, Otto-Von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg, 39120, Germany
| | | | - Sarah E Flanagan
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX2 5DW, UK.
| |
Collapse
|
8
|
Park S, Hall MN. Metabolic reprogramming in hepatocellular carcinoma: mechanisms and therapeutic implications. Exp Mol Med 2025; 57:515-523. [PMID: 40025169 PMCID: PMC11958682 DOI: 10.1038/s12276-025-01415-2] [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: 09/17/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 03/04/2025] Open
Abstract
Hepatocellular carcinoma features extensive metabolic reprogramming. This includes alterations in major biochemical pathways such as glycolysis, the pentose phosphate pathway, amino acid metabolism and fatty acid metabolism. Moreover, there is a complex interplay among these altered pathways, particularly involving acetyl-CoA (coenzyme-A) metabolism and redox homeostasis, which in turn influences reprogramming of other metabolic pathways. Understanding these metabolic changes and their interactions with cellular signaling pathways offers potential strategies for the targeted treatment of hepatocellular carcinoma and improved patient outcomes. This review explores the specific metabolic alterations observed in hepatocellular carcinoma and highlights their roles in the progression of the disease.
Collapse
Affiliation(s)
- Sujin Park
- Center for Genome Engineering, Institute for Basic Science, Daejeon, Republic of Korea.
| | | |
Collapse
|
9
|
Ham J, Yang W, Kim HY. Tissue-Specific Metabolic Reprogramming in Innate Lymphoid Cells and Its Impact on Disease. Immune Netw 2025; 25:e3. [PMID: 40078781 PMCID: PMC11896661 DOI: 10.4110/in.2025.25.e3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 03/14/2025] Open
Abstract
Recent advances have highlighted the crucial role of metabolic reprogramming in shaping the functions of innate lymphoid cells (ILCs), which are vital for tissue immunity and homeostasis. As tissue-resident cells, ILCs dynamically respond to local environmental cues, with tissue-derived metabolites such as short-chain fatty acids and amino acids directly modulating their effector functions. The metabolic states of ILC subsets-ILC1, ILC2, and ILC3-are closely linked to their ability to produce cytokines, sustain survival, and drive proliferation. This review provides a comprehensive analysis of how key metabolic pathways, including glycolysis, oxidative phosphorylation, and fatty acid oxidation, influence ILC activation and function. Furthermore, we explore the complex interactions between these metabolic pathways and tissue-specific metabolites, which can shape ILC-mediated immune responses in health and disease. Understanding these interactions reveals new insights into the pathogenesis of conditions such as asthma, inflammatory bowel disease, and cancer. A deeper understanding of these mechanisms may not only advance our knowledge of disease pathogenesis but also lead to the development of novel therapeutic strategies targeting metabolic pathways in ILCs to treat tissue-specific immune disorders.
Collapse
Affiliation(s)
- Jongho Ham
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Life Science, SRC Center for Multitasking Macrophage Research Center, Ewha Womans University, Seoul 03760, Korea
| | - Wooseok Yang
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Life Science, SRC Center for Multitasking Macrophage Research Center, Ewha Womans University, Seoul 03760, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
| |
Collapse
|
10
|
Jain A, Das R, Giri M, Mane P, Shard A. Carbohydrate kinase inhibition: a promising strategy in cancer treatment. Drug Discov Today 2025; 30:104308. [PMID: 39912130 DOI: 10.1016/j.drudis.2025.104308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/21/2025] [Accepted: 01/30/2025] [Indexed: 02/07/2025]
Abstract
Carbohydrate kinases (CKs) are pivotal in various biological processes, including energy consumption, cell signaling, and biosynthesis. They are a group of enzymes that facilitate the phosphorylation of carbohydrates, playing a crucial role in cellular metabolism. These enzymes facilitate the transfer of a phosphate group from a high-energy donor like ATP to a specified location on a carbohydrate substrate. Dysregulated kinase activity drives tumor growth and progression. Inhibitors targeting these enzymes have been developed and used in cancer therapy. The CK family encompasses three major types: hexokinases, ribokinases, and phosphatidylinositol kinases, with inhibitors of paramount importance in cancer treatment. This review explores the role of CKs in cancer and its inhibitors, providing insights into improving existing inhibitors and designing new ones.
Collapse
Affiliation(s)
- Archit Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Rudradip Das
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Muskan Giri
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Pranita Mane
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
11
|
Mady YH, Kalbermatter CG, Khan M, Schläfli AM, Mehmeti R, Zlobec I, Christe L, Tschan MP. Reliable hexokinase 3 protein detection in human cell lines and primary tissue. Eur J Histochem 2025; 69:4175. [PMID: 40071468 PMCID: PMC11956552 DOI: 10.4081/ejh.2025.4175] [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: 12/20/2024] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Accurate differentiation of homologous proteins that share high sequence identity remains a significant challenge in biomedical research, as conventional antibodies often lack sufficient specificity, leading to potential misinterpretations. This issue is particularly evident in the study of hexokinases, a family of isoenzymes that catalyze the first step of glycolysis by phosphorylating glucose. Beyond their canonical metabolic roles, hexokinases play critical non-glycolytic functions, especially in cancer biology. However, their unique tissue distributions and context-dependent roles are often obscured by the overlapping specificities of commercially available antibodies, which can produce misleading results. In this study, we rigorously evaluated a panel of antibodies targeting hexokinase isoenzyme 3 (HK3), highlighting the widespread issue of cross-reactivity and insufficient validation. Through this process, we identified and validated a highly specific antibody for HK3, demonstrating its reliability in western blot and immunohistochemistry applications. Using this validated tool, we reveal the distinct localization of HK3 in myeloid cell populations, providing new insights into its potential functional roles in these cells. This work addresses a critical gap in antibody specificity and establishes HK3 as a uniquely expressed gene in myeloid and immune cells and is absent in other cell types under basal conditions. Providing a foundation for future investigations into its context-dependent functions.
Collapse
Affiliation(s)
- Yasmeen H. Mady
- Institute of Tissue Medicine and Pathology, University of Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Carmen G. Kalbermatter
- Institute of Tissue Medicine and Pathology, University of Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Maarij Khan
- Institute of Tissue Medicine and Pathology, University of Bern, Switzerland
| | - Anna M. Schläfli
- Institute of Tissue Medicine and Pathology, University of Bern, Switzerland
| | - Rina Mehmeti
- Institute of Tissue Medicine and Pathology, University of Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Inti Zlobec
- Institute of Tissue Medicine and Pathology, University of Bern, Switzerland
| | - Lucine Christe
- Institute of Tissue Medicine and Pathology, University of Bern, Switzerland
| | - Mario P. Tschan
- Institute of Tissue Medicine and Pathology, University of Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| |
Collapse
|
12
|
Jiao J, Zhao Y, Li Q, Jin S, Liu Z. LncRNAs in tumor metabolic reprogramming and tumor microenvironment remodeling. Front Immunol 2024; 15:1467151. [PMID: 39539540 PMCID: PMC11557318 DOI: 10.3389/fimmu.2024.1467151] [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: 07/19/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
The tumor microenvironment (TME) is a complex and dynamic ecosystem composed of tumor cells, immune cells, supporting cells, and the extracellular matrix. Typically, the TME is characterized by an immunosuppressive state. To meet the demands of rapid proliferation, cancer cells undergo metabolic reprogramming, which enhances their biosynthesis and bioenergy supply. Immune cells require similar nutrients for activation and proliferation, leading to competition and immunosuppression within the TME. Additionally, tumor metabolites inhibit immune cell activation and function. Consequently, an immunosuppressed and immune-tolerant TME promotes cancer cell proliferation and metastasis. Long non-coding RNAs (lncRNAs), a category of non-coding RNA longer than 200 nucleotides, regulate tumor metabolic reprogramming by interacting with key enzymes, transporters, and related signaling pathways involved in tumor metabolism. Furthermore, lncRNAs can interact with both cellular and non-cellular components in the TME, thereby facilitating tumor growth, metastasis, drug resistance, and inducing immunosuppression. Recent studies have demonstrated that lncRNAs play a crucial role in reshaping the TME by regulating tumor metabolic reprogramming. In this discussion, we explore the potential mechanisms through which lncRNAs regulate tumor metabolic reprogramming to remodel the TME. Additionally, we examine the prospects of lncRNAs as targets for anti-tumor therapy and as biomarkers for tumor prognosis.
Collapse
Affiliation(s)
- Jianhang Jiao
- Department of Orthopedics, The Second Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Yangzhi Zhao
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Qimei Li
- Department of Radiation Oncology, The Second Affiliated Hospital of Jilin University, Changchun, China
| | - Shunzi Jin
- NHC Key Laboratory of Radiobiology, Jilin University, Changchun, China
| | - Zhongshan Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Paradoski BT, Hou S, Mejia EM, Olayinka-Adefemi F, Fowke D, Hatch GM, Saleem A, Banerji V, Hay N, Zeng H, Marshall AJ. PI3K-dependent reprogramming of hexokinase isoforms controls glucose metabolism and functional responses of B lymphocytes. iScience 2024; 27:110939. [PMID: 39635128 PMCID: PMC11615188 DOI: 10.1016/j.isci.2024.110939] [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: 03/07/2024] [Revised: 07/12/2024] [Accepted: 09/10/2024] [Indexed: 12/07/2024] Open
Abstract
B lymphocyte activation triggers metabolic reprogramming essential for B cell differentiation and mounting a healthy immune response. Here, we investigate the regulation and function of glucose-phosphorylating enzyme hexokinase 2 (HK2) in B cells. We report that both activation-dependent expression and mitochondrial localization of HK2 are regulated by the phosphatidylinositol 3-kinase (PI3K) signaling pathway. B cell-specific deletion of HK2 in mice caused mild perturbations in B cell development. HK2-deficient B cells show impaired functional responses in vitro and adapt to become less dependent on glucose and more dependent on glutamine. HK2 deficiency impairs glycolysis, alters metabolite profiles, and alters flux of labeled glucose carbons into downstream pathways. Upon immunization, HK2-deficient mice exhibit impaired germinal center, plasmablast, and antibody responses. HK2 expression in primary human chronic lymphocytic leukemia (CLL) cells was associated with recent proliferation and could be reduced by PI3K inhibition. Our study implicates PI3K-dependent modulation of HK2 in B cell metabolic reprogramming.
Collapse
Affiliation(s)
| | - Sen Hou
- Departments of Immunology, University of Manitoba, Winnipeg, Canada
| | - Edgard M. Mejia
- Departments of Immunology, University of Manitoba, Winnipeg, Canada
| | | | - Danielle Fowke
- Departments of Immunology, University of Manitoba, Winnipeg, Canada
| | - Grant M. Hatch
- Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Canada
- The Children’s Hospital Research Institute of Manitoba, Winnipeg, Canada
| | - Ayesha Saleem
- The Children’s Hospital Research Institute of Manitoba, Winnipeg, Canada
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, Canada
| | - Versha Banerji
- Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- Paul Albrechtsen Research Institute, Cancer Care Manitoba, Winnipeg, Canada
| | - Nissim Hay
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL, USA
| | - Hu Zeng
- Department of Immunology and Division of Rheumatology, Mayo Clinic, Rochester, MN, USA
| | - Aaron J. Marshall
- Departments of Immunology, University of Manitoba, Winnipeg, Canada
- Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Canada
- Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- Paul Albrechtsen Research Institute, Cancer Care Manitoba, Winnipeg, Canada
| |
Collapse
|
14
|
Manning BD, Dibble CC. Growth Signaling Networks Orchestrate Cancer Metabolic Networks. Cold Spring Harb Perspect Med 2024; 14:a041543. [PMID: 38438221 PMCID: PMC11444256 DOI: 10.1101/cshperspect.a041543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Normal cells grow and divide only when instructed to by signaling pathways stimulated by exogenous growth factors. A nearly ubiquitous feature of cancer cells is their capacity to grow independent of such signals, in an uncontrolled, cell-intrinsic manner. This property arises due to the frequent oncogenic activation of core growth factor signaling pathway components, including receptor tyrosine kinases, PI3K-AKT, RAS-RAF, mTORC1, and MYC, leading to the aberrant propagation of pro-growth signals independent of exogenous growth factors. The growth of both normal and cancer cells requires the acquisition of nutrients and their anabolic conversion to the primary macromolecules underlying biomass production (protein, nucleic acids, and lipids). The core growth factor signaling pathways exert tight regulation of these metabolic processes and the oncogenic activation of these pathways drive the key metabolic properties of cancer cells and tumors. Here, we review the molecular mechanisms through which these growth signaling pathways control and coordinate cancer metabolism.
Collapse
Affiliation(s)
- Brendan D Manning
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Christian C Dibble
- Department of Pathology, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
15
|
Khanijou JK, Hee YT, Scipion CPM, Chen X, Selvarajoo K. Systems biology approach for enhancing limonene yield by re-engineering Escherichia coli. NPJ Syst Biol Appl 2024; 10:109. [PMID: 39353984 PMCID: PMC11445242 DOI: 10.1038/s41540-024-00440-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/19/2024] [Indexed: 10/03/2024] Open
Abstract
Engineered microorganisms have emerged as viable alternatives for limonene production. However, issues such as low enzyme abundance or activities, and regulatory feedback/forward inhibition may reduce yields. To understand the underlying metabolism, we adopted a systems biology approach for an engineered limonene-producing Escherichia coli strain K-12 MG1655. Firstly, we generated time-series metabolomics data and, secondly, developed a dynamic model based on enzyme dynamics to track the native metabolic networks and the engineered mevalonate pathway. After several iterations of model fitting with experimental profiles, which also included 13C-tracer studies, we performed in silico knockouts (KOs) of all enzymes to identify bottleneck(s) for optimal limonene yields. The simulations indicated that ALDH/ADH (aldehyde dehydrogenase/alcohol dehydrogenase) and LDH (lactate dehydrogenase) suppression, and HK (hexokinase) enhancement would increase limonene yields. Experimental confirmation was achieved, where ALDH-ADH and LDH KOs, and HK overexpression improved limonene yield by 8- to 11-fold. Our systems biology approach can guide microbial strain re-engineering for optimal target production.
Collapse
Affiliation(s)
- Jasmeet Kaur Khanijou
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, Nanos, Singapore, 138669, Singapore
| | - Yan Ting Hee
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis St, Matrix, Singapore, 138671, Singapore
| | | | - Xixian Chen
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, Nanos, Singapore, 138669, Singapore
| | - Kumar Selvarajoo
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis St, Matrix, Singapore, 138671, Singapore.
- Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore (NUS), Singapore, Singapore.
- School of Biological Sciences, Nanyang Technological University (NTU), Singapore, Singapore.
| |
Collapse
|
16
|
Yang JK, Kim J, Ahn YH, Bae SH, Baek MJ, Lee SH, Moon JS. Inhibition of P2RX7 contributes to cytotoxicity by suppression of glycolysis and AKT activation in human hepatocellular carcinoma. BMB Rep 2024; 57:459-464. [PMID: 39219047 PMCID: PMC11524825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/07/2024] [Accepted: 09/01/2024] [Indexed: 09/04/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer. HCC occurs people with chronic liver diseases. The purinergic receptor P2X 7 (P2RX7) is involved in tumor proliferation and growth. Also, P2RX7 is associated with tumor invasion and metastatic dissemination. High glucose utilization is important for the survival of various types of tumors. However, the role of P2RX7 in glucose metabolism and cellular survival of HCC remains unclear. Here, our results show that the gene and protein levels of P2RX7 were elevated in tumor cells of patients with HCC. The pharmacological inhibition of P2RX7 by A-804598, a selective P2RX7 antagonist, and genetic inhibition by P2RX7 knockdown suppressed the glycolytic activity by reduction of hexokinase 2 (HK2), a key enzyme of the glycolysis pathway, in human HCC cells. Also, both A-804598 treatment and P2RX7 knockdown induced cytotoxicity via inhibition of AKT activation which is critical for tumor cell survival in human HCC cells. Moreover, A-804598 treatment and P2RX7 knockdown increased cytotoxicity and caspase-3 activation in human HCC cells. These results suggest that inhibition of P2RX7 contributes to cytotoxicity by suppression of glycolysis and AKT activation in human HCC. [BMB Reports 2024; 57(10): 459-464].
Collapse
Affiliation(s)
- Jae Kook Yang
- Department of Internal Medicine, Soonchunhyang University College of Medicine Cheonan Hospital, Cheonan 31151, Korea
| | - Junhyung Kim
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Korea
| | - Young Hyeon Ahn
- Department of Internal Medicine, Soonchunhyang University College of Medicine Cheonan Hospital, Cheonan 31151, Korea
| | - Sang Ho Bae
- Department of Surgery, Soonchunhyang University College of Medicine Cheonan Hospital, Cheonan 31151, Korea
| | - Moo-Jun Baek
- Department of Surgery, Soonchunhyang University College of Medicine Cheonan Hospital, Cheonan 31151, Korea
| | - Sae Hwan Lee
- Department of Internal Medicine, Soonchunhyang University College of Medicine Cheonan Hospital, Cheonan 31151, Korea
| | - Jong-Seok Moon
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Korea
- Department of Pathology, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
| |
Collapse
|
17
|
Ye Q, Gao C, Xiao H, Ruan S, Wang Y, Li X, Chang Y, Zhao C, Wang H, Han B, Ding J. Feeding Behavior, Gut Microbiota, and Transcriptome Analysis Reveal Individual Growth Differences in the Sea Urchin Strongylocentrotus intermedius. BIOLOGY 2024; 13:705. [PMID: 39336132 PMCID: PMC11428599 DOI: 10.3390/biology13090705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024]
Abstract
Growth differentiation among farmed sea urchins (Strongylocentrotus intermedius) poses a significant challenge to aquaculture, with there being a limited understanding of the underlying molecular mechanisms. In this study, sea urchins with varying growth rates, reared under identical conditions, were analyzed for feeding behavior, gut microbiota, and transcriptomes. Large-sized sea urchins demonstrated significantly higher feeding ability and longer duration than smaller ones. The dominant phyla across all size groups were Campylobacterota, Proteobacteria, and Firmicutes, with Campylobacterota showing the highest abundance in small-sized sea urchins (82.6%). However, the families Lachnospiraceae and Pseudomonadaceae were significantly less prevalent in small-sized sea urchins. Transcriptome analysis identified 214, 544, and 732 differentially expressed genes (DEGs) in the large vs. medium, large vs. small, and medium vs. small comparisons, respectively. Gene Ontology and KEGG pathway analyses associated DEGs with key processes such as steroid biosynthesis, protein processing within the endoplasmic reticulum, and nucleotide sugar metabolism. Variations in phagosomes and signaling pathways indicated that size differences are linked to disparities in energy expenditure and stress responses. These findings provide a foundation for future investigations into the regulatory mechanisms underlying growth differences in S. intermedius and provide clues for the screening of molecular markers useful to improve sea urchin production.
Collapse
Affiliation(s)
- Qi Ye
- Key Laboratory of Mariculture and Stock Enhancement in North China's Sea (Ministry of Agriculture and Rural Affairs), Dalian Ocean University, Dalian 116023, China
| | - Chuang Gao
- Key Laboratory of Mariculture and Stock Enhancement in North China's Sea (Ministry of Agriculture and Rural Affairs), Dalian Ocean University, Dalian 116023, China
| | - Haoran Xiao
- Key Laboratory of Mariculture and Stock Enhancement in North China's Sea (Ministry of Agriculture and Rural Affairs), Dalian Ocean University, Dalian 116023, China
| | - Shuchao Ruan
- Key Laboratory of Mariculture and Stock Enhancement in North China's Sea (Ministry of Agriculture and Rural Affairs), Dalian Ocean University, Dalian 116023, China
| | - Yongjie Wang
- Key Laboratory of Mariculture and Stock Enhancement in North China's Sea (Ministry of Agriculture and Rural Affairs), Dalian Ocean University, Dalian 116023, China
| | - Xiaonan Li
- Key Laboratory of Mariculture and Stock Enhancement in North China's Sea (Ministry of Agriculture and Rural Affairs), Dalian Ocean University, Dalian 116023, China
| | - Yaqing Chang
- Key Laboratory of Mariculture and Stock Enhancement in North China's Sea (Ministry of Agriculture and Rural Affairs), Dalian Ocean University, Dalian 116023, China
| | - Chong Zhao
- Key Laboratory of Mariculture and Stock Enhancement in North China's Sea (Ministry of Agriculture and Rural Affairs), Dalian Ocean University, Dalian 116023, China
| | - Heng Wang
- Key Laboratory of Mariculture and Stock Enhancement in North China's Sea (Ministry of Agriculture and Rural Affairs), Dalian Ocean University, Dalian 116023, China
| | - Bing Han
- Key Laboratory of Mariculture and Stock Enhancement in North China's Sea (Ministry of Agriculture and Rural Affairs), Dalian Ocean University, Dalian 116023, China
| | - Jun Ding
- Key Laboratory of Mariculture and Stock Enhancement in North China's Sea (Ministry of Agriculture and Rural Affairs), Dalian Ocean University, Dalian 116023, China
| |
Collapse
|
18
|
Fan J, Liu S, Ye W, Zhang X, Shi W. miR-483-5p-Containing exosomes treatment ameliorated deep vein thrombosis‑induced inflammatory response. Eur J Pharm Biopharm 2024; 202:114384. [PMID: 38950718 DOI: 10.1016/j.ejpb.2024.114384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024]
Abstract
Peripheral vascular condition, known as deep vein thrombosis (DVT), is a common ailment that may lead to deadly pulmonary embolism. Inflammation is closely connected to venous thrombosis, which results in blood stasis, leading to ischemia and hypoxia, as indicated by research. The objective of this research was to investigate the mechanism by which exosomes derived from adipose stem cells (ADSCs) prevent deep vein thrombosis. Our data showed that Exo-483 effectively reduced the thrombus weight in DVT rats by intravenous injection. Exo-483 decreased the expression of tissue factor (TF) protein, the influx of inflammatory cells into the thrombosed vein wall, and the levels of cytokines in the serum. Furthermore, Exo-483 suppressed the expression of Mitogen-activated protein kinase 1 (MAPK1) and decreased the expression of NLRP3 inflammasomes. In an oxygen-glucose deprivation (OGD) cell model, the tube-forming and migratory abilities of primary human umbilical vein endothelial cells (HUVEC) and EA.hy926 cells were suppressed by Exo-483 pretreatment.Exo-483 is also linked to regulating Dynamin-related protein 1 (DRP1) production downstream of MAPK1.By decreasing the mitochondrial localization and phosphorylation at the S616 site of DRP1, it diminishes the expression of NLRP3 inflammasomes. Moreover, according to Bioinformatics analysis, miR-483-5p was anticipated to target MAPK1. The research conducted by our team revealed that the miR-483-5p exosome derived from ADSCs exhibited anti-inflammatory properties through the modulation of downstream DRP1-NLRP3 expression by targeting MAPK1.The findings of this research propose that miR-483-5p may be regarded as an innovative treatment target for DVT.
Collapse
Affiliation(s)
- Jing Fan
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Sikai Liu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Wenhai Ye
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xiujin Zhang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Wanyin Shi
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| |
Collapse
|
19
|
Tong Y, Liu X, Wu L, Xiang Y, Wang J, Cheng Y, Zhang C, Han B, Wang L, Yan D. Hexokinase 2 nonmetabolic function-mediated phosphorylation of IκBα enhances pancreatic ductal adenocarcinoma progression. Cancer Sci 2024; 115:2673-2685. [PMID: 38801832 PMCID: PMC11309947 DOI: 10.1111/cas.16204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/29/2024] Open
Abstract
Aberrant signaling in tumor cells induces nonmetabolic functions of some metabolic enzymes in many cellular activities. As a key glycolytic enzyme, the nonmetabolic function of hexokinase 2 (HK2) plays a role in tumor immune evasion. However, whether HK2, dependent of its nonmetabolic activity, plays a role in human pancreatic ductal adenocarcinoma (PDAC) tumorigenesis remains unclear. Here, we demonstrated that HK2 acts as a protein kinase and phosphorylates IκBα at T291 in PDAC cells, activating NF-κB, which enters the nucleus and promotes the expression of downstream targets under hypoxia. HK2 nonmetabolic activity-promoted activation of NF-κB promotes the proliferation, migration, and invasion of PDAC cells. These findings provide new insights into the multifaceted roles of HK2 in tumor development and underscore the potential of targeting HK2 protein kinase activity for PDAC treatment.
Collapse
Affiliation(s)
- Yingying Tong
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Xin Liu
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Lihui Wu
- Zhejiang Provincial Key Laboratory of Pancreatic DiseaseThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Yaoxian Xiang
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Jing Wang
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Yurong Cheng
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Chan Zhang
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Baojuan Han
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Li Wang
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Dong Yan
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
20
|
Zhou L, Lian C, He Y, Chi X, Chen H, Zhong Z, Wang M, Cao L, Wang H, Zhang H, Li C. Toxicology assessment of deep-sea mining impacts on Gigantidas platifrons: A comparative in situ and laboratory metal exposure study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 933:173184. [PMID: 38750754 DOI: 10.1016/j.scitotenv.2024.173184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
Deep-sea toxicology is essential for deep-sea environmental impact assessment. Yet most toxicology experiments are conducted solely in laboratory settings, overlooking the complexities of the deep-sea environment. Here we carried out metal exposure experiments in both the laboratory and in situ, to compare and evaluate the response patterns of Gigantidas platifrons to metal exposure (copper [Cu] or cadmium [Cd] at 100 μg/L for 48 h). Metal concentrations, traditional biochemical parameters, and fatty acid composition were assessed in deep-sea mussel gills. The results revealed significant metal accumulation in deep-sea mussel gills in both laboratory and in situ experiments. Metal exposure could induce oxidative stress, neurotoxicity, an immune response, altered energy metabolism, and changes to fatty acid composition in mussel gills. Interestingly, the metal accumulating capability, biochemical response patterns, and fatty acid composition each varied under differing experimental systems. In the laboratory setting, Cd-exposed mussels exhibited a higher value for integrated biomarker response (IBR) while in situ the Cu-exposed mussels instead displayed a higher IBR value. This study emphasizes the importance of performing deep-sea toxicology experiments in situ and contributes valuable data to a standardized workflow for deep-sea toxicology assessment.
Collapse
Affiliation(s)
- Li Zhou
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Chao Lian
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Yameng He
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Xupeng Chi
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Hao Chen
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Zhaoshan Zhong
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Minxiao Wang
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Lei Cao
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Hao Wang
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Huan Zhang
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Chaolun Li
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 10049, China; Laoshan Laboratory, Qingdao 266237, China.
| |
Collapse
|
21
|
Lv X, Yang L, Xie Y, Momeni MR. Non-coding RNAs and exosomal non-coding RNAs in lung cancer: insights into their functions. Front Cell Dev Biol 2024; 12:1397788. [PMID: 38859962 PMCID: PMC11163066 DOI: 10.3389/fcell.2024.1397788] [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: 03/08/2024] [Accepted: 05/02/2024] [Indexed: 06/12/2024] Open
Abstract
Lung cancer is the second most common form of cancer worldwide Research points to the pivotal role of non-coding RNAs (ncRNAs) in controlling and managing the pathology by controlling essential pathways. ncRNAs have all been identified as being either up- or downregulated among individuals suffering from lung cancer thus hinting that they may play a role in either promoting or suppressing the spread of the disease. Several ncRNAs could be effective non-invasive biomarkers to diagnose or even serve as effective treatment options for those with lung cancer, and several molecules have emerged as potential targets of interest. Given that ncRNAs are contained in exosomes and are implicated in the development and progression of the malady. Herein, we have summarized the role of ncRNAs in lung cancer. Moreover, we highlight the role of exosomal ncRNAs in lung cancer.
Collapse
Affiliation(s)
- Xiaolong Lv
- Department of Cardiothoracic Surgery, The People’s Hospital of Changshou, Chongqing, China
| | - Lei Yang
- Department of Cardiothoracic Surgery, The People’s Hospital of Tongliang District, Chongqing, China
| | - Yunbo Xie
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | |
Collapse
|
22
|
Yu SK, Yu T, Wang YM, Sun A, Liu J, Lu KH. CCT6A facilitates lung adenocarcinoma progression and glycolysis via STAT1/HK2 axis. J Transl Med 2024; 22:460. [PMID: 38750462 PMCID: PMC11094951 DOI: 10.1186/s12967-024-05284-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Chaperonin Containing TCP1 Subunit 6 A (CCT6A) is a prominent protein involved in the folding and stabilization of newly synthesized proteins. However, its roles and underlying mechanisms in lung adenocarcinoma (LUAD), one of the most aggressive cancers, remain elusive. METHODS Our study utilized in vitro cell phenotype experiments to assess CCT6A's impact on the proliferation and invasion capabilities of LUAD cell lines. To delve into CCT6A's intrinsic mechanisms affecting glycolysis and proliferation in lung adenocarcinoma, we employed transcriptomic sequencing and liquid chromatography-mass spectrometry analysis. Co-immunoprecipitation (Co-IP) and chromatin immunoprecipitation (CHIP) assays were also conducted to substantiate the mechanism. RESULTS CCT6A was found to be significantly overexpressed in LUAD and associated with a poorer prognosis. The silencing of CCT6A inhibited the proliferation and migration of LUAD cells and elevated apoptosis rates. Mechanistically, CCT6A interacted with STAT1 protein, forming a complex that enhances the stability of STAT1 by protecting it from ubiquitin-mediated degradation. This, in turn, facilitated the transcription of hexokinase 2 (HK2), a critical enzyme in aerobic glycolysis, thereby stimulating LUAD's aerobic glycolysis and progression. CONCLUSION Our findings reveal that the CCT6A/STAT1/HK2 axis orchestrated a reprogramming of glucose metabolism and thus promoted LUAD progression. These insights position CCT6A as a promising candidate for therapeutic intervention in LUAD treatment.
Collapse
Affiliation(s)
- Shao-Kun Yu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Yu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu-Ming Wang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ao Sun
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jia Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kai-Hua Lu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
23
|
Das A, Martinez-Ruiz GU, Bouladoux N, Stacy A, Moraly J, Vega-Sendino M, Zhao Y, Lavaert M, Ding Y, Morales-Sanchez A, Harly C, Seedhom MO, Chari R, Awasthi P, Ikeuchi T, Wang Y, Zhu J, Moutsopoulos NM, Chen W, Yewdell JW, Shapiro VS, Ruiz S, Taylor N, Belkaid Y, Bhandoola A. Transcription factor Tox2 is required for metabolic adaptation and tissue residency of ILC3 in the gut. Immunity 2024; 57:1019-1036.e9. [PMID: 38677292 PMCID: PMC11096055 DOI: 10.1016/j.immuni.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/13/2024] [Accepted: 04/03/2024] [Indexed: 04/29/2024]
Abstract
Group 3 innate lymphoid cells (ILC3) are the major subset of gut-resident ILC with essential roles in infections and tissue repair, but how they adapt to the gut environment to maintain tissue residency is unclear. We report that Tox2 is critical for gut ILC3 maintenance and function. Gut ILC3 highly expressed Tox2, and depletion of Tox2 markedly decreased ILC3 in gut but not at central sites, resulting in defective control of Citrobacter rodentium infection. Single-cell transcriptional profiling revealed decreased expression of Hexokinase-2 in Tox2-deficient gut ILC3. Consistent with the requirement for hexokinases in glycolysis, Tox2-/- ILC3 displayed decreased ability to utilize glycolysis for protein translation. Ectopic expression of Hexokinase-2 rescued Tox2-/- gut ILC3 defects. Hypoxia and interleukin (IL)-17A each induced Tox2 expression in ILC3, suggesting a mechanism by which ILC3 adjusts to fluctuating environments by programming glycolytic metabolism. Our results reveal the requirement for Tox2 to support the metabolic adaptation of ILC3 within the gastrointestinal tract.
Collapse
Affiliation(s)
- Arundhoti Das
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Gustavo Ulises Martinez-Ruiz
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA; Faculty of Medicine, Research Division, National Autonomous University of Mexico, Mexico City, Mexico; Children's Hospital of Mexico Federico Gomez, Mexico City, Mexico
| | - Nicolas Bouladoux
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, NIAID, NIH, Bethesda, MD, USA
| | - Apollo Stacy
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, NIAID, NIH, Bethesda, MD, USA; Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Josquin Moraly
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Maria Vega-Sendino
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Yongge Zhao
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Marieke Lavaert
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Yi Ding
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Abigail Morales-Sanchez
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA; Children's Hospital of Mexico Federico Gomez, Mexico City, Mexico
| | - Christelle Harly
- Université de Nantes, CNRS, Inserm, CRCINA, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France
| | - Mina O Seedhom
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Raj Chari
- Genome Modification Core, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Parirokh Awasthi
- Mouse Modeling Core, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Tomoko Ikeuchi
- Oral Immunity and Infection Section, NIDCR, NIH, Bethesda, MD, USA
| | - Yueqiang Wang
- Shenzhen Typhoon HealthCare, Shenzhen, Guangdong, China
| | - Jinfang Zhu
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | | | - WanJun Chen
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD, USA
| | | | | | - Sergio Ruiz
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Naomi Taylor
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, NIAID, NIH, Bethesda, MD, USA
| | - Avinash Bhandoola
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA.
| |
Collapse
|
24
|
Simeroth S, Yu P. The role of lymphatic endothelial cell metabolism in lymphangiogenesis and disease. Front Cardiovasc Med 2024; 11:1392816. [PMID: 38798921 PMCID: PMC11119333 DOI: 10.3389/fcvm.2024.1392816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/18/2024] [Indexed: 05/29/2024] Open
Abstract
Lymphatic endothelial cells (LECs) line lymphatic vessels, which play an important role in the transport of lymph fluid throughout the human body. An organized lymphatic network develops via a process termed "lymphangiogenesis." During development, LECs respond to growth factor signaling to initiate the formation of a primary lymphatic vascular network. These LECs display a unique metabolic profile, preferring to undergo glycolysis even in the presence of oxygen. In addition to their reliance on glycolysis, LECs utilize other metabolic pathways such as fatty acid β-oxidation, ketone body oxidation, mitochondrial respiration, and lipid droplet autophagy to support lymphangiogenesis. This review summarizes the current understanding of metabolic regulation of lymphangiogenesis. Moreover, it highlights how LEC metabolism is implicated in various pathological conditions.
Collapse
Affiliation(s)
- Summer Simeroth
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Pengchun Yu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
25
|
Shi C, Wang L, Xu J, Li A, Wang C, Zhu X, Wang W, Yu Q, Han L. Effect of glycolysis on water holding capacity during postmortem aging of Jersey cattle-yak meat. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:3039-3046. [PMID: 38057148 DOI: 10.1002/jsfa.13195] [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: 10/19/2023] [Revised: 11/18/2023] [Accepted: 12/07/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Postmortem muscle moisture loss leads to a decrease in carcass weight and can adversely impact overall meat quality. Therefore, it is critical to investigate water holding capacity (WHC) to enhance meat quality. Current research has primarily focused on examining the correlation between signaling molecules and meat quality in relation to the glycolysis effect on muscle WHC. But there exists a significant knowledge gap regarding the mechanism of WHC in Jersey cattle-yak meat. RESULTS Jersey cattle-yak meat pH decreased and then increased during postmortem aging. Lactate content, cooking loss, pressing loss, drip loss and centrifuging loss of Jersey cattle-yak meat increased and then decreased during postmortem aging. The glycogen content of Jersey cattle-yak meat was significantly higher than that of yak meat at 6-120 h, being 8.40% higher than that of yak meat at 120 h. The activity of key glycolytic enzymes hexokinase (HK), pyruvate kinase (PK), phosphofructokinase (PFK) and lactate dehydrogenase (LDH) in Jersey cattle-yak meat was lower than that in yak meat. Correlation analysis showed that Jersey cattle-yak meat WHC was positively correlated with the activity of HK, PK, PFK and LDH. CONCLUSIONS The WHC of Jersey cattle-yak meat was higher than that of Gannan yak meat, and it was significantly positively correlated with the activity of key enzymes of the glycolytic signaling pathway. Therefore, the glycolysis rate can be reduced by inhibiting enzyme activity to improve Jersey cattle-yak meat WHC and meat quality. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Chaoxue Shi
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Linlin Wang
- College of Food Science and Technology, Southwest Minzu University, Chengdu, China
| | - Jin Xu
- Gannan Tibetan Autonomous Prefecture Animal Husbandry Technical Service Center, Gannan, China
| | - Aixia Li
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Changfeng Wang
- Wudu District Market Supervision Administration, Longnan, China
| | - Xijin Zhu
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Wanlin Wang
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Qunli Yu
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Ling Han
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
26
|
John S, Calmettes G, Xu S, Ribalet B. Real-time resolution studies of the regulation of lactate production by hexokinases binding to mitochondria in single cells. PLoS One 2024; 19:e0300150. [PMID: 38457438 PMCID: PMC10923494 DOI: 10.1371/journal.pone.0300150] [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: 11/24/2023] [Accepted: 02/21/2024] [Indexed: 03/10/2024] Open
Abstract
During hypoxia accumulation of lactate may be a key factor in acidosis-induced tissue damage. Binding of hexokinase (HK) to the outer membrane of mitochondria may have a protective effect under these conditions. We have investigated the regulation of lactate metabolism by hexokinases (HKs), using HEK293 cells in which the endogenous hexokinases have been knocked down to enable overexpression of wild type and mutant HKs. To assess the real-time changes in intracellular lactate levels the cells were also transfected with a lactate specific FRET probe. In the HKI/HKII double knockdown HEK cells, addition of extracellular pyruvate caused a large and sustained decrease in lactate. Upon inhibition of the mitochondrial electron transfer chain by NaCN this effect was reversed as a rapid increase in lactate developed which was followed by a slow and sustained increase in the continued presence of the inhibitor. Incubation of the HKI/HKII double knockdown HEK cells with the inhibitor of the malic enzyme, ME1*, blocked the delayed accumulation of lactate evoked by NaCN. With replacement by overexpression of HKI or HKII the accumulation of intracellular lactate evoked by NaCN was prevented. Blockage of the pentose phosphate pathway with the inhibitor 6-aminonicotinamide (6-AN) abolished the protective effect of HK expression, with NaCN causing again a sustained increase in lactate. The effect of HK was dependent on HK's catalytic activity and interaction with the mitochondrial outer membrane (MOM). Based on these data we propose that transformation of glucose into G6P by HK activates the pentose phosphate pathway which increases the production of NADPH, which then blocks the activity of the malic enzyme to transform malate into pyruvate and lactate.
Collapse
Affiliation(s)
- Scott John
- Department of Medicine (Division of Cardiology), David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Guillaume Calmettes
- Department of Medicine (Division of Cardiology), David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Shili Xu
- California NanoSystems Institute (CNSI) 2151, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Bernard Ribalet
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| |
Collapse
|
27
|
Sarkar A, Pawar SV, Chopra K, Jain M. Gamut of glycolytic enzymes in vascular smooth muscle cell proliferation: Implications for vascular proliferative diseases. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167021. [PMID: 38216067 DOI: 10.1016/j.bbadis.2024.167021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the media of the blood vessels and are responsible for maintaining vascular tone. Emerging evidence confirms that VSMCs possess high plasticity. During vascular injury, VSMCs switch from a "contractile" phenotype to an extremely proliferative "synthetic" phenotype. The balance between both strongly affects the progression of vascular remodeling in many cardiovascular pathologies such as restenosis, atherosclerosis and aortic aneurism. Proliferating cells demand high energy requirements and to meet this necessity, alteration in cellular bioenergetics seems to be essential. Glycolysis, fatty acid metabolism, and amino acid metabolism act as a fuel for VSMC proliferation. Metabolic reprogramming of VSMCs is dynamically variable that involves multiple mechanisms and encompasses the coordination of various signaling molecules, proteins, and enzymes. Here, we systemically reviewed the metabolic changes together with the possible treatments that are still under investigation underlying VSMC plasticity which provides a promising direction for the treatment of diseases associated with VSMC proliferation. A better understanding of the interaction between metabolism with associated signaling may uncover additional targets for better therapeutic strategies in vascular disorders.
Collapse
Affiliation(s)
- Ankan Sarkar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Sandip V Pawar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Kanwaljit Chopra
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Manish Jain
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| |
Collapse
|
28
|
Song H, Wang B, Zhao G, Lu S, Zhang D, Kong J, Li J, Zhang X, Lyu Y, Liu L. Discovery and biochemical characterization of two hexokinases from Crassostrea gigas. Protein Expr Purif 2024; 215:106408. [PMID: 38008389 DOI: 10.1016/j.pep.2023.106408] [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: 10/18/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 11/28/2023]
Abstract
Hexokinases (HKs) play a vital role in glucose metabolism, which controls the first committed step catalyzing the production of glucose-6-phosphate from glucose. Two HKs (CGIHK1 and CGIHK2) from the Pacific oyster Crassostrea giga were cloned and characterized. CGIHK1 and CGIHK2 were recombinantly expressed in Escherichia coli and successfully purified by the Ni-NTA column. The optimum pH of the two enzymes was pH 8.0 and 8.5, respectively. The optimum temperature of the two enzymes was 42 °C and 50 °C, respectively. Both enzymes showed a clear requirement for divalent magnesium and were strongly inhibited by SDS. CGIHK1 exhibited highly strict substrate specificity to glucose, while CGIHK2 could also catalyze other 11 monosaccharide substrates. This is the first report on the in vitro biosynthesis of glucose-6-phosphate by the hexokinases from Crassostrea gigas. The facile expression and purification procedures combined with different substrate specificities make CGIHK1 and CGIHK2 candidates for the biosynthesis of glucose-6-phosphate and other sugar-phosphates.
Collapse
Affiliation(s)
- Huibo Song
- College of Agricultural and Biological Engineering (College of Tree Peony), Heze University, Heze, 274015, China; Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Bo Wang
- College of Agricultural and Biological Engineering (College of Tree Peony), Heze University, Heze, 274015, China
| | - Guihong Zhao
- College of Agricultural and Biological Engineering (College of Tree Peony), Heze University, Heze, 274015, China.
| | - Shihai Lu
- Shandong Bigtree Dreyfus Special Meals Food Co., Ltd, Heze, 274000, China
| | - Dahu Zhang
- Shandong Bigtree Dreyfus Special Meals Food Co., Ltd, Heze, 274000, China
| | - Jianbiao Kong
- Heze Product Inspection and Testing Research Institute, Heze, 274000, China
| | - Jianxin Li
- Heze Institute for Food and Drug Control. Heze, 274000, China
| | - Xiaoyang Zhang
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, 224051, China
| | - Yongmei Lyu
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, 224051, China
| | - Li Liu
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
29
|
Ni Y, Zhuang Z. DDX24 promotes tumor progression by mediating hexokinase-1 induced glycolysis in gastric cancer. Cell Signal 2024; 114:110995. [PMID: 38043669 DOI: 10.1016/j.cellsig.2023.110995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/10/2023] [Accepted: 11/26/2023] [Indexed: 12/05/2023]
Abstract
Metabolic reprogramming allows tumor cells to meet high demand of biogenesis and increased energy for rapid proliferation. Gastric cancer (GC) ranks among the most prevalent malignancies globally. Exploring the underlying mechanisms of glycolytic reprogramming in GC could provide new therapeutic target for GC treatment. Here, we showed that DEAD-box helicase 24 (DDX24) played a critical role in hexokinase-1 (HK1) induced glycolysis. DDX24 expression was significantly elevated in GC tissues and was closely associated with worse survival in GC patients. In addition, DDX24 promoted glucose uptake and lactate production in GC cells. Mechanistically, DDX24 could bind the HK1 mRNA and positively regulated HK1 level at the transcriptional level. Moreover, DDX24 promoted the proliferation, migration, and invasion ability of GC cells by upregulating HK1. Collectively, these results suggested that DDX24 was a critical player in the regulation of glycolytic reprogramming and also implicated DDX24 as a valuable therapeutic target for GC.
Collapse
Affiliation(s)
- Yuanyuan Ni
- Department of Oncology, the Second Affiliated Hospital of Soochow University, Suzhou 215008, Jiangsu Province, PR China; Department of Radiation Oncology, the Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, Jiangsu Province, PR China
| | - Zhixiang Zhuang
- Department of Oncology, the Second Affiliated Hospital of Soochow University, Suzhou 215008, Jiangsu Province, PR China.
| |
Collapse
|
30
|
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
|
31
|
Wegener AJ, Hyer MM, Targett I, Kloster A, Shaw GA, Rodriguez AMM, Dyer SK, Neigh GN. Behavior, synaptic mitochondria, and microglia are differentially impacted by chronic adolescent stress and repeated endotoxin exposure in male and female rats. Stress 2024; 27:2299971. [PMID: 38179979 PMCID: PMC11064104 DOI: 10.1080/10253890.2023.2299971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/16/2023] [Indexed: 01/06/2024] Open
Abstract
Early life adversity and chronic inflammation have both been associated with cognitive impairment and neural compromise. In this study, we investigated the interactions between a history of chronic adolescent stress (CAS) and repeated endotoxin exposure on behavior, synaptic mitochondria, and microglia in adult male and female Wistar rats. Adult rats from chronic stress and control conditions were exposed to either repeated endotoxin (lipopolysaccharide; LPS) or saline injections every 3 days for 9 weeks. In both sexes, repeated LPS, regardless of stress history, impaired working memory in the Y maze. Regarding spatial memory, LPS impaired function for females; whereas, CAS altered function in males. Although males had an increase in anxiety-like behavior shortly after CAS, there were no long-term effects on anxiety-like behavior or social interaction observed in males or females. Stress did not alter synaptic mitochondrial function in either sex. Repeated LPS altered synaptic mitochondrial function such that ATP production was increased in females only. There were no observed increases in IBA-1 positive cells within the hippocampus for either sex. However, LPS and CAS altered microglia morphology in females. Impact of repeated LPS was evident at the terminal endpoint with increased spleen weight in both sexes and decreased adrenal weight in males only. Circulating cytokines were not impacted by repeated LPS at the terminal endpoint, but evidence of CAS effects on cytokines in females were evident. These data suggest a long-term impact of chronic stress and an impact of repeated endotoxin challenge in adulthood; however, not all physiological and behavioral metrics examined were impacted by the paradigm employed in this study and the two environmental challenges rarely interacted.
Collapse
Affiliation(s)
- A J Wegener
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - M M Hyer
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - I Targett
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - A Kloster
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - G A Shaw
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - A M M Rodriguez
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - S K Dyer
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - G N Neigh
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
32
|
Wang Q, Liu J, Chen Z, Zheng J, Wang Y, Dong J. Targeting metabolic reprogramming in hepatocellular carcinoma to overcome therapeutic resistance: A comprehensive review. Biomed Pharmacother 2024; 170:116021. [PMID: 38128187 DOI: 10.1016/j.biopha.2023.116021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/23/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Hepatocellular carcinoma (HCC) poses a heavy burden on human health with high morbidity and mortality rates. Systematic therapy is crucial for advanced and mid-term HCC, but faces a significant challenge from therapeutic resistance, weakening drug effectiveness. Metabolic reprogramming has gained attention as a key contributor to therapeutic resistance. Cells change their metabolism to meet energy demands, adapt to growth needs, or resist environmental pressures. Understanding key enzyme expression patterns and metabolic pathway interactions is vital to comprehend HCC occurrence, development, and treatment resistance. Exploring metabolic enzyme reprogramming and pathways is essential to identify breakthrough points for HCC treatment. Targeting metabolic enzymes with inhibitors is key to addressing these points. Inhibitors, combined with systemic therapeutic drugs, can alleviate resistance, prolong overall survival for advanced HCC, and offer mid-term HCC patients a chance for radical resection. Advances in metabolic research methods, from genomics to metabolomics and cells to organoids, help build the HCC metabolic reprogramming network. Recent progress in biomaterials and nanotechnology impacts drug targeting and effectiveness, providing new solutions for systemic therapeutic drug resistance. This review focuses on metabolic enzyme changes, pathway interactions, enzyme inhibitors, research methods, and drug delivery targeting metabolic reprogramming, offering valuable references for metabolic approaches to HCC treatment.
Collapse
Affiliation(s)
- Qi Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Juan Liu
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 100021, China; Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China; Institute for Organ Transplant and Bionic Medicine, Tsinghua University, Beijing 102218, China; Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, China.
| | - Ziye Chen
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Jingjing Zheng
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yunfang Wang
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 100021, China; Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China; Institute for Organ Transplant and Bionic Medicine, Tsinghua University, Beijing 102218, China; Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China; Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, China.
| | - Jiahong Dong
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun 130021, China; Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 100021, China; Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China; Institute for Organ Transplant and Bionic Medicine, Tsinghua University, Beijing 102218, China; Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, China.
| |
Collapse
|
33
|
Palma FR, Gantner BN, Sakiyama MJ, Kayzuka C, Shukla S, Lacchini R, Cunniff B, Bonini MG. ROS production by mitochondria: function or dysfunction? Oncogene 2024; 43:295-303. [PMID: 38081963 DOI: 10.1038/s41388-023-02907-z] [Citation(s) in RCA: 129] [Impact Index Per Article: 129.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/01/2023] [Accepted: 11/21/2023] [Indexed: 01/31/2024]
Abstract
In eukaryotic cells, ATP generation is generally viewed as the primary function of mitochondria under normoxic conditions. Reactive oxygen species (ROS), in contrast, are regarded as the by-products of respiration, and are widely associated with dysfunction and disease. Important signaling functions have been demonstrated for mitochondrial ROS in recent years. Still, their chemical reactivity and capacity to elicit oxidative damage have reinforced the idea that ROS are the products of dysfunctional mitochondria that accumulate during disease. Several studies support a different model, however, by showing that: (1) limited oxygen availability results in mitochondria prioritizing ROS production over ATP, (2) ROS is an essential adaptive mitochondrial signal triggered by various important stressors, and (3) while mitochondria-independent ATP production can be easily engaged by most cells, there is no known replacement for ROS-driven redox signaling. Based on these observations and other evidence reviewed here, we highlight the role of ROS production as a major mitochondrial function involved in cellular adaptation and stress resistance. As such, we propose a rekindled view of ROS production as a primary mitochondrial function as essential to life as ATP production itself.
Collapse
Affiliation(s)
- Flavio R Palma
- Department of Medicine, Division of Hematology Oncology, Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Chicago, Northwestern University, Chicago, IL, USA
| | - Benjamin N Gantner
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Marcelo J Sakiyama
- Department of Medicine, Division of Hematology Oncology, Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Chicago, Northwestern University, Chicago, IL, USA
| | - Cezar Kayzuka
- Department of Pharmacology, Ribeirao Preto College of Nursing, University of Sao Paulo, Sao Paulo, Brazil
| | - Sanjeev Shukla
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Sao Paulo, Brazil
| | - Brian Cunniff
- Department of Pathology and Laboratory Medicine, Larner School of Medicine, University of Vermont, Burlington, VT, USA
| | - Marcelo G Bonini
- Department of Medicine, Division of Hematology Oncology, Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Chicago, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
34
|
Liu B, Lu Y, Taledaohan A, Qiao S, Li Q, Wang Y. The Promoting Role of HK II in Tumor Development and the Research Progress of Its Inhibitors. Molecules 2023; 29:75. [PMID: 38202657 PMCID: PMC10779805 DOI: 10.3390/molecules29010075] [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: 10/26/2023] [Revised: 12/09/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Increased glycolysis is a key characteristic of malignant cells that contributes to their high proliferation rates and ability to develop drug resistance. The glycolysis rate-limiting enzyme hexokinase II (HK II) is overexpressed in most tumor cells and significantly affects tumor development. This paper examines the structure of HK II and the specific biological factors that influence its role in tumor development, as well as the potential of HK II inhibitors in antitumor therapy. Furthermore, we identify and discuss the inhibitors of HK II that have been reported in the literature.
Collapse
Affiliation(s)
- Bingru Liu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; (B.L.); (Y.L.); (A.T.)
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Yu Lu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; (B.L.); (Y.L.); (A.T.)
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
- Department of Core Facility Center, Capital Medical University, Beijing 100069, China
| | - Ayijiang Taledaohan
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; (B.L.); (Y.L.); (A.T.)
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Shi Qiao
- Civil Aviation Medical Center, Civil Aviation Administration of China, Beijing 100123, China;
| | - Qingyan Li
- Civil Aviation Medical Center, Civil Aviation Administration of China, Beijing 100123, China;
| | - Yuji Wang
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; (B.L.); (Y.L.); (A.T.)
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
- Department of Core Facility Center, Capital Medical University, Beijing 100069, China
| |
Collapse
|
35
|
Li Q, Chen Y, Liu H, Tian Y, Yin G, Xie Q. Targeting glycolytic pathway in fibroblast-like synoviocytes for rheumatoid arthritis therapy: challenges and opportunities. Inflamm Res 2023; 72:2155-2167. [PMID: 37940690 DOI: 10.1007/s00011-023-01807-y] [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: 04/08/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by hyperplastic synovium, pannus formation, immune cell infiltration, and potential articular cartilage damage. Notably, fibroblast-like synoviocytes (FLS), especially rheumatoid arthritis fibroblast-like synoviocytes (RAFLS), exhibit specific overexpression of glycolytic enzymes, resulting in heightened glycolysis. This elevated glycolysis serves to generate ATP and plays a pivotal role in immune regulation, angiogenesis, and adaptation to hypoxia. Key glycolytic enzymes, such as hexokinase 2 (HK2), phosphofructose-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), and pyruvate kinase M2 (PKM2), significantly contribute to the pathogenic behavior of RAFLS. This increased glycolysis activity is regulated by various signaling pathways. MATERIALS AND METHODS A comprehensive literature search was conducted to retrieve relevant studies published from January 1, 2010, to the present, focusing on RAFLS glycolysis, RA pathogenesis, glycolytic regulation pathways, and small-molecule drugs targeting glycolysis. CONCLUSION This review provides a thorough exploration of the pathological and physiological characteristics of three crucial glycolytic enzymes in RA. It delves into their putative regulatory mechanisms, shedding light on their significance in RAFLS. Furthermore, the review offers an up-to-date overview of emerging small-molecule candidate drugs designed to target these glycolytic enzymes and the upstream signaling pathways that regulate them. By enhancing our understanding of the pathogenic mechanisms of RA and highlighting the pivotal role of glycolytic enzymes, this study contributes to the development of innovative anti-rheumatic therapies.
Collapse
Affiliation(s)
- Qianwei Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuehong Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Huan Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yunru Tian
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Geng Yin
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.
- Department of General Practice, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
36
|
Chen J, Yang S, Li Y, Ziwen X, Zhang P, Song Q, Yao Y, Pei H. De novo nucleotide biosynthetic pathway and cancer. Genes Dis 2023; 10:2331-2338. [PMID: 37554216 PMCID: PMC10404870 DOI: 10.1016/j.gendis.2022.04.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/18/2022] [Indexed: 11/27/2022] Open
Abstract
De novo nucleotide biosynthetic pathway is a highly conserved and essential biochemical pathway in almost all organisms. Both purine nucleotides and pyrimidine nucleotides are necessary for cell metabolism and proliferation. Thus, the dysregulation of the de novo nucleotide biosynthetic pathway contributes to the development of many human diseases, such as cancer. It has been shown that many enzymes in this pathway are overactivated in different cancers. In this review, we summarize and update the current knowledge on the de novo nucleotide biosynthetic pathway, regulatory mechanisms, its role in tumorigenesis, and potential targeting opportunities.
Collapse
Affiliation(s)
- Jie Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430062, China
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057, USA
| | - Siqi Yang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430062, China
| | - Yingge Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430062, China
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057, USA
| | - Xu Ziwen
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057, USA
| | - Pingfeng Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430062, China
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430062, China
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430062, China
| | - Huadong Pei
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057, USA
| |
Collapse
|
37
|
Khalifa A, Guijarro A, Ravera S, Bertola N, Adorni MP, Papotti B, Raffaghello L, Benelli R, Becherini P, Namatalla A, Verzola D, Reverberi D, Monacelli F, Cea M, Pisciotta L, Bernini F, Caffa I, Nencioni A. Cyclic fasting bolsters cholesterol biosynthesis inhibitors' anticancer activity. Nat Commun 2023; 14:6951. [PMID: 37907500 PMCID: PMC10618279 DOI: 10.1038/s41467-023-42652-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 10/17/2023] [Indexed: 11/02/2023] Open
Abstract
Identifying oncological applications for drugs that are already approved for other medical indications is considered a possible solution for the increasing costs of cancer treatment. Under the hypothesis that nutritional stress through fasting might enhance the antitumour properties of at least some non-oncological agents, by screening drug libraries, we find that cholesterol biosynthesis inhibitors (CBIs), including simvastatin, have increased activity against cancers of different histology under fasting conditions. We show fasting's ability to increase CBIs' antitumour effects to depend on the reduction in circulating insulin, insulin-like growth factor-1 and leptin, which blunts the expression of enzymes from the cholesterol biosynthesis pathway and enhances cholesterol efflux from cancer cells. Ultimately, low cholesterol levels through combined fasting and CBIs reduce AKT and STAT3 activity, oxidative phosphorylation and energy stores in the tumour. Our results support further studies of CBIs in combination with fasting-based dietary regimens in cancer treatment and highlight the value of fasting for drug repurposing in oncology.
Collapse
Affiliation(s)
- Amr Khalifa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Ana Guijarro
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Silvia Ravera
- Department of Experimental Medicine, University of Genoa, Via Leon Battista Alberti 2, 16132, Genoa, Italy
| | - Nadia Bertola
- Department of Experimental Medicine, University of Genoa, Via Leon Battista Alberti 2, 16132, Genoa, Italy
| | - Maria Pia Adorni
- Department of Medicine and Surgery, University of Parma, 43125, Parma, Italy
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Lizzia Raffaghello
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Roberto Benelli
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Pamela Becherini
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
| | - Asmaa Namatalla
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
| | - Daniela Verzola
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
| | - Daniele Reverberi
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Michele Cea
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Livia Pisciotta
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Franco Bernini
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Irene Caffa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy.
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy.
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| |
Collapse
|
38
|
Rico A, Valls A, Guembelzu G, Azpitarte M, Aiastui A, Zufiria M, Jaka O, López de Munain A, Sáenz A. Altered expression of proteins involved in metabolism in LGMDR1 muscle is lost in cell culture conditions. Orphanet J Rare Dis 2023; 18:315. [PMID: 37817200 PMCID: PMC10565977 DOI: 10.1186/s13023-023-02873-5] [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: 06/20/2023] [Accepted: 08/24/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Limb-girdle muscular dystrophy R1 calpain 3-related (LGMDR1) is an autosomal recessive muscular dystrophy due to mutations in the CAPN3 gene. While the pathophysiology of this disease has not been clearly established yet, Wnt and mTOR signaling pathways impairment in LGMDR1 muscles has been reported. RESULTS A reduction in Akt phosphorylation ratio and upregulated expression of proteins implicated in glycolysis (HK-II) and in fructose and lactate transport (GLUT5 and MCT1) in LGMDR1 muscle was observed. In vitro analysis to establish mitochondrial and glycolytic functions of primary cultures were performed, however, no differences between control and patients were observed. Additionally, gene expression analysis showed a lack of correlation between primary myoblasts/myotubes and LGMDR1 muscle while skin fibroblasts and CD56- cells showed a slightly better correlation with muscle. FRZB gene was upregulated in all the analyzed cell types (except in myoblasts). CONCLUSIONS Proteins implicated in metabolism are deregulated in LGMDR1 patients' muscle. Obtained results evidence the limited usefulness of primary myoblasts/myotubes for LGMDR1 gene expression and metabolic studies. However, since FRZB is the only gene that showed upregulation in all the analyzed cell types it is suggested its role as a key regulator of the pathophysiology of the LGMDR1 muscle fiber. The Wnt signaling pathway inactivation, secondary to FRZB upregulation, and GLUT5 overexpression may participate in the impaired adipogenesis in LGMD1R patients.
Collapse
Affiliation(s)
- Anabel Rico
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, CIBER, Spanish Ministry of Science and Innovation, Carlos III Health Institute, Madrid, Spain
| | - Andrea Valls
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, CIBER, Spanish Ministry of Science and Innovation, Carlos III Health Institute, Madrid, Spain
| | - Garazi Guembelzu
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, CIBER, Spanish Ministry of Science and Innovation, Carlos III Health Institute, Madrid, Spain
| | - Margarita Azpitarte
- Cell Culture, Histology and Multidisciplinary 3D Printing Platform, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Ana Aiastui
- Department of Neurology, Donostialdea Integrated Health Organization, San Sebastián, Spain
| | - Mónica Zufiria
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, CIBER, Spanish Ministry of Science and Innovation, Carlos III Health Institute, Madrid, Spain
| | - Oihane Jaka
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, CIBER, Spanish Ministry of Science and Innovation, Carlos III Health Institute, Madrid, Spain
| | - Adolfo López de Munain
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, CIBER, Spanish Ministry of Science and Innovation, Carlos III Health Institute, Madrid, Spain
- Department of Neurology, Donostialdea Integrated Health Organization, San Sebastián, Spain
- Department of Neurosciences, University of the Basque Country UPV-EHU, San Sebastián, Spain
- Faculty of Medicine, University of Deusto, Bilbao, Spain
| | - Amets Sáenz
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián, Spain.
- CIBERNED, CIBER, Spanish Ministry of Science and Innovation, Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
39
|
Peng M, Li H, Cao H, Huang Y, Yu W, Shen C, Gu J. Dual FGFR and VEGFR inhibition synergistically restrain hexokinase 2-dependent lymphangiogenesis and immune escape in intrahepatic cholangiocarcinoma. J Gastroenterol 2023; 58:908-924. [PMID: 37433897 PMCID: PMC10423168 DOI: 10.1007/s00535-023-02012-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 06/18/2023] [Indexed: 07/13/2023]
Abstract
BACKGROUND Therapies for cholangiocarcinoma are largely limited and ineffective. Herein, we examined the role of the FGF and VEGF pathways in regulating lymphangiogenesis and PD-L1 expression in intrahepatic cholangiocarcinoma (iCCA). METHODS The lymphangiogenic functions of FGF and VEGF were evaluated in lymphatic endothelial cells (LECs) and iCCA xenograft mouse models. The relationship between VEGF and hexokinase 2 (HK2) was validated in LECs by western blot, immunofluorescence, ChIP and luciferase reporter assays. The efficacy of the combination therapy was assessed in LECs and xenograft models. Microarray analysis was used to evaluate the pathological relationships of FGFR1 and VEGFR3 with HK2 in human lymphatic vessels. RESULTS FGF promoted lymphangiogenesis through c-MYC-dependent modulation of HK2 expression. VEGFC also upregulated HK2 expression. Mechanistically, VEGFC phosphorylated components of the PI3K/Akt/mTOR axis to upregulate HIF-1α expression at the translational level, and HIF-1α then bound to the HK2 promoter region to activate its transcription. More importantly, dual FGFR and VEGFR inhibition with infigratinib and SAR131675 almost completely inhibited lymphangiogenesis, and significantly suppressed iCCA tumor growth and progression by reducing PD-L1 expression in LECs. CONCLUSIONS Dual FGFR and VEGFR inhibition inhibits lymphangiogenesis through suppression of c-MYC-dependent and HIF-1α-mediated HK2 expression, respectively. HK2 downregulation decreased glycolytic activity and further attenuated PD-L1 expression. Our findings suggest that dual FGFR and VEGFR blockade is an effective novel combination strategy to inhibit lymphangiogenesis and improve immunocompetence in iCCA.
Collapse
Affiliation(s)
- Min Peng
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, 210009, Jiangsu, China
| | - Hui Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Huan Cao
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yamei Huang
- Department of Pathology and Pathophysiology, Medical School of Southeast University, Nanjing, 210009, Jiangsu, China
| | - Weiping Yu
- Department of Pathology and Pathophysiology, Medical School of Southeast University, Nanjing, 210009, Jiangsu, China
| | - Chuanlai Shen
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, 210009, Jiangsu, China.
| | - Jinyang Gu
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Transplantation, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
40
|
Rho H, Terry AR, Chronis C, Hay N. Hexokinase 2-mediated gene expression via histone lactylation is required for hepatic stellate cell activation and liver fibrosis. Cell Metab 2023; 35:1406-1423.e8. [PMID: 37463576 PMCID: PMC11748916 DOI: 10.1016/j.cmet.2023.06.013] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/03/2023] [Accepted: 06/20/2023] [Indexed: 07/20/2023]
Abstract
Lactate was implicated in the activation of hepatic stellate cells (HSCs). However, the mechanism by which lactate exerts its effect remains elusive. Using RNA-seq and CUT&Tag chromatin profiling, we found that induction of hexokinase 2 (HK2) expression in activated HSCs is required for induced gene expression by histone lactylation but not histone acetylation. Inhibiting histone lactylation by Hk2 deletion or pharmacological inhibition of lactate production diminishes HSC activation, whereas exogenous lactate but not acetate supplementation rescues the activation phenotype. Thus, lactate produced by activated HSCs determines the HSC fate via histone lactylation. We found that histone acetylation competes with histone lactylation, which could explain why class I HDAC (histone deacetylase) inhibitors impede HSC activation. Finally, HSC-specific or systemic deletion of HK2 inhibits HSC activation and liver fibrosis in vivo. Therefore, we provide evidence that HK2 may be an effective therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Hyunsoo Rho
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Alexander R Terry
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Constantinos Chronis
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Nissim Hay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA; Research and Development Section, Jesse Brown VA Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
41
|
Catalano G, Zaza A, Banella C, Pelosi E, Castelli G, de Marinis E, Smigliani A, Travaglini S, Ottone T, Divona M, Del Principe MI, Buccisano F, Maurillo L, Ammatuna E, Testa U, Nervi C, Venditti A, Voso MT, Noguera NI. MCL1 regulates AML cells metabolism via direct interaction with HK2. Metabolic signature at onset predicts overall survival in AMLs' patients. Leukemia 2023; 37:1600-1610. [PMID: 37349598 DOI: 10.1038/s41375-023-01946-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/31/2023] [Accepted: 06/14/2023] [Indexed: 06/24/2023]
Abstract
We characterize the metabolic background in distinct Acute Myeloid Leukemias (AMLs), by comparing the metabolism of primary AML blasts isolated at diagnosis with that of normal hematopoietic maturing progenitors, using the Seahorse XF Agilent. Leukemic cells feature lower spare respiratory (SRC) and glycolytic capacities as compared to hematopoietic precursors (i.e. day 7, promyelocytes). According with Proton Leak (PL) values, AML blasts can be grouped in two well defined populations. The AML group with blasts presenting high PL or high basal OXPHOS plus high SRC levels had shorter overall survival time and significantly overexpressed myeloid cell leukemia 1 (MCL1) protein. We demonstrate that MCL1 directly binds to Hexokinase 2 (HK2) on the outer mitochondrial membrane (OMM). Overall, these results suggest that high PL and high SRC plus high basal OXPHOS levels at disease onset, arguably with the concourse of MCL1/HK2 action, are significantly linked with shorter overall survival time in AML. Our data describe a new function for MCL1 protein in AMLs' cells: by forming a complex with HK2, MCL1 co-localizes to VDAC on the OMM, thus inducing glycolysis and OXPHOS, ultimately conferring metabolic plasticity and promoting resistance to therapy.
Collapse
Affiliation(s)
- Gianfranco Catalano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy
| | - Alessandra Zaza
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy
| | - Cristina Banella
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy
- Department of Health Sciences, University of Florence and Meyer Children's University Hospital, Florence, Italy
| | - Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elisabetta de Marinis
- Department of Medical and Surgical Sciences and Biotechnologies, University of Roma La Sapienza, Rome, Italy
| | - Ariela Smigliani
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy
| | - Serena Travaglini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy
| | - Mariadomenica Divona
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | | - Francesco Buccisano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Luca Maurillo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Emanuele Ammatuna
- Department of Hematology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Clara Nervi
- Department of Hematology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Adriano Venditti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy.
| | - Nelida Ines Noguera
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy.
| |
Collapse
|
42
|
Lin J, Fang W, Xiang Z, Wang Q, Cheng H, Chen S, Fang J, Liu J, Wang Q, Lu Z, Ma L. Glycolytic enzyme HK2 promotes PD-L1 expression and breast cancer cell immune evasion. Front Immunol 2023; 14:1189953. [PMID: 37377974 PMCID: PMC10291184 DOI: 10.3389/fimmu.2023.1189953] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Immune therapies targeting the PD-1/PD-L1 pathway have been employed in the treatment of breast cancer, which requires aerobic glycolysis to sustain breast cancer cells growth. However, whether PD-L1 expression is regulated by glycolysis in breast cancer cells remains to be further elucidated. Here, we demonstrate that glycolytic enzyme hexokinase 2 (HK2) plays a crucial role in upregulating PD-L1 expression. Under high glucose conditions, HK2 acts as a protein kinase and phosphorylates IκBα at T291 in breast cancer cells, leading to the rapid degradation of IκBα and activation of NF-κB, which enters the nucleus and promotes PD-L1 expression. Immunohistochemistry staining of human breast cancer specimens and bioinformatics analyses reveals a positive correlation between HK2 and PD-L1 expression levels, which are inversely correlated with immune cell infiltration and survival time of breast cancer patients. These findings uncover the intrinsic and instrumental connection between aerobic glycolysis and PD-L1 expression-mediated tumor cell immune evasion and underscore the potential to target the protein kinase activity of HK2 for breast cancer treatment.
Collapse
Affiliation(s)
- Jichun Lin
- Department of Oncology, the Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wenshuo Fang
- Department of Oncology, the Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Zhuo Xiang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Qingqing Wang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Huapeng Cheng
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Shimin Chen
- Department of Oncology, the Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jing Fang
- Department of Oncology, the Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jia Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Qiang Wang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Leina Ma
- Department of Oncology, the Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| |
Collapse
|
43
|
Zong Y, Wang X, Cui B, Xiong X, Wu A, Lin C, Zhang Y. Decoding the regulatory roles of non-coding RNAs in cellular metabolism and disease. Mol Ther 2023; 31:1562-1576. [PMID: 37113055 PMCID: PMC10277898 DOI: 10.1016/j.ymthe.2023.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 04/29/2023] Open
Abstract
Non-coding RNAs, including long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), are being studied extensively in a variety of fields. Their roles in metabolism have received increasing attention in recent years but are not yet clear. The regulation of glucose, fatty acid, and amino acid metabolism is an imperative physiological process that occurs in living organisms and takes part in cancer and cardiovascular diseases. Here, we summarize the important roles played by non-coding RNAs in glucose metabolism, fatty acid metabolism, and amino acid metabolism, as well as the mechanisms involved. We also summarize the therapeutic advances for non-coding RNAs in diseases such as obesity, cardiovascular disease, and some metabolic diseases. Overall, non-coding RNAs are indispensable factors in metabolism and have a significant role in the three major metabolisms, which may be exploited as therapeutic targets in the future.
Collapse
Affiliation(s)
- Yuru Zong
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Xuliang Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Bing Cui
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Xiaowei Xiong
- Department of Cardiology and Macrovascular Disease, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Andrew Wu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chunru Lin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Yaohua Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
44
|
Rebane-Klemm E, Reinsalu L, Puurand M, Shevchuk I, Bogovskaja J, Suurmaa K, Valvere V, Moreno-Sanchez R, Kaambre T. Colorectal polyps increase the glycolytic activity. Front Oncol 2023; 13:1171887. [PMID: 37342183 PMCID: PMC10277630 DOI: 10.3389/fonc.2023.1171887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/23/2023] [Indexed: 06/22/2023] Open
Abstract
In colorectal cancer (CRC) energy metabolism research, the precancerous stage of polyp has remained rather unexplored. By now, it has been shown that CRC has not fully obtained the glycolytic phenotype proposed by O. Warburg and rather depends on mitochondrial respiration. However, the pattern of metabolic adaptations during tumorigenesis is still unknown. Understanding the interplay between genetic and metabolic changes that initiate tumor development could provide biomarkers for diagnosing cancer early and targets for new cancer therapeutics. We used human CRC and polyp tissue material and performed high-resolution respirometry and qRT-PCR to detect changes on molecular and functional level with the goal of generally describing metabolic reprogramming during CRC development. Colon polyps were found to have a more glycolytic bioenergetic phenotype than tumors and normal tissues. This was supported by a greater GLUT1, HK, LDHA, and MCT expression. Despite the increased glycolytic activity, cells in polyps were still able to maintain a highly functional OXPHOS system. The mechanisms of OXPHOS regulation and the preferred substrates are currently unclear and would require further investigation. During polyp formation, intracellular energy transfer pathways become rearranged mainly by increasing the expression of mitochondrial adenylate kinase (AK) and creatine kinase (CK) isoforms. Decreased glycolysis and maintenance of OXPHOS activity, together with the downregulation of the CK system and the most common AK isoforms (AK1 and AK2), seem to play a relevant role in CRC development.
Collapse
Affiliation(s)
- Egle Rebane-Klemm
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
- Department of Chemistry and Biotechnology, School of Science, Tallinn University of Technology, Tallinn, Estonia
| | - Leenu Reinsalu
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
- Department of Chemistry and Biotechnology, School of Science, Tallinn University of Technology, Tallinn, Estonia
| | - Marju Puurand
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Igor Shevchuk
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Jelena Bogovskaja
- Clinic of Diagnostics, North Estonia Medical Centre, Tallinn, Estonia
| | - Kulliki Suurmaa
- Department of Gastroenterology, West Tallinn Central Hospital, Tallinn, Estonia
| | - Vahur Valvere
- Oncology and Hematology Clinic, North Estonia Medical Centre, Tallinn, Estonia
| | - Rafael Moreno-Sanchez
- Laboratorio de Control Metabólico, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Los Reyes Iztacala, Barrio de los Árboles/Barrio de los Héroes, Tlalnepantla, Mexico
| | - Tuuli Kaambre
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| |
Collapse
|
45
|
Liu LX, Heng JH, Deng DX, Zhao H, Zheng ZY, Liao LD, Lin W, Xu XE, Li EM, Xu LY. Sulconazole induces PANoptosis by triggering oxidative stress and inhibiting glycolysis to increase radiosensitivity in esophageal cancer. Mol Cell Proteomics 2023; 22:100551. [PMID: 37076047 DOI: 10.1016/j.mcpro.2023.100551] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023] Open
Abstract
Esophageal cancer is the seventh most common cancer in the world. Although traditional treatment methods such as radiotherapy and chemotherapy have good effects, their side effects and drug resistance remain problematic. The repositioning of drug function provides new ideas for the research and development of anticancer drugs. We previously showed that the Food and Drug Administration (FDA)-approved drug sulconazole can effectively inhibit the growth of esophageal cancer cells, but its molecular mechanism is not clear. Here, our study demonstrated that sulconazole had a broad spectrum of anticancer effects. It can not only inhibit the proliferation, but also inhibit the migration of esophageal cancer cells. Both transcriptomic sequencing and proteomic sequencing showed that sulconazole could promote various types of programmed cell death and inhibit glycolysis and its related pathways. Experimentally, we found that sulconazole induced apoptosis, pyroptosis, necroptosis and ferroptosis. Mechanistically, sulconazole triggered mitochondrial oxidative stress and inhibited glycolysis. Finally, we showed that low-dose sulconazole can increase radiosensitivity of esophageal cancer cells. Taken together, these new findings provide strong laboratory evidence for the clinical application of sulconazole in esophageal cancer.
Collapse
Affiliation(s)
- Lu-Xin Liu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Jing-Hua Heng
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Dan-Xia Deng
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Hui Zhao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China; Guangdong Esophageal Cancer Research Institute, Shantou Sub-center, Cancer Research Center, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Zhen-Yuan Zheng
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China; Guangdong Esophageal Cancer Research Institute, Shantou Sub-center, Cancer Research Center, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Lian-Di Liao
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Wan Lin
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China; Guangdong Esophageal Cancer Research Institute, Shantou Sub-center, Cancer Research Center, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Xiu-E Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China.
| | - Li-Yan Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China; Guangdong Esophageal Cancer Research Institute, Shantou Sub-center, Cancer Research Center, Shantou University Medical College, Shantou 515041, Guangdong, China.
| |
Collapse
|
46
|
Cannabidiol alters mitochondrial bioenergetics via VDAC1 and triggers cell death in hormone-refractory prostate cancer. Pharmacol Res 2023; 189:106683. [PMID: 36736415 DOI: 10.1016/j.phrs.2023.106683] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/18/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023]
Abstract
In spite of the huge advancements in both diagnosis and interventions, hormone refractory prostate cancer (HRPC) remains a major hurdle in prostate cancer (PCa). Metabolic reprogramming plays a key role in PCa oncogenesis and resistance. However, the dynamics between metabolism and oncogenesis are not fully understood. Here, we demonstrate that two multi-target natural products, cannabidiol (CBD) and cannabigerol (CBG), suppress HRPC development in the TRansgenic Adenocarcinoma of the Mouse Prostate (TRAMP) model by reprogramming metabolic and oncogenic signaling. Mechanistically, CBD increases glycolytic capacity and inhibits oxidative phosphorylation in enzalutamide-resistant HRPC cells. This action of CBD originates from its effect on metabolic plasticity via modulation of VDAC1 and hexokinase II (HKII) coupling on the outer mitochondrial membrane, which leads to strong shifts of mitochondrial functions and oncogenic signaling pathways. The effect of CBG on enzalutamide-resistant HRPC cells was less pronounced than CBD and only partially attributable to its action on mitochondria. However, when optimally combined, these two cannabinoids exhibited strong anti-tumor effects in TRAMP mice, even when these had become refractory to enzalutamide, thus pointing to their therapeutical potential against PCa.
Collapse
|
47
|
He H, Xiao L, Wang J, Guo D, Lu Z. Aerobic glycolysis promotes tumor immune evasion and tumor cell stemness through the noncanonical function of hexokinase 2. Cancer Commun (Lond) 2023; 43:387-390. [PMID: 36604859 PMCID: PMC10009661 DOI: 10.1002/cac2.12404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/22/2022] [Accepted: 12/27/2022] [Indexed: 01/07/2023] Open
Affiliation(s)
- Haiyan He
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P. R. China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P. R. China.,Cancer Center, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Liwei Xiao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P. R. China.,Cancer Center, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Juhong Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China.,State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Dong Guo
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P. R. China.,Cancer Center, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P. R. China.,Cancer Center, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| |
Collapse
|
48
|
The Role of Reprogrammed Glucose Metabolism in Cancer. Metabolites 2023; 13:metabo13030345. [PMID: 36984785 PMCID: PMC10051753 DOI: 10.3390/metabo13030345] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Cancer cells reprogram their metabolism to meet biosynthetic needs and to adapt to various microenvironments. Accelerated glycolysis offers proliferative benefits for malignant cells by generating glycolytic products that move into branched pathways to synthesize proteins, fatty acids, nucleotides, and lipids. Notably, reprogrammed glucose metabolism and its associated events support the hallmark features of cancer such as sustained cell proliferation, hijacked apoptosis, invasion, metastasis, and angiogenesis. Overproduced enzymes involved in the committed steps of glycolysis (hexokinase, phosphofructokinase-1, and pyruvate kinase) are promising pharmacological targets for cancer therapeutics. In this review, we summarize the role of reprogrammed glucose metabolism in cancer cells and how it can be manipulated for anti-cancer strategies.
Collapse
|
49
|
Su W, Li J, Jiang L, Lei L, Li H. Hexokinase 2-mediated glycolysis supports inflammatory responses to Porphyromonas gingivalis in gingival fibroblasts. BMC Oral Health 2023; 23:103. [PMID: 36793034 PMCID: PMC9933269 DOI: 10.1186/s12903-023-02807-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND When infected with Porphyromonas gingivalis, gingival fibroblasts undergo metabolic reprogramming, and rely on aerobic glycolysis rather than oxidative phosphorylation for rapid energy replenishment. Hexokinases (HKs) are catalysts for glucose metabolism, and HK2 constitutes the major HK inducible isoform. The objective of this study is to determine whether HK2-mediated glycolysis promotes inflammatory responses in inflamed gingiva. METHODS Levels of glycolysis-related genes were assessed in normal and inflamed gingiva. Human gingival fibroblasts were harvested and infected with Porphyromonas gingivalis in order to mimic periodontal inflammation. 2-deoxy-d-glucose, an analogue of glucose, was used to block HK2-mediated glycolysis, while small interfering RNA was used to knock down HK2 expression. The mRNA and protein levels of genes were analyzed by real-time quantitative PCR and western blotting, respectively. HK2 activity and lactate production were assessed by ELISA. Cell proliferation was assessed by confocal microscopy. The generation of reactive oxygen species was assessed by flow cytometry. RESULTS Elevated expression of HK2 and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 was observed in the inflamed gingiva. P. gingivalis infection was shown to promote glycolysis in human gingival fibroblasts, as evidenced by increased gene transcription of HK2 and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3, cell glucose consumption, and HK2 activity. Inhibition and knockdown of HK2 resulted in reduced cytokine production, cell proliferation, and reactive oxygen species generation. Furthermore, P. gingivalis infection activated the hypoxia-inducible factor-1α signaling pathway, thus promoting HK2-mediated glycolysis and proinflammatory responses. CONCLUSIONS HK2-mediated glycolysis promotes inflammatory responses in gingival tissues, and therefore glycolysis can be targeted in order to inhibit the progression of periodontal inflammation.
Collapse
Affiliation(s)
- Wenqi Su
- grid.41156.370000 0001 2314 964XDepartment of Periodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, #30 Zhongyang Road, Nanjing, 210008 Jiangsu China ,grid.41156.370000 0001 2314 964XCentral Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jingwen Li
- grid.41156.370000 0001 2314 964XDepartment of Periodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, #30 Zhongyang Road, Nanjing, 210008 Jiangsu China ,grid.41156.370000 0001 2314 964XCentral Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lishan Jiang
- grid.41156.370000 0001 2314 964XDepartment of Periodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, #30 Zhongyang Road, Nanjing, 210008 Jiangsu China ,grid.41156.370000 0001 2314 964XCentral Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lang Lei
- grid.41156.370000 0001 2314 964XDepartment of Orthodontics, Medical School of Nanjing University, Nanjing Stomatological Hospital, Nanjing, China
| | - Houxuan Li
- Department of Periodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, #30 Zhongyang Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
50
|
Guo D, Meng Y, Jiang X, Lu Z. Hexokinases in cancer and other pathologies. CELL INSIGHT 2023; 2:100077. [PMID: 37192912 PMCID: PMC10120283 DOI: 10.1016/j.cellin.2023.100077] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/28/2022] [Accepted: 01/02/2023] [Indexed: 05/18/2023]
Abstract
Glucose metabolism is indispensable for cell growth and survival. Hexokinases play pivotal roles in glucose metabolism through canonical functions of hexokinases as well as in immune response, cell stemness, autophagy, and other cellular activities through noncanonical functions. The aberrant regulation of hexokinases contributes to the development and progression of pathologies, including cancer and immune diseases.
Collapse
Affiliation(s)
- Dong Guo
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ying Meng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoming Jiang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|