1
|
Jain A, Das R, Giri M, Mane P, Shard A. Carbohydrate kinase inhibition: a promising strategy in cancer treatment. Drug Discov Today 2025:104308. [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] [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
|
2
|
Ricci L, Cardaci S. A novel glucose sensor fuelling cancer growth. Cell Death Differ 2024; 31:1579-1581. [PMID: 39379701 PMCID: PMC11618296 DOI: 10.1038/s41418-024-01400-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/10/2024] Open
Affiliation(s)
- Luisa Ricci
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Simone Cardaci
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.
| |
Collapse
|
3
|
Xing Y, Ge J, Wang Y, Zhou X, Yuan Z, Lv M, Zhao M, Liu L, Gong D, Geng T, Xie K. Mitochondrial HKDC1 suppresses oxidative stress and apoptosis by regulating mitochondrial function in goose fatty liver. Int J Biol Macromol 2024; 282:137222. [PMID: 39491705 DOI: 10.1016/j.ijbiomac.2024.137222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Different from human non-alcoholic fatty liver disease (NAFLD), goose fatty liver is physiological with no inflammation. Consistently, mitochondrial dysfunction, oxidative stress and apoptosis are rarely seen in goose fatty liver. Hexokinase domain-containing protein 1 (HKDC1) is involved in maintaining systemic glucose homeostasis, and its absence causes mitochondrial dysfunction. Here, we demonstrated that mitochondrial outer membrane-bound HKDC1 (mHKDC1) had an expression pattern different from that of whole-cell HKDC1 (wHKDC1). Data indicated that the protein level of whole-cell HKDC1 (wHKDC1) was increased but mHKDC1 was decreased in mouse fatty liver. Interestingly, both the protein levels of wHKDC1 and mHKDC1 were significantly increased in goose fatty liver. Treatment of goose or mouse hepatocytes with fatty liver-related factors could influence the expression of wHKDC1 and mHKDC1, but the influence on wHKDC1 was not identical to mHKDC1. HKDC1 overexpression in goose hepatocytes increased wHKDC1 and mHKDC1 expression, mitochondrial membrane potential (MMP), mitochondrial respiratory chain activity, and suppressed reactive oxygen species (ROS) generation, apoptosis and cytokine-cytokine receptor signaling pathway. In addition, mutations in mitochondrial signal peptide or activation domain of HKDC1 altered MMP or ROS levels. In conclusion, HKDC1, particularly mHKDC1, may protect goose fatty liver by regulating mitochondrial function, ROS generation, apoptosis, and inflammation-related pathways.
Collapse
Affiliation(s)
- Ya Xing
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jing Ge
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yuqing Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Xiaoyi Zhou
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zijin Yuan
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Mengqing Lv
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Minmeng Zhao
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Long Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Daoqing Gong
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Tuoyu Geng
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China.
| | - Kaizhou Xie
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
4
|
Li T, Zhu F, Dai L, Hogstrand C, Li B, Yue X, Wang J, Yu L, Li D. Effects of 2-ethylhexyl diphenyl phosphate (EHDPP) on glycolipid metabolism in male adult zebrafish revealed by targeted lipidomic analyses. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174248. [PMID: 38936724 DOI: 10.1016/j.scitotenv.2024.174248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
The present study aims to evaluate the effects of 2-ethylhexyldiphenyl phosphate (EHDPP) on glycolipid metabolism in vivo. Adult male zebrafish were exposed to various concentrations (0, 1, 10, 100 and 250 μg/L) of EHDPP for 28 days, and changes in lipid and glucose levels were measured. Results indicated significant liver damages in the 100 and 250 μg/L EHDPP groups, which both exhibited significant decreases in hepatic somatic index (HSI), elevated activities of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in serum and liver, as well as hepatocyte vacuolation and nuclear pyknosis. Exposure to 100 and 250 μg/L EHDPP led to significant reductions in serum and liver cholesterol (TC), triglycerides (TGs), and lipid droplet deposition, indicating a significant inhibition of EHDPP on hepatic lipid accumulation. Lipidomic analyses manifested that 250 μg/L EHDPP reduced the levels of 103 lipid metabolites which belong to glycerides (TGs, diglycerides, and monoglycerides), fatty acyles (fatty acids), sterol lipids (cholesterol, bile acids), sphingolipids, and glycerophospholipids, and downregulated genes involved in de novo synthesis of fatty acids (fas, acc, srebp1, and dagt2), while upregulated genes involved in fatty acid β-oxidation (pparα and cpt1). KEGG analyses revealed that EHDPP significantly disrupted glycerolipid metabolism, steroid biosynthesis and fatty acid biosynthesis pathways. Collectively, the results showed that EHDPP induced lipid reduction in zebrafish liver, possibly through inhibiting lipid synthesis and disrupting glycerolipid metabolism. Our findings provide a theoretical basis for evaluating the ecological hazards and health effects of EHDPP on glycolipid metabolism.
Collapse
Affiliation(s)
- Tao Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Fengyue Zhu
- National Agricultural Science Observing and Experimental Station of Chongqing, China; Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430073, China
| | - Lili Dai
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430073, China
| | - Christer Hogstrand
- King's College London, Franklin-Wilkins Building, 150 Stamford St., London SE1 9NH, United Kingdom
| | - Boqun Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Xikai Yue
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Jianghua Wang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Liqin Yu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan, China; Engineering Research Center of Green development for Conventional Aquatic, Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China.
| | - Dapeng Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan, China; Engineering Research Center of Green development for Conventional Aquatic, Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| |
Collapse
|
5
|
Lyu SI, Simon AG, Jung JO, Fretter C, SchrÖder W, Bruns CJ, Schmidt T, Quaas A, Knipper K. Hexokinase 2 as an independent risk factor for worse patient survival in esophageal adenocarcinoma and as a potential therapeutic target protein: A retrospective, single‑center cohort study. Oncol Lett 2024; 28:495. [PMID: 39211305 PMCID: PMC11358717 DOI: 10.3892/ol.2024.14628] [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: 03/15/2024] [Accepted: 06/26/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer cells exhibit a distinct metabolic profile that features an upregulation of less efficient glycolysis accompanied by lactate production for energy generation, in contract to the characteristic metabolism of normal cells. Consequently, cancer research has focused on the enzymes that participate in these cancer metabolic pathways. Among them, hexokinase 2 (HK2) has an important position as the initial enzyme in the glycolytic pathway. Increased expression levels of HK2 have been correlated with an increased risk of poor patient outcomes and advanced tumor stages in a number of malignant tumors, such as gastric carcinoma. The present study aimed to investigate the specific role of HK2 in patients diagnosed with esophageal adenocarcinoma. A total of 643 patients with esophageal adenocarcinoma were included. Immunohistochemical staining and HK2 mRNA in situ probes were used to investigate the association of HK2 expression levels with clinical and molecular tumor characteristics. Patients who exhibited high HK2 expression levels demonstrated significantly reduced overall survival (OS) times compared with patients who exhibited low HK2 expression levels (29.6 vs. 39.9 months, respectively; P=0.027). Furthermore, high HK2 expression levels were demonstrated to be an independent risk factor for reduced patient survival (hazard ratio, 1.65; 95% CI, 1.09-2.50; P=0.018). Significantly reduced patient survival was also demonstrated in the subgroups of male patients, patients with primarily resected tumors, patients with HER2-negative tumors and patients with tumors exhibiting Y chromosome loss. Elevated expression of HK2 was identified as a risk factor for unfavorable patient survival in esophageal adenocarcinoma. This revelation suggests the potential for future diagnostic and therapeutic avenues tailored to this specific patient subset. Identifying patients with high HK2 expression may pinpoint a higher-risk cohort, paving the way for comprehensive prospective studies that could advocate for intensified monitoring and more aggressive therapeutic regimens. Furthermore, the targeted inhibition of HK2 could hold promise as a strategy to potentially enhance patient outcomes.
Collapse
Affiliation(s)
- Su Ir Lyu
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, D-50937 Cologne, Germany
| | - Adrian Georg Simon
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, D-50937 Cologne, Germany
| | - Jin-On Jung
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, University of Cologne, D-50937 Cologne, Germany
| | - Caroline Fretter
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, D-50937 Cologne, Germany
| | - Wolfgang SchrÖder
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, University of Cologne, D-50937 Cologne, Germany
| | - Christiane J. Bruns
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, University of Cologne, D-50937 Cologne, Germany
| | - Thomas Schmidt
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, University of Cologne, D-50937 Cologne, Germany
| | - Alexander Quaas
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, D-50937 Cologne, Germany
| | - Karl Knipper
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, University of Cologne, D-50937 Cologne, Germany
| |
Collapse
|
6
|
Fan L, Tian C, Yang W, Liu X, Dhungana Y, Yang W, Tan H, Glazer ES, Yu J, Peng J, Ma L, Ni M, Zhu L. HKDC1 promotes liver cancer stemness under hypoxia through stabilizing β-catenin. Hepatology 2024:01515467-990000000-01019. [PMID: 39250463 DOI: 10.1097/hep.0000000000001085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 08/19/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND AND AIMS Hexokinases (HKs), a group of enzymes catalyzing the first step of glycolysis, have been shown to play important roles in liver metabolism and tumorigenesis. Our recent studies identified hexokinase domain containing 1 (HKDC1) as a top candidate associated with liver cancer metastasis. We aimed to compare its cell-type specificity with other HKs upregulated in liver cancer and investigate the molecular mechanisms underlying its involvement in liver cancer metastasis. APPROACH AND RESULTS We found that, compared to HK1 and HK2, the other 2 commonly upregulated HKs in liver cancer, HKDC1 was most strongly associated with the metastasis potential of tumors and organoids derived from 2 liver cancer mouse models we previously established. RNA in situ hybridization and single-cell RNA-seq analysis revealed that HKDC1 was specifically upregulated in malignant cells in HCC and cholangiocarcinoma patient tumors, whereas HK1 and HK2 were widespread across various tumor microenvironment lineages. An unbiased metabolomic profiling demonstrated that HKDC1 overexpression in HCC cells led to metabolic alterations distinct from those from HK1 and HK2 overexpression, with HKDC1 particularly impacting the tricarboxylic acid cycle. HKDC1 was prometastatic in HCC orthotopic and tail vein injection mouse models. Molecularly, HKDC1 was induced by hypoxia and bound to glycogen synthase kinase 3β to stabilize β-catenin, leading to enhanced stemness of HCC cells. CONCLUSIONS Overall, our findings underscore HKDC1 as a prometastatic HK specifically expressed in the malignant compartment of primary liver tumors, thereby providing a mechanistic basis for targeting this enzyme in advanced liver cancer.
Collapse
Affiliation(s)
- Li Fan
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Cheng Tian
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Wentao Yang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Xiaoli Liu
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Yogesh Dhungana
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Wenjian Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Haiyan Tan
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Evan S Glazer
- Departments of Surgery and Cancer Center, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Lichun Ma
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Min Ni
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Liqin Zhu
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
7
|
Pilic J, Gottschalk B, Bourgeois B, Habisch H, Koshenov Z, Oflaz FE, Erdogan YC, Miri SM, Yiğit EN, Aydın MŞ, Öztürk G, Eroglu E, Shoshan-Barmatz V, Madl T, Graier WF, Malli R. Hexokinase 1 forms rings that regulate mitochondrial fission during energy stress. Mol Cell 2024; 84:2732-2746.e5. [PMID: 38981483 DOI: 10.1016/j.molcel.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 04/30/2024] [Accepted: 06/11/2024] [Indexed: 07/11/2024]
Abstract
Metabolic enzymes can adapt during energy stress, but the consequences of these adaptations remain understudied. Here, we discovered that hexokinase 1 (HK1), a key glycolytic enzyme, forms rings around mitochondria during energy stress. These HK1-rings constrict mitochondria at contact sites with the endoplasmic reticulum (ER) and mitochondrial dynamics protein (MiD51). HK1-rings prevent mitochondrial fission by displacing the dynamin-related protein 1 (Drp1) from mitochondrial fission factor (Mff) and mitochondrial fission 1 protein (Fis1). The disassembly of HK1-rings during energy restoration correlated with mitochondrial fission. Mechanistically, we identified that the lack of ATP and glucose-6-phosphate (G6P) promotes the formation of HK1-rings. Mutations that affect the formation of HK1-rings showed that HK1-rings rewire cellular metabolism toward increased TCA cycle activity. Our findings highlight that HK1 is an energy stress sensor that regulates the shape, connectivity, and metabolic activity of mitochondria. Thus, the formation of HK1-rings may affect mitochondrial function in energy-stress-related pathologies.
Collapse
Affiliation(s)
- Johannes Pilic
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, 8010 Graz, Austria
| | - Benjamin Gottschalk
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, 8010 Graz, Austria
| | - Benjamin Bourgeois
- BioTechMed Graz, Mozartgasse 12/2, 8010 Graz, Austria; Otto Loewi Research Center, Medical Chemistry, Medical University of Graz, 8010 Graz, Austria
| | - Hansjörg Habisch
- Otto Loewi Research Center, Medical Chemistry, Medical University of Graz, 8010 Graz, Austria
| | - Zhanat Koshenov
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, 8010 Graz, Austria
| | - Furkan E Oflaz
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, 8010 Graz, Austria
| | - Yusuf C Erdogan
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, 8010 Graz, Austria
| | - Seyed M Miri
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810 Istanbul, Türkiye; Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, 34956 Istanbul, Türkiye
| | - Esra N Yiğit
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810 Istanbul, Türkiye; Department of Physiology, International School of Medicine, Istanbul Medipol University, 34810 Istanbul, Türkiye
| | - Mehmet Ş Aydın
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810 Istanbul, Türkiye
| | - Gürkan Öztürk
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810 Istanbul, Türkiye
| | - Emrah Eroglu
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810 Istanbul, Türkiye; Department of Physiology, International School of Medicine, Istanbul Medipol University, 34810 Istanbul, Türkiye
| | - Varda Shoshan-Barmatz
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | - Tobias Madl
- BioTechMed Graz, Mozartgasse 12/2, 8010 Graz, Austria; Otto Loewi Research Center, Medical Chemistry, Medical University of Graz, 8010 Graz, Austria
| | - Wolfgang F Graier
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, 8010 Graz, Austria; BioTechMed Graz, Mozartgasse 12/2, 8010 Graz, Austria
| | - Roland Malli
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, 8010 Graz, Austria; BioTechMed Graz, Mozartgasse 12/2, 8010 Graz, Austria; Center for Medical Research, CF Bioimaging, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria.
| |
Collapse
|
8
|
Guo N, Luo Q, Zheng Q, Yang S, Zhang S. Current status and progress of research on the ADP-dependent glucokinase gene. Front Oncol 2024; 14:1358904. [PMID: 38590647 PMCID: PMC10999526 DOI: 10.3389/fonc.2024.1358904] [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: 12/21/2023] [Accepted: 02/16/2024] [Indexed: 04/10/2024] Open
Abstract
ADP-dependent glucokinase (ADPGK) produces glucose-6-phosphate with adenosine diphosphate (ADP) as the phosphate group donor, in contrast to ATP-dependent hexokinases (HKs). Originally found in archaea, ADPGK is involved in glycolysis. However, its biological function in most eukaryotic organisms is still unclear, and the molecular mechanism of action requires further investigation. This paper provides a concise overview of ADPGK's origin, biological function and clinical application. It aims to furnish scientific information for the diagnosis and treatment of human metabolic diseases, neurological disorders, and malignant tumours, and to suggest new strategies for the development of targeted drugs.
Collapse
Affiliation(s)
- Ningjing Guo
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Qiong Luo
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Qixian Zheng
- Department of Respiratory Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Sheng Yang
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Suyun Zhang
- Department of Internal Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
9
|
Zhang Y, Wang M, Ye L, Shen S, Zhang Y, Qian X, Zhang T, Yuan M, Ye Z, Cai J, Meng X, Qiu S, Liu S, Liu R, Jia W, Yang X, Zhang H, Zhong X, Gao P. HKDC1 promotes tumor immune evasion in hepatocellular carcinoma by coupling cytoskeleton to STAT1 activation and PD-L1 expression. Nat Commun 2024; 15:1314. [PMID: 38351096 PMCID: PMC10864387 DOI: 10.1038/s41467-024-45712-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/31/2024] [Indexed: 02/16/2024] Open
Abstract
Immune checkpoint blockade (ICB) has shown considerable promise for treating various malignancies, but only a subset of cancer patients benefit from immune checkpoint inhibitor therapy because of immune evasion and immune-related adverse events (irAEs). The mechanisms underlying how tumor cells regulate immune cell response remain largely unknown. Here we show that hexokinase domain component 1 (HKDC1) promotes tumor immune evasion in a CD8+ T cell-dependent manner by activating STAT1/PD-L1 in tumor cells. Mechanistically, HKDC1 binds to and presents cytosolic STAT1 to IFNGR1 on the plasma membrane following IFNγ-stimulation by associating with cytoskeleton protein ACTA2, resulting in STAT1 phosphorylation and nuclear translocation. HKDC1 inhibition in combination with anti-PD-1/PD-L1 enhances in vivo T cell antitumor response in liver cancer models in male mice. Clinical sample analysis indicates a correlation among HKDC1 expression, STAT1 phosphorylation, and survival in patients with hepatocellular carcinoma treated with atezolizumab (anti-PD-L1). These findings reveal a role for HKDC1 in regulating immune evasion by coupling cytoskeleton with STAT1 activation, providing a potential combination strategy to enhance antitumor immune responses.
Collapse
Affiliation(s)
- Yi Zhang
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Mingjie Wang
- School of Medicine, South China University of Technology, Guangzhou, China
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ling Ye
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Shengqi Shen
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yuxi Zhang
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiaoyu Qian
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Tong Zhang
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Mengqiu Yuan
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Zijian Ye
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Jin Cai
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiang Meng
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Shiqiao Qiu
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Shengzhi Liu
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Rui Liu
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Weidong Jia
- Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, the First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Xianzhu Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China.
| | - Huafeng Zhang
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Xiuying Zhong
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Ping Gao
- School of Medicine, South China University of Technology, Guangzhou, China.
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
10
|
Cui M, Yamano K, Yamamoto K, Yamamoto-Imoto H, Minami S, Yamamoto T, Matsui S, Kaminishi T, Shima T, Ogura M, Tsuchiya M, Nishino K, Layden BT, Kato H, Ogawa H, Oki S, Okada Y, Isaka Y, Kosako H, Matsuda N, Yoshimori T, Nakamura S. HKDC1, a target of TFEB, is essential to maintain both mitochondrial and lysosomal homeostasis, preventing cellular senescence. Proc Natl Acad Sci U S A 2024; 121:e2306454120. [PMID: 38170752 PMCID: PMC10786298 DOI: 10.1073/pnas.2306454120] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2024] Open
Abstract
Mitochondrial and lysosomal functions are intimately linked and are critical for cellular homeostasis, as evidenced by the fact that cellular senescence, aging, and multiple prominent diseases are associated with concomitant dysfunction of both organelles. However, it is not well understood how the two important organelles are regulated. Transcription factor EB (TFEB) is the master regulator of lysosomal function and is also implicated in regulating mitochondrial function; however, the mechanism underlying the maintenance of both organelles remains to be fully elucidated. Here, by comprehensive transcriptome analysis and subsequent chromatin immunoprecipitation-qPCR, we identified hexokinase domain containing 1 (HKDC1), which is known to function in the glycolysis pathway as a direct TFEB target. Moreover, HKDC1 was upregulated in both mitochondrial and lysosomal stress in a TFEB-dependent manner, and its function was critical for the maintenance of both organelles under stress conditions. Mechanistically, the TFEB-HKDC1 axis was essential for PINK1 (PTEN-induced kinase 1)/Parkin-dependent mitophagy via its initial step, PINK1 stabilization. In addition, the functions of HKDC1 and voltage-dependent anion channels, with which HKDC1 interacts, were essential for the clearance of damaged lysosomes and maintaining mitochondria-lysosome contact. Interestingly, HKDC1 regulated mitophagy and lysosomal repair independently of its prospective function in glycolysis. Furthermore, loss function of HKDC1 accelerated DNA damage-induced cellular senescence with the accumulation of hyperfused mitochondria and damaged lysosomes. Our results show that HKDC1, a factor downstream of TFEB, maintains both mitochondrial and lysosomal homeostasis, which is critical to prevent cellular senescence.
Collapse
Affiliation(s)
- Mengying Cui
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
| | - Koji Yamano
- Ubiquitin Project, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo156-8506, Japan
- Department of Biomolecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo113-8510, Japan
| | - Kenichi Yamamoto
- Department of Statistical Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
| | - Hitomi Yamamoto-Imoto
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
| | - Satoshi Minami
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
- Department of Nephrology, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
| | - Takeshi Yamamoto
- Department of Nephrology, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
| | - Sho Matsui
- Department of Nephrology, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
| | - Tatsuya Kaminishi
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka565-0871, Japan
| | - Takayuki Shima
- Department of Biochemistry, Nara Medical University, Kashihara, Nara634-8521, Japan
| | - Monami Ogura
- Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka565-0871, Japan
| | - Megumi Tsuchiya
- Laboratory of Nuclear Dynamics Group, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka565-0871, Japan
| | - Kohei Nishino
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima770-8503, Japan
| | - Brian T. Layden
- Division of Endocrinology, Diabetes, and Metabolism, University of Illinois Chicago, Chicago, IL60612
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL60612
| | - Hisakazu Kato
- Department of Medical Biochemistry, Graduate School of Medicine/Frontier Bioscience, Osaka University, Suita, Osaka565-0871, Japan
| | - Hidesato Ogawa
- Laboratory of Nuclear Dynamics Group, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka565-0871, Japan
| | - Shinya Oki
- Department of Drug Discovery Medicine, Graduate School of Medicine, Kyoto University, Kyoto606-8501, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka565-0871, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center, World Premier International Research Center (WPI-IFReC), Osaka University, Suita, Osaka565-0871, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima770-8503, Japan
| | - Noriyuki Matsuda
- Ubiquitin Project, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo156-8506, Japan
- Department of Biomolecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo113-8510, Japan
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka565-0871, Japan
- Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka565-0871, Japan
| | - Shuhei Nakamura
- Department of Biochemistry, Nara Medical University, Kashihara, Nara634-8521, Japan
| |
Collapse
|
11
|
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: 60] [Impact Index Per Article: 30.0] [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
|
12
|
Pusec CM, Ilievski V, De Jesus A, Farooq Z, Zapater JL, Sweis N, Ismail H, Khan MW, Ardehali H, Cordoba-Chacon J, Layden BT. Liver-specific overexpression of HKDC1 increases hepatocyte size and proliferative capacity. Sci Rep 2023; 13:8034. [PMID: 37198225 PMCID: PMC10192376 DOI: 10.1038/s41598-023-33924-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 04/20/2023] [Indexed: 05/19/2023] Open
Abstract
A primary role of the liver is to regulate whole body glucose homeostasis. Glucokinase (GCK) is the main hexokinase (HK) expressed in hepatocytes and functions to phosphorylate the glucose that enters via GLUT transporters to become glucose-6-phosphate (G6P), which subsequently commits glucose to enter downstream anabolic and catabolic pathways. In the recent years, hexokinase domain-containing-1 (HKDC1), a novel 5th HK, has been characterized by our group and others. Its expression profile varies but has been identified to have low basal expression in normal liver but increases during states of stress including pregnancy, nonalcoholic fatty liver disease (NAFLD), and liver cancer. Here, we have developed a stable overexpression model of hepatic HKDC1 in mice to examine its effect on metabolic regulation. We found that HKDC1 overexpression, over time, causes impaired glucose homeostasis in male mice and shifts glucose metabolism towards anabolic pathways with an increase in nucleotide synthesis. Furthermore, we observed these mice to have larger liver sizes due to greater hepatocyte proliferative potential and cell size, which in part, is mediated via yes-associated protein (YAP) signaling.
Collapse
Affiliation(s)
- Carolina M Pusec
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Vladimir Ilievski
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Adam De Jesus
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zeenat Farooq
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Joseph L Zapater
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Nadia Sweis
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Hagar Ismail
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Md Wasim Khan
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Hossein Ardehali
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jose Cordoba-Chacon
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Brian T Layden
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
- Jesse Brown VA Medical Center, Chicago, IL, USA.
| |
Collapse
|
13
|
Farooq Z, Ismail H, Bhat SA, Layden BT, Khan MW. Aiding Cancer's "Sweet Tooth": Role of Hexokinases in Metabolic Reprogramming. Life (Basel) 2023; 13:946. [PMID: 37109475 PMCID: PMC10141071 DOI: 10.3390/life13040946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/24/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
Hexokinases (HKs) convert hexose sugars to hexose-6-phosphate, thus trapping them inside cells to meet the synthetic and energetic demands. HKs participate in various standard and altered physiological processes, including cancer, primarily through the reprogramming of cellular metabolism. Four canonical HKs have been identified with different expression patterns across tissues. HKs 1-3 play a role in glucose utilization, whereas HK 4 (glucokinase, GCK) also acts as a glucose sensor. Recently, a novel fifth HK, hexokinase domain containing 1 (HKDC1), has been identified, which plays a role in whole-body glucose utilization and insulin sensitivity. Beyond the metabolic functions, HKDC1 is differentially expressed in many forms of human cancer. This review focuses on the role of HKs, particularly HKDC1, in metabolic reprogramming and cancer progression.
Collapse
Affiliation(s)
- Zeenat Farooq
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Hagar Ismail
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sheraz Ahmad Bhat
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Brian T. Layden
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| | - Md. Wasim Khan
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
14
|
Wang MQ, Chen YR, Xu HW, Zhan JR, Suo DQ, Wang JJ, Ma YZ, Guan XY, Li Y, Zhu SL. HKDC1 upregulation promotes glycolysis and disease progression, and confers chemoresistance onto gastric cancer. Cancer Sci 2022; 114:1365-1377. [PMID: 36519789 PMCID: PMC10067396 DOI: 10.1111/cas.15692] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
There is increasing evidence that hexokinase is involved in cell proliferation and migration. However, the function of the hexokinase domain containing protein-1 (HKDC1) in gastric cancer (GC) remains unclear. Immunohistochemistry analysis and big data mining were used to evaluate the correlation between HKDC1 expression and clinical features in GC. In addition, the biological function and molecular mechanism of HKDC1 in GC were studied by in vitro and in vivo assays. Our study indicated that HKDC1 expression was upregulated in GC tissues compared with adjacent nontumor tissues. High expression of HKDC1 was associated with worse prognosis. Functional experiments demonstrated that HKDC1 upregulation promoted glycolysis, cell proliferation, and tumorigenesis. In addition, HKDC1 could enhance GC invasion and metastasis by inducing epithelial-mesenchymal transition (EMT). Abrogation of HKDC1 could effectively attenuate its oncogenic and metastatic function. Moreover, HKDC1 promoted GC proliferation and migration in vivo. HKDC1 overexpression conferred chemoresistance to cisplatin, oxaliplatin, and 5-fluorouracil (5-Fu) onto GC cells. Furthermore, nuclear factor kappa-B (NF-κB) inhibitor PS-341 could attenuate tumorigenesis, metastasis, and drug resistance ability induced by HKDC1 overexpression in GC cells. Our results highlight a critical role of HKDC1 in promoting glycolysis, tumorigenesis, and EMT of GC cells via activating the NF-κB pathway. In addition, HKDC1-mediated drug resistance was associated with DNA damage repair, which further activated NF-κB signaling. HKDC1 upregulation may be used as a potential indicator for choosing an effective chemotherapy regimen for GC patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Mei-Qian Wang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yi-Ru Chen
- Department of Gastroenterology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Hui-Wen Xu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jia-Rong Zhan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Da-Qin Suo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ji-Jin Wang
- Department of radiotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuan-Zhen Ma
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin-Yuan Guan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Departments of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Yan Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Sen-Lin Zhu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
15
|
Zhang L, Wang Y, Jia H, Liu X, Zhang R, Guan J. Transcriptome and metabolome analyses reveal the regulatory effects of compound probiotics on cecal metabolism in heat-stressed broilers. Poult Sci 2022; 102:102323. [PMID: 36436366 PMCID: PMC9706624 DOI: 10.1016/j.psj.2022.102323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/30/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
The effect of compound probiotics on the caecum of broilers under heat stress was assessed in this study. A total of 400 twenty-eight-day-old AA male broilers were randomly divided into 4 treatment groups, where each group had 5 replicates of 20 broilers. The 4 treatment groups were a heat stress control group (broilers receiving a normal diet) and groups HP I, HP II, and HP Ⅲ, consisting of broilers receiving 1, 5, and 10 g of compound probiotics added to each kilogram of feed, respectively. Compound probiotics (L. casei, L. acidophilus, and B. lactis at a ratio of 1:1:2) were used to formulate a compound probiotic powder, with 1 × 1010 CFU/g of effective viable bacteria. Heat stress treatment was performed at 32 ± 1°C from 9:00 to 17:00 every day from 28 d to 42 d. In d 28 to 42, compared with the HC group, the ADG of broilers in the HP II and III groups was significantly increased (P < 0.05); the ADFI difference between groups was not significant (P > 0.05); the FCR of HP II and III broilers was significantly decreased (P < 0.05); and the FCR of the HP I group increased, but the difference was not significant (P > 0.05). Transcriptome results demonstrate that 665 differential genes were screened (DEGs; upregulated: 366, downregulated: 299). The DEGs were enriched in the B cell receptor signaling pathway, the intestinal immune network for IgA synthesis, the Fc epsilon RI signaling pathway, and other signaling pathways, according to KEGG enrichment analysis. Metabolome analysis identified 92 differential metabolites (DAMs; upregulated: 48, downregulated: 44). KEGG enrichment analysis indicated significant enrichment of Pantothenate and CoA biosynthesis and beta-Alanine metabolism. The combined transcriptome and metabolome analysis revealed that the DAMs and DEGs were mostly involved in beta-alanine metabolism, arginine biosynthesis, amino sugar and nucleotide sugar, and alanine, aspartate, and glutamate metabolism. The results of this study suggest that the addition of compound probiotics has a positive effect on intestinal metabolites, improving the growth performance and contributing to the overall health of broilers under heat stress.
Collapse
|
16
|
Zhong W, Kollipara A, Liu Y, Wang Y, O’Connell CM, Poston TB, Yount K, Wiesenfeld HC, Hillier SL, Li Y, Darville T, Zheng X. Genetic susceptibility loci for Chlamydia trachomatis endometrial infection influence expression of genes involved in T cell function, tryptophan metabolism and epithelial integrity. Front Immunol 2022; 13:1001255. [PMID: 36248887 PMCID: PMC9562917 DOI: 10.3389/fimmu.2022.1001255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives Identify genetic loci of enhanced susceptibility to Chlamydial trachomatis (Ct) upper genital tract infection in women. Methods We performed an integrated analysis of DNA genotypes and blood-derived mRNA profiles from 200 Ct-exposed women to identify expression quantitative trait loci (eQTL) and determine their association with endometrial chlamydial infection using a mediation test. We further evaluated the effect of a lead eQTL on the expression of CD151 by immune cells from women with genotypes associated with low and high whole blood expression of CD151, respectively. Results We identified cis-eQTLs modulating mRNA expression of 81 genes (eGenes) associated with altered risk of ascending infection. In women with endometrial infection, eGenes involved in proinflammatory signaling were upregulated. Downregulated eGenes included genes involved in T cell functions pivotal for chlamydial control. eGenes encoding molecules linked to metabolism of tryptophan, an essential chlamydial nutrient, and formation of epithelial tight junctions were also downregulated in women with endometrial infection. A lead eSNP rs10902226 was identified regulating CD151, a tetrospanin molecule important for immune cell adhesion and migration and T cell proliferation. Further in vitro experiments showed that women with a CC genotype at rs10902226 had reduced rates of endometrial infection with increased CD151 expression in whole blood and T cells when compared to women with a GG genotype. Conclusions We discovered genetic variants associated with altered risk for Ct ascension. A lead eSNP for CD151 is a candidate genetic marker for enhanced CD4 T cell function and reduced susceptibility.
Collapse
Affiliation(s)
- Wujuan Zhong
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Avinash Kollipara
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yutong Liu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yuhan Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Catherine M. O’Connell
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Taylor B. Poston
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kacy Yount
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Harold C. Wiesenfeld
- The University of Pittsburgh School of Medicine and the Magee-Womens Research Institute, Pittsburgh, PA, United States
| | - Sharon L. Hillier
- The University of Pittsburgh School of Medicine and the Magee-Womens Research Institute, Pittsburgh, PA, United States
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Toni Darville
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Xiaojing Zheng
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
17
|
Doke M, McLaughlin JP, Cai JJ, Pendyala G, Kashanchi F, Khan MA, Samikkannu T. HIV-1 Tat and cocaine impact astrocytic energy reservoirs and epigenetic regulation by influencing the LINC01133-hsa-miR-4726-5p-NDUFA9 axis. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 29:243-258. [PMID: 35892093 PMCID: PMC9307901 DOI: 10.1016/j.omtn.2022.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/04/2022] [Indexed: 06/15/2023]
Abstract
Clinical research has proven that HIV-positive (HIV+) individuals with cocaine abuse show behavioral and neurocognitive disorders. Noncoding RNAs (ncRNAs), such as long ncRNAs (lncRNAs) and microRNAs (miRNAs), are known to regulate gene expression in the contexts of HIV infection and drug abuse. However, there are no specific lncRNA or miRNA biomarkers associated with HIV-1 Transactivator of transcription protein (Tat) and cocaine coexposure. In the central nervous system (CNS), astrocytes are the primary regulators of energy metabolism, and impairment of the astrocytic energy supply can trigger neurodegeneration. The aim of this study was to uncover the roles of lncRNAs and miRNAs in the regulation of messenger RNA (mRNA) targets affected by HIV infection and cocaine abuse. Integrative bioinformatics analysis revealed altered expression of 10 lncRNAs, 10 miRNAs, and 4 mRNA/gene targets in human primary astrocytes treated with cocaine and HIV-1 Tat. We assessed the alterations in the expression of two miRNAs, hsa-miR-2355 and hsa-miR-4726-5p; four lncRNAs, LINC01133, H19, HHIP-AS1, and NOP14-AS1; and four genes, NDUFA9, KYNU, HKDC1, and LIPG. The results revealed interactions in the LINC01133-hsa-miR-4726-5p-NDUFA9 axis that may eventually help us understand cocaine- and HIV-1 Tat-induced astrocyte dysfunction that may ultimately result in neurodegeneration.
Collapse
Affiliation(s)
- Mayur Doke
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, TX 78363, USA
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - James J. Cai
- Veterinary Integrative Biosciences, Texas A&M University, TAMU 4458, College Station, TX 77845, USA
| | - Gurudutt Pendyala
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA
| | - Fatah Kashanchi
- National Center for Biodefense and Infectious Disease, Laboratory of Molecular Virology, George Mason University, Manassas, VA 20110, USA
| | - Mansoor A. Khan
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, TX 78363, USA
| | - Thangavel Samikkannu
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, TX 78363, USA
| |
Collapse
|
18
|
Fuhr L, Basti A, Brás TS, Duarte MF, Relógio A. Antiproliferative Effects of Cynara Cardunculus in Colorectal Cancer Cells Are Modulated by the Circadian Clock. Int J Mol Sci 2022; 23:ijms23169130. [PMID: 36012399 PMCID: PMC9409071 DOI: 10.3390/ijms23169130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/25/2022] Open
Abstract
The circadian clock generates 24 h rhythms in behavioural, cellular and molecular processes. Malfunctions of the clock are associated with enhanced susceptibility to cancer, worse treatment response and poor prognosis. Clock-controlled genes are involved in cellular processes associated with tumour development and progression including metabolism of drugs and the cell cycle. Cynara cardunculus, a plant of the Asteraceae family, has been reported to have antiproliferative effects on breast cancer cells. Here, we used the human colorectal cancer (CRC) cell line HCT116 and its knockout variants for different core-clock genes (BMAL1, PER2, NR1D1), to investigate the treatment effect of C. cardunculus lipophilic leaf extract under different clock scenarios. Our results show a direct effect of C. cardunculus on the circadian phenotype of the cells, as indicated by alterations in the phase, amplitude, and period length of core-clock gene oscillations. Furthermore, our data indicate a role for the circadian clock in sensitivity to C. cardunculus treatment. In particular, the treatment inhibited proliferation and induced cytotoxicity and apoptosis in a clock knockout-specific manner, in CRC cells. These results point to a potential effect of C. cardunculus lipophilic leaf extracts as a modulator of the circadian clock, in addition to its anti-proliferative properties.
Collapse
Affiliation(s)
- Luise Fuhr
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt—Universität zu Berlin, Institute for Theoretical Biology, 10115 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt—Universität zu Berlin, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, 13353 Berlin, Germany
| | - Alireza Basti
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt—Universität zu Berlin, Institute for Theoretical Biology, 10115 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt—Universität zu Berlin, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, 13353 Berlin, Germany
- Institute for Systems Medicine, Faculty of Human Medicine, MSH Medical School Hamburg—University of Applied Sciences and Medical University, 20457 Hamburg, Germany
| | - Teresa Silva Brás
- Alentejo Biotechnology Center for Agriculture and Agro-Food (CEBAL) and Instituto Politécnico de Beja (IPBeja), 7801-908 Beja, Portugal
- MED—Mediterranean Institute for Agriculture, Environment and Development & CHANGE—Global Change and Sustainability Institute, CEBAL, 7801-908 Beja, Portugal
| | - Maria F. Duarte
- Alentejo Biotechnology Center for Agriculture and Agro-Food (CEBAL) and Instituto Politécnico de Beja (IPBeja), 7801-908 Beja, Portugal
- MED—Mediterranean Institute for Agriculture, Environment and Development & CHANGE—Global Change and Sustainability Institute, CEBAL, 7801-908 Beja, Portugal
| | - Angela Relógio
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt—Universität zu Berlin, Institute for Theoretical Biology, 10115 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt—Universität zu Berlin, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, 13353 Berlin, Germany
- Institute for Systems Medicine, Faculty of Human Medicine, MSH Medical School Hamburg—University of Applied Sciences and Medical University, 20457 Hamburg, Germany
- Correspondence:
| |
Collapse
|
19
|
Chen H, Wang SH, Chen C, Yu XY, Zhu JN, Mansell T, Novakovic B, Saffery R, Baker PN, Han TL, Zhang H. A novel role of FoxO3a in the migration and invasion of trophoblast cells: from metabolic remodeling to transcriptional reprogramming. Mol Med 2022; 28:92. [PMID: 35941589 PMCID: PMC9358829 DOI: 10.1186/s10020-022-00522-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Background The forkhead box O3a protein (FoxO3a) has been reported to be involved in the migration and invasion of trophoblast, but its underlying mechanisms unknown. In this study, we aim to explore the transcriptional and metabolic regulations of FoxO3a on the migration and invasion of early placental development.
Methods Lentiviral vectors were used to knock down the expression of FoxO3a of the HTR8/SVneo cells. Western blot, matrigel invasion assay, wound healing assay, seahorse, gas-chromatography-mass spectrometry (GC–MS) based metabolomics, fluxomics, and RNA-seq transcriptomics were performed. Results We found that FoxO3a depletion restrained the migration and invasion of HTR8/SVneo cells. Metabolomics, fluxomics, and seahorse demonstrated that FoxO3a knockdown resulted in a switch from aerobic to anaerobic respiration and increased utilization of aromatic amino acids and long-chain fatty acids from extracellular nutrients. Furthermore, our RNA-seq also demonstrated that the expression of COX-2 and MMP9 decreased after FoxO3a knockdown, and these two genes were closely associated with the migration/invasion progress of trophoblast cells. Conclusions Our results suggested novel biological roles of FoxO3a in early placental development. FoxO3a exerts an essential effect on trophoblast migration and invasion owing to the regulations of COX2, MMP9, aromatic amino acids, energy metabolism, and oxidative stress.
Collapse
Affiliation(s)
- Hao Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, Chongqing, China
| | - Shi-Han Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, Chongqing, China
| | - Chang Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Xin-Yang Yu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China
| | - Jia-Nan Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, Chongqing, China
| | - Toby Mansell
- Molecular Immunity, Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Boris Novakovic
- Molecular Immunity, Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Richard Saffery
- Molecular Immunity, Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Philip N Baker
- Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK
| | - Ting-Li Han
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Hua Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China. .,Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
20
|
Khan MW, Terry AR, Priyadarshini M, Ilievski V, Farooq Z, Guzman G, Cordoba-Chacon J, Ben-Sahra I, Wicksteed B, Layden BT. The hexokinase "HKDC1" interaction with the mitochondria is essential for liver cancer progression. Cell Death Dis 2022; 13:660. [PMID: 35902556 PMCID: PMC9334634 DOI: 10.1038/s41419-022-04999-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/28/2022] [Accepted: 06/07/2022] [Indexed: 01/21/2023]
Abstract
Liver cancer (LC) is the fourth leading cause of death from cancer malignancies. Recently, a putative fifth hexokinase, hexokinase domain containing 1 (HKDC1), was shown to have significant overexpression in LC compared to healthy liver tissue. Using a combination of in vitro and in vivo tools, we examined the role of HKDC1 in LC development and progression. Importantly, HKDC1 ablation stops LC development and progression via its action at the mitochondria by promoting metabolic reprogramming and a shift of glucose flux away from the TCA cycle. HKDC1 ablation leads to mitochondrial dysfunction resulting in less cellular energy, which cannot be compensated by enhanced glucose uptake. Moreover, we show that the interaction of HKDC1 with the mitochondria is essential for its role in LC progression, and without this interaction, mitochondrial dysfunction occurs. As HKDC1 is highly expressed in LC cells, but only to a minimal degree in hepatocytes under normal conditions, targeting HKDC1, specifically its interaction with the mitochondria, may represent a highly selective approach to target cancer cells in LC.
Collapse
Affiliation(s)
- Md. Wasim Khan
- grid.185648.60000 0001 2175 0319Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Alexander R. Terry
- grid.185648.60000 0001 2175 0319Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607 USA
| | - Medha Priyadarshini
- grid.185648.60000 0001 2175 0319Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Vladimir Ilievski
- grid.185648.60000 0001 2175 0319Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Zeenat Farooq
- grid.185648.60000 0001 2175 0319Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Grace Guzman
- grid.412973.a0000 0004 0434 4425Department of Pathology, College of Medicine, Cancer Center, University of Illinois Hospital and Health Science Chicago, Chicago, IL 60612 USA
| | - Jose Cordoba-Chacon
- grid.185648.60000 0001 2175 0319Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Issam Ben-Sahra
- grid.16753.360000 0001 2299 3507Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611 USA
| | - Barton Wicksteed
- grid.185648.60000 0001 2175 0319Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Brian T. Layden
- grid.185648.60000 0001 2175 0319Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA ,grid.280892.90000 0004 0419 4711Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612 USA
| |
Collapse
|
21
|
Zapater JL, Wicksteed B, Layden BT. Enterocyte HKDC1 Modulates Intestinal Glucose Absorption in Male Mice Fed a High-fat Diet. Endocrinology 2022; 163:6569855. [PMID: 35435980 PMCID: PMC9078327 DOI: 10.1210/endocr/bqac050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Indexed: 11/24/2022]
Abstract
Hexokinase domain containing protein-1, or HKDC1, is a widely expressed hexokinase that is genetically associated with elevated 2-hour gestational blood glucose levels during an oral glucose tolerance test, suggesting a role for HKDC1 in postprandial glucose regulation during pregnancy. Our earlier studies utilizing mice containing global HKDC1 knockdown, as well as hepatic HKDC1 overexpression and knockout, indicated that HKDC1 is important for whole-body glucose homeostasis in aging and pregnancy, through modulation of glucose tolerance, peripheral tissue glucose utilization, and hepatic energy storage. However, our knowledge of the precise role(s) of HKDC1 in regulating postprandial glucose homeostasis under normal and diabetic conditions is lacking. Since the intestine is the main entry portal for dietary glucose, here we have developed an intestine-specific HKDC1 knockout mouse model, HKDC1Int-/-, to determine the in vivo role of intestinal HKDC1 in regulating glucose homeostasis. While no overt glycemic phenotype was observed, aged HKDC1Int-/- mice fed a high-fat diet exhibited an increased glucose excursion following an oral glucose load compared with mice expressing intestinal HKDC1. This finding resulted from glucose entry via the intestinal epithelium and is not due to differences in insulin levels, enterocyte glucose utilization, or reduction in peripheral skeletal muscle glucose uptake. Assessment of intestinal glucose transporters in high-fat diet-fed HKDC1Int-/- mice suggested increased apical GLUT2 expression in the fasting state. Taken together, our results indicate that intestinal HKDC1 contributes to the modulation of postprandial dietary glucose transport across the intestinal epithelium under conditions of enhanced metabolic stress, such as high-fat diet.
Collapse
Affiliation(s)
- Joseph L Zapater
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, Medical Research Service, Chicago, IL 60612, USA
| | - Barton Wicksteed
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Brian T Layden
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, Medical Research Service, Chicago, IL 60612, USA
- Correspondence: Brian T. Layden, MD, PhD, 835 South Wolcott Avenue, Suite 625E (M/C 640), Chicago, IL, 60612, USA.
| |
Collapse
|
22
|
Hou S, Lei S, Peng H, Weng L, Lv S, Li M, Zhao D. Down-regulating HK2 inhibits proliferation of endometrial stromal cells through a noncanonical pathway involving phosphorylation of STAT1 in endometriosis. Biol Reprod 2022; 107:488-499. [PMID: 35470850 DOI: 10.1093/biolre/ioac081] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/26/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Endometriosis is a benign gynecologic disease that causes chronic pelvic pain, dysmenorrhea and infertility and shares several characteristics with malignant tumors, afflicting women of reproductive age. Hexokinase 2 (HK2) plays an essential role as the first rate-limiting enzyme in the metabolic glycolysis pathway, and its abnormal elevation in tumors is associated with tumor genesis and metastasis. However, the expression and role of HK2 in endometriosis remain unclear. METHODS We sequenced the primary endometrial stromal cells from patients with endometrioma and utilized immunohistochemistry, quantitative real-time PCR and western blot to determine the expression of HK2. Then wound healing assays, cell invasion assays, cell proliferation assays were performed to explore the functions of HK2 in endometrial stromal cells. Furthermore, mice models of endometriosis were used to observe the effects of HK2 inhibitors in vivo. Lastly, glycolysis metabolism detection and transcriptome sequencing were carried out in HK2-knockdown endometrial stromal cells to analyze the mechanism of HK2 affecting cell function. RESULTS Endometrial stromal cells of endometrioma displayed active glycolysis metabolism and elevated expression of HK2. Downregulating HK2 reduced the migration, invasion and proliferation capacity of endometrial stromal cells. Knockdown of HK2 induced upregulation of signal transducer and activator of transcription 1 (STAT1) and their phosphorylation to attenuate the proliferation of endometrial stromal cells. CONCLUSIONS HK2 is associated with the migration, invasion and proliferation of endometrial stromal cells, which might provide new insights into the pathogenesis and treatment of endometriosis.
Collapse
Affiliation(s)
- Shuhui Hou
- Department of Obstetrics and Gynecology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China.,Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, People's Republic of China
| | - Shating Lei
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, People's Republic of China
| | - Haiyan Peng
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, People's Republic of China
| | - Lichun Weng
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, People's Republic of China
| | - Siji Lv
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, People's Republic of China
| | - Mingqing Li
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Laboratory for Reproductive Immunology, Shanghai Medical School, Fudan University, Shanghai 200080, People's Republic of China
| | - Dong Zhao
- Department of Obstetrics and Gynecology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
| |
Collapse
|
23
|
Sofer S, Lamkiewicz K, Armoza Eilat S, Partouche S, Marz M, Moskovits N, Stemmer SM, Shlomai A, Sklan EH. A genome-wide CRISPR activation screen reveals Hexokinase 1 as a critical factor in promoting resistance to multi-kinase inhibitors in hepatocellular carcinoma cells. FASEB J 2022; 36:e22191. [PMID: 35147243 DOI: 10.1096/fj.202101507rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/27/2021] [Accepted: 01/20/2022] [Indexed: 01/05/2023]
Abstract
Hepatocellular carcinoma (HCC) is often diagnosed at an advanced stage and is, therefore, treated with systemic drugs, such as tyrosine-kinase inhibitors (TKIs). These drugs, however, offer only modest survival benefits due to the rapid development of drug resistance. To identify genes implicated in TKI resistance, a cluster of regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 activation screen was performed in hepatoma cells treated with regorafenib, a TKI used as second-line therapy for advanced HCC. The screen results show that Hexokinase 1 (HK1), catalyzing the first step in glucose metabolism, is a top candidate for conferring TKI resistance. Compatible with this, HK1 was upregulated in regorafenib-resistant cells. Using several experimental approaches, both in vitro and in vivo, we show that TKI resistance correlates with HK1 expression. Furthermore, an HK inhibitor resensitized resistant cells to TKI treatment. Together, our data indicate that HK1 may function as a critical factor modulating TKI resistance in hepatoma cells and, therefore, may serve as a biomarker for treatment success.
Collapse
Affiliation(s)
- Summer Sofer
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Kevin Lamkiewicz
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University, Jena, Germany.,European Virus Bioinformatics Center, Jena, Germany
| | - Shir Armoza Eilat
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shirly Partouche
- Felsenstein Medical Research Center, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel
| | - Manja Marz
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University, Jena, Germany.,European Virus Bioinformatics Center, Jena, Germany.,Leibniz Institute for Age Research-Fritz Lipmann Institute, Jena, Germany
| | - Neta Moskovits
- PDX Laboratory, Felsenstein Medical Research Center and the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Salomon M Stemmer
- PDX Laboratory, Felsenstein Medical Research Center and the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Institute of Oncology, Davidoff Center, Rabin Medical Center, Petah-Tikva, Israel
| | - Amir Shlomai
- Felsenstein Medical Research Center and the Department of Medicine D, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Ella H Sklan
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
24
|
Dong L, Lu D, Chen R, Lin Y, Zhu H, Zhang Z, Cai S, Cui P, Song G, Rao D, Yi X, Wu Y, Song N, Liu F, Zou Y, Zhang S, Zhang X, Wang X, Qiu S, Zhou J, Wang S, Zhang X, Shi Y, Figeys D, Ding L, Wang P, Zhang B, Rodriguez H, Gao Q, Gao D, Zhou H, Fan J. Proteogenomic characterization identifies clinically relevant subgroups of intrahepatic cholangiocarcinoma. Cancer Cell 2022; 40:70-87.e15. [PMID: 34971568 DOI: 10.1016/j.ccell.2021.12.006] [Citation(s) in RCA: 159] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/19/2021] [Accepted: 12/08/2021] [Indexed: 02/08/2023]
Abstract
We performed proteogenomic characterization of intrahepatic cholangiocarcinoma (iCCA) using paired tumor and adjacent liver tissues from 262 patients. Integrated proteogenomic analyses prioritized genetic aberrations and revealed hallmarks of iCCA pathogenesis. Aflatoxin signature was associated with tumor initiation, proliferation, and immune suppression. Mutation-associated signaling profiles revealed that TP53 and KRAS co-mutations may contribute to iCCA metastasis via the integrin-FAK-SRC pathway. FGFR2 fusions activated the Rho GTPase pathway and could be a potential source of neoantigens. Proteomic profiling identified four patient subgroups (S1-S4) with subgroup-specific biomarkers. These proteomic subgroups had distinct features in prognosis, genetic alterations, microenvironment dysregulation, tumor microbiota composition, and potential therapeutics. SLC16A3 and HKDC1 were further identified as potential prognostic biomarkers associated with metabolic reprogramming of iCCA cells. This study provides a valuable resource for researchers and clinicians to further identify molecular pathogenesis and therapeutic opportunities in iCCA.
Collapse
Affiliation(s)
- Liangqing Dong
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Dayun Lu
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China
| | - Ran Chen
- University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Youpei Lin
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Hongwen Zhu
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Zhou Zhang
- Burning Rock Biotech, Shanghai 201114, China
| | - Shangli Cai
- Burning Rock Biotech, Shanghai 201114, China
| | - Peng Cui
- Burning Rock Biotech, Shanghai 201114, China
| | - Guohe Song
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Dongning Rao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Xinpei Yi
- Lester and Sue Smith Breast Center, Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yingcheng Wu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Nixue Song
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China
| | - Fen Liu
- University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Yunhao Zou
- University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Shu Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Xiaoming Zhang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoying Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Shuangjian Qiu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China; Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Shisheng Wang
- Frontiers Science Center for Disease-related Molecular Network, Institutes for Systems Genetics, Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xu Zhang
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Shanghai Institute of Materia Medica-University of Ottawa Joint Research Center in Systems and Personalized Pharmacology, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yongyong Shi
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), The Collaborative Innovation Center for Brain Science, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Daniel Figeys
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Shanghai Institute of Materia Medica-University of Ottawa Joint Research Center in Systems and Personalized Pharmacology, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Li Ding
- Department of Medicine, McDonnell Genome Institute, Siteman Cancer Center, Washington University, St. Louis, MI 63108, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NewYork, NY 10029, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China; Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200433, China.
| | - Daming Gao
- University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Hu Zhou
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; Shanghai Institute of Materia Medica-University of Ottawa Joint Research Center in Systems and Personalized Pharmacology, 555 Zuchongzhi Road, Shanghai 201203, China.
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China; Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
25
|
Zapater JL, Lednovich KR, Khan MW, Pusec CM, Layden BT. Hexokinase domain-containing protein-1 in metabolic diseases and beyond. Trends Endocrinol Metab 2022; 33:72-84. [PMID: 34782236 PMCID: PMC8678314 DOI: 10.1016/j.tem.2021.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 12/16/2022]
Abstract
Glucose phosphorylation by hexokinases (HKs) traps glucose in cells and facilitates its usage in metabolic processes dependent on cellular needs. HK domain-containing protein-1 (HKDC1) is a recently discovered protein with wide expression containing HK activity, first noted through a genome-wide association study (GWAS) to be linked with gestational glucose homeostasis during pregnancy. Since then, HKDC1 has been observed to be expressed in many human tissues. Moreover, studies have shown that HKDC1 plays a role in glucose homeostasis by which it may affect the progression of many pathophysiological conditions such as gestational diabetes mellitus (GDM), nonalcoholic steatohepatitis (NASH), and cancer. Here, we review the key studies contributing to our current understanding of the roles of HKDC1 in human pathophysiological conditions and potential therapeutic interventions.
Collapse
Affiliation(s)
- Joseph L Zapater
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Kristen R Lednovich
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Md Wasim Khan
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Carolina M Pusec
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Brian T Layden
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Jesse Brown VA Medical Center, Chicago, IL, USA.
| |
Collapse
|
26
|
Hon KW, Zainal Abidin SA, Othman I, Naidu R. The Crosstalk Between Signaling Pathways and Cancer Metabolism in Colorectal Cancer. Front Pharmacol 2021; 12:768861. [PMID: 34887764 PMCID: PMC8650587 DOI: 10.3389/fphar.2021.768861] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most frequently diagnosed cancers worldwide. Metabolic reprogramming represents an important cancer hallmark in CRC. Reprogramming core metabolic pathways in cancer cells, such as glycolysis, glutaminolysis, oxidative phosphorylation, and lipid metabolism, is essential to increase energy production and biosynthesis of precursors required to support tumor initiation and progression. Accumulating evidence demonstrates that activation of oncogenes and loss of tumor suppressor genes regulate metabolic reprogramming through the downstream signaling pathways. Protein kinases, such as AKT and c-MYC, are the integral components that facilitate the crosstalk between signaling pathways and metabolic pathways in CRC. This review provides an insight into the crosstalk between signaling pathways and metabolic reprogramming in CRC. Targeting CRC metabolism could open a new avenue for developing CRC therapy by discovering metabolic inhibitors and repurposing protein kinase inhibitors/monoclonal antibodies.
Collapse
Affiliation(s)
| | | | | | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
27
|
Huang G, Zhang J, Gong L, Huang Y, Liu D. A glycolysis-based three-gene signature predicts survival in patients with lung squamous cell carcinoma. BMC Cancer 2021; 21:626. [PMID: 34044809 PMCID: PMC8161559 DOI: 10.1186/s12885-021-08360-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 05/13/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lung cancer is one of the most lethal and most prevalent malignant tumors worldwide, and lung squamous cell carcinoma (LUSC) is one of the major histological subtypes. Although numerous biomarkers have been found to be associated with prognosis in LUSC, the prediction effect of a single gene biomarker is insufficient, especially for glycolysis-related genes. Therefore, we aimed to develop a novel glycolysis-related gene signature to predict survival in patients with LUSC. METHODS The mRNA expression files and LUSC clinical information were obtained from The Cancer Genome Atlas (TCGA) dataset. RESULTS Based on Gene Set Enrichment Analysis (GSEA), we found 5 glycolysis-related gene sets that were significantly enriched in LUSC tissues. Univariate and multivariate Cox proportional regression models were performed to choose prognostic-related gene signatures. Based on a Cox proportional regression model, a risk score for a three-gene signature (HKDC1, ALDH7A1, and MDH1) was established to divide patients into high-risk and low-risk subgroups. Multivariate Cox regression analysis indicated that the risk score for this three-gene signature can be used as an independent prognostic indicator in LUSC. Additionally, based on the cBioPortal database, the rate of genomic alterations in the HKDC1, ALDH7A1, and MDH1 genes were 1.9, 1.1, and 5% in LUSC patients, respectively. CONCLUSION A glycolysis-based three-gene signature could serve as a novel biomarker in predicting the prognosis of patients with LUSC and it also provides additional gene targets that can be used to cure LUSC patients.
Collapse
Affiliation(s)
- Guichuan Huang
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), No 98 Fenghuang Road, Huichuan District, Zunyi, 563000, China
| | - Jing Zhang
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Ling Gong
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), No 98 Fenghuang Road, Huichuan District, Zunyi, 563000, China
| | - Yi Huang
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), No 98 Fenghuang Road, Huichuan District, Zunyi, 563000, China
| | - Daishun Liu
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), No 98 Fenghuang Road, Huichuan District, Zunyi, 563000, China.
| |
Collapse
|
28
|
Ciscato F, Ferrone L, Masgras I, Laquatra C, Rasola A. Hexokinase 2 in Cancer: A Prima Donna Playing Multiple Characters. Int J Mol Sci 2021; 22:ijms22094716. [PMID: 33946854 PMCID: PMC8125560 DOI: 10.3390/ijms22094716] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 12/21/2022] Open
Abstract
Hexokinases are a family of ubiquitous exose-phosphorylating enzymes that prime glucose for intracellular utilization. Hexokinase 2 (HK2) is the most active isozyme of the family, mainly expressed in insulin-sensitive tissues. HK2 induction in most neoplastic cells contributes to their metabolic rewiring towards aerobic glycolysis, and its genetic ablation inhibits malignant growth in mouse models. HK2 can dock to mitochondria, where it performs additional functions in autophagy regulation and cell death inhibition that are independent of its enzymatic activity. The recent definition of HK2 localization to contact points between mitochondria and endoplasmic reticulum called Mitochondria Associated Membranes (MAMs) has unveiled a novel HK2 role in regulating intracellular Ca2+ fluxes. Here, we propose that HK2 localization in MAMs of tumor cells is key in sustaining neoplastic progression, as it acts as an intersection node between metabolic and survival pathways. Disrupting these functions by targeting HK2 subcellular localization can constitute a promising anti-tumor strategy.
Collapse
Affiliation(s)
- Francesco Ciscato
- Dipartimento di Scienze Biomediche, Università di Padova, 35131 Padova, Italy; (L.F.); (I.M.); (C.L.)
- Correspondence: (F.C.); (A.R.)
| | - Lavinia Ferrone
- Dipartimento di Scienze Biomediche, Università di Padova, 35131 Padova, Italy; (L.F.); (I.M.); (C.L.)
| | - Ionica Masgras
- Dipartimento di Scienze Biomediche, Università di Padova, 35131 Padova, Italy; (L.F.); (I.M.); (C.L.)
- Institute of Neuroscience, National Research Council, 56124 Pias, Italy
| | - Claudio Laquatra
- Dipartimento di Scienze Biomediche, Università di Padova, 35131 Padova, Italy; (L.F.); (I.M.); (C.L.)
| | - Andrea Rasola
- Dipartimento di Scienze Biomediche, Università di Padova, 35131 Padova, Italy; (L.F.); (I.M.); (C.L.)
- Correspondence: (F.C.); (A.R.)
| |
Collapse
|
29
|
Massey V, Parrish A, Argemi J, Moreno M, Mello A, García-Rocha M, Altamirano J, Odena G, Dubuquoy L, Louvet A, Martinez C, Adrover A, Affò S, Morales-Ibanez O, Sancho-Bru P, Millán C, Alvarado-Tapias E, Morales-Arraez D, Caballería J, Mann J, Cao S, Sun Z, Shah V, Cameron A, Mathurin P, Snider N, Villanueva C, Morgan TR, Guinovart J, Vadigepalli R, Bataller R. Integrated Multiomics Reveals Glucose Use Reprogramming and Identifies a Novel Hexokinase in Alcoholic Hepatitis. Gastroenterology 2021; 160:1725-1740.e2. [PMID: 33309778 PMCID: PMC8613537 DOI: 10.1053/j.gastro.2020.12.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 11/06/2020] [Accepted: 12/01/2020] [Indexed: 02/02/2023]
Abstract
BACKGROUND & AIMS We recently showed that alcoholic hepatitis (AH) is characterized by dedifferentiation of hepatocytes and loss of mature functions. Glucose metabolism is tightly regulated in healthy hepatocytes. We hypothesize that AH may lead to metabolic reprogramming of the liver, including dysregulation of glucose metabolism. METHODS We performed integrated metabolomic and transcriptomic analyses of liver tissue from patients with AH or alcoholic cirrhosis or normal liver tissue from hepatic resection. Focused analyses of chromatin immunoprecipitation coupled to DNA sequencing was performed. Functional in vitro studies were performed in primary rat and human hepatocytes and HepG2 cells. RESULTS Patients with AH exhibited specific changes in the levels of intermediates of glycolysis/gluconeogenesis, the tricarboxylic acid cycle, and monosaccharide and disaccharide metabolism. Integrated analysis of the transcriptome and metabolome showed the used of alternate energetic pathways, metabolite sinks and bottlenecks, and dysregulated glucose storage in patients with AH. Among genes involved in glucose metabolism, hexokinase domain containing 1 (HKDC1) was identified as the most up-regulated kinase in patients with AH. Histone active promoter and enhancer markers were increased in the HKDC1 genomic region. High HKDC1 levels were associated with the development of acute kidney injury and decreased survival. Increased HKDC1 activity contributed to the accumulation of glucose-6-P and glycogen in primary rat hepatocytes. CONCLUSIONS Altered metabolite levels and messenger RNA expression of metabolic enzymes suggest the existence of extensive reprogramming of glucose metabolism in AH. Increased HKDC1 expression may contribute to dysregulated glucose metabolism and represents a novel biomarker and therapeutic target for AH.
Collapse
Affiliation(s)
- Veronica Massey
- Division of Gastroenterology and Hepatology, Departments of Medicine and Nutrition, and Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, North Carolina
| | - Austin Parrish
- Daniel Baugh Institute, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Josepmaria Argemi
- Department of Gastroenterology and Hepatology, Division of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; Liver Unit, Clinica Universidad de Navarra. Hepatology Program, Center for Applied Medical Research, IdisNA, Pamplona, Spain
| | - Montserrat Moreno
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Aline Mello
- Department of Gastroenterology and Hepatology, Division of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Mar García-Rocha
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Jose Altamirano
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; Liver Unit, Internal Medicine Department, Hospital Universitari Vall d'Hebrón, Vall d'Hebrón Institut de Recerca, Barcelona, Spain
| | - Gemma Odena
- Division of Gastroenterology and Hepatology, Departments of Medicine and Nutrition, and Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, North Carolina
| | - Laurent Dubuquoy
- Service des Maladies de l'appareil digestif, CHU Lille, Inserm LIRIC-UMR995, University of Lille, Lille, France
| | - Alexandre Louvet
- Service des Maladies de l'appareil digestif, CHU Lille, Inserm LIRIC-UMR995, University of Lille, Lille, France
| | - Carlos Martinez
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Anna Adrover
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Silvia Affò
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | | | - Pau Sancho-Bru
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Cristina Millán
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Edilmar Alvarado-Tapias
- Department of Gastroenterology, Hospital Santa Creu i Sant Pau, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, Madrid, Spain
| | - Dalia Morales-Arraez
- Department of Gastroenterology and Hepatology, Division of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Juan Caballería
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; Liver Unit, Hospital Clínic, CIBER de Enfermedades Hepáticas y Digestivas, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Jelena Mann
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Sheng Cao
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Zhaoli Sun
- Johns Hopkins School of Medicine, Department of Surgery and Transplant Biology Research Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Vijay Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Andrew Cameron
- Johns Hopkins School of Medicine, Department of Surgery and Transplant Biology Research Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Phillipe Mathurin
- Service des Maladies de l'appareil digestif, CHU Lille, Inserm LIRIC-UMR995, University of Lille, Lille, France
| | - Natasha Snider
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina
| | - Càndid Villanueva
- Department of Gastroenterology, Hospital Santa Creu i Sant Pau, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, Madrid, Spain; Institut de Recerca, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), Barcelona, Spain
| | - Timothy R Morgan
- Gastroenterology Services, VA Long Beach Healthcare, VA Long Beach Healthcare System, Long Beach, California
| | - Joan Guinovart
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ramon Bataller
- Division of Gastroenterology and Hepatology, Departments of Medicine and Nutrition, and Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, North Carolina; Department of Gastroenterology and Hepatology, Division of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.
| |
Collapse
|
30
|
The Synthetic Flavonoid Derivative GL-V9 Induces Apoptosis and Autophagy in Cutaneous Squamous Cell Carcinoma via Suppressing AKT-Regulated HK2 and mTOR Signals. Molecules 2020; 25:molecules25215033. [PMID: 33143000 PMCID: PMC7663336 DOI: 10.3390/molecules25215033] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/20/2020] [Accepted: 10/24/2020] [Indexed: 02/07/2023] Open
Abstract
Cutaneous squamous-cell carcinoma (cSCC) is one of most common type of non-black skin cancer. The malignancy degree and the death risk of cSCC patients are significantly higher than basal cell carcinoma patients. GL-V9 is a synthesized flavonoid derived from natural active ingredient wogonin and shows potent growth inhibitory effects in liver and breast cancer cells. In this study, we investigated the anti-cSCC effect and the underlying mechanism of GL-V9. The results showed that GL-V9 induced both apoptosis and autophagy in human cSCC cell line A431 cells, and prevented the growth progression of chemical induced primary skin cancer in mice. Metabolomics assay showed that GL-V9 potentially affected mitochondrial function, inhibiting glucose metabolism and Warburg effect. Further mechanism studies demonstrated that AKT played important roles in the anti-cSCC effect of GL-V9. On one hand, GL-V9 suppressed AKT-modulated mitochondrial localization of HK2 and promoted the protein degradation of HK2, resulting in cell apoptosis and glycolytic inhibition. On the other hand, GL-V9 induced autophagy via inhibiting Akt/mTOR pathway. Interestingly, though the autophagy induced by GL-V9 potentially antagonized its effect of apoptosis induction, the anti-cSCC effect of GL-V9 was not diluted. All above, our studies suggest that GL-V9 is a potent candidate for cSCC treatment.
Collapse
|
31
|
Han W, Shi J, Cao J, Dong B, Guan W. Emerging Roles and Therapeutic Interventions of Aerobic Glycolysis in Glioma. Onco Targets Ther 2020; 13:6937-6955. [PMID: 32764985 PMCID: PMC7371605 DOI: 10.2147/ott.s260376] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/26/2020] [Indexed: 12/20/2022] Open
Abstract
Glioma is the most common type of intracranial malignant tumor, with a great recurrence rate due to its infiltrative growth, treatment resistance, intra- and intertumoral genetic heterogeneity. Recently, accumulating studies have illustrated that activated aerobic glycolysis participated in various cellular and clinical activities of glioma, thus influencing the efficacy of radiotherapy and chemotherapy. However, the glycolytic process is too complicated and ambiguous to serve as a novel therapy for glioma. In this review, we generalized the implication of key enzymes, glucose transporters (GLUTs), signalings and transcription factors in the glycolytic process of glioma. In addition, we summarized therapeutic interventions via the above aspects and discussed promising clinical applications for glioma.
Collapse
Affiliation(s)
- Wei Han
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| | - Jia Shi
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| | - Jiachao Cao
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| | - Bo Dong
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| | - Wei Guan
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| |
Collapse
|
32
|
Garcia SN, Guedes RC, Marques MM. Unlocking the Potential of HK2 in Cancer Metabolism and Therapeutics. Curr Med Chem 2020; 26:7285-7322. [PMID: 30543165 DOI: 10.2174/0929867326666181213092652] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/26/2018] [Accepted: 11/06/2018] [Indexed: 12/24/2022]
Abstract
Glycolysis is a tightly regulated process in which several enzymes, such as Hexokinases (HKs), play crucial roles. Cancer cells are characterized by specific expression levels of several isoenzymes in different metabolic pathways and these features offer possibilities for therapeutic interventions. Overexpression of HKs (mostly of the HK2 isoform) have been consistently reported in numerous types of cancer. Moreover, deletion of HK2 has been shown to decrease cancer cell proliferation without explicit side effects in animal models, which suggests that targeting HK2 is a viable strategy for cancer therapy. HK2 inhibition causes a substantial decrease of glycolysis that affects multiple pathways of central metabolism and also destabilizes the mitochondrial outer membrane, ultimately enhancing cell death. Although glycolysis inhibition has met limited success, partly due to low selectivity for specific isoforms and excessive side effects of the reported HK inhibitors, there is ample ground for progress. The current review is focused on HK2 inhibition, envisaging the development of potent and selective anticancer agents. The information on function, expression, and activity of HKs is presented, along with their structures, known inhibitors, and reported effects of HK2 ablation/inhibition. The structural features of the different isozymes are discussed, aiming to stimulate a more rational approach to the design of selective HK2 inhibitors with appropriate drug-like properties. Particular attention is dedicated to a structural and sequence comparison of the structurally similar HK1 and HK2 isoforms, aiming to unveil differences that could be explored therapeutically. Finally, several additional catalytic- and non-catalytic roles on different pathways and diseases, recently attributed to HK2, are reviewed and their implications briefly discussed.
Collapse
Affiliation(s)
- Sara N Garcia
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal.,iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Rita C Guedes
- iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - M Matilde Marques
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| |
Collapse
|
33
|
Guo W, Kuang Y, Wu J, Wen D, Zhou A, Liao Y, Song H, Xu D, Wang T, Jing B, Li K, Hu M, Ling J, Wang Q, Wu W. Hexokinase 2 Depletion Confers Sensitization to Metformin and Inhibits Glycolysis in Lung Squamous Cell Carcinoma. Front Oncol 2020; 10:52. [PMID: 32083006 PMCID: PMC7005048 DOI: 10.3389/fonc.2020.00052] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 01/13/2020] [Indexed: 12/21/2022] Open
Abstract
Lung squamous cell carcinomas (SCCs) are highly aggressive tumors, and there is currently no effective targeted therapy owing to the lack of specific mutation targets. Compared with lung adenocarcinoma (ADCs), lung SCCs reportedly utilized higher levels of glucose metabolism to meet the anabolic and catabolic needs required to sustain rapid tumor growth. Hexokinase 2 (HK2) is an enzyme that catalyzes the rate-limit and first committed step in glucose metabolism. Here, we investigated the expression and effect of HK2 in lung SCCs. We found a significantly higher HK2 expression in lung SCCs, but not lung ADC or normal tissues. HK2 depletion or inhibition decreased the glycolysis and tumor growth via activating AMPK signaling pathway, which downregulated mTORC1 activity. Furthermore, we found an increased oxygen respiration rate compensating for HK2 depletion. Thus, metformin treatment showed combinatorial therapeutic value, which resulted in greater induction of lung SCC apoptosis in vitro and in vivo. Our study suggests that HK2 depletion in combination with metformin might be a novel effective strategy for lung SCCs therapy.
Collapse
Affiliation(s)
- Wenzheng Guo
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanbin Kuang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Dalian Medical University, Dalian, China.,Department of Respiratory Medicine, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jingjing Wu
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Donghua Wen
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Aiping Zhou
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yueling Liao
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyong Song
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongliang Xu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tong Wang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Jing
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kaimi Li
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Hu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Ling
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Wenjuan Wu
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Khan MW, Priyadarshini M, Cordoba-Chacon J, Becker TC, Layden BT. Hepatic hexokinase domain containing 1 (HKDC1) improves whole body glucose tolerance and insulin sensitivity in pregnant mice. Biochim Biophys Acta Mol Basis Dis 2019; 1865:678-687. [PMID: 30543855 PMCID: PMC6387585 DOI: 10.1016/j.bbadis.2018.11.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/19/2018] [Accepted: 11/26/2018] [Indexed: 02/04/2023]
Abstract
Hexokinase domain containing 1, a recently discovered putative fifth hexokinase, is hypothesized to play key roles in glucose metabolism. Specifically, during pregnancy in a recent genome wide association study (GWAS), a strong correlation between HKDC1 and 2-h plasma glucose in pregnant women from different ethnic backgrounds was shown. Our earlier work also reported diminished glucose tolerance during pregnancy in our whole body HKDC1 heterozygous mice. Therefore, we hypothesized that HKDC1 plays important roles in gestational metabolism, and designed this study to assess the role of hepatic HKDC1 in whole body glucose utilization and insulin action during pregnancy. We overexpressed human HKDC1 in mouse liver by injecting a human HKDC1 adenoviral construct; whereas, for the liver-specific HKDC1 knockout model, we used AAV-Cre constructs in our HKDC1fl/fl mice. Both groups of mice were subjected to metabolic testing before and during pregnancy on gestation day 17-18. Our results indicate that hepatic HKDC1 overexpression during pregnancy leads to improved whole-body glucose tolerance and enhanced hepatic and peripheral insulin sensitivity while hepatic HKDC1 knockout results in diminished glucose tolerance. Further, we observed reduced gluconeogenesis with hepatic HKDC1 overexpression while HKDC1 knockout led to increased gluconeogenesis. These changes were associated with significantly enhanced ketone body production in HKDC1 overexpressing mice, indicating that these mice shift their metabolic needs from glucose reliance to greater fat oxidation and ketone utilization during fasting. Taken together, our results indicate that hepatic HKDC1 contributes to whole body glucose disposal, insulin sensitivity, and aspects of nutrient balance during pregnancy.
Collapse
Affiliation(s)
- Md Wasim Khan
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, IL, USA
| | - Medha Priyadarshini
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, IL, USA
| | - Jose Cordoba-Chacon
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, IL, USA
| | - Thomas C Becker
- Duke Molecular Physiology Institute, Department of Internal Medicine, Duke University Medical Center, Durham, NC, USA
| | - Brian T Layden
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, IL, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA.
| |
Collapse
|
35
|
Pusec CM, De Jesus A, Khan MW, Terry AR, Ludvik AE, Xu K, Giancola N, Pervaiz H, Daviau Smith E, Ding X, Harrison S, Chandel NS, Becker TC, Hay N, Ardehali H, Cordoba-Chacon J, Layden BT. Hepatic HKDC1 Expression Contributes to Liver Metabolism. Endocrinology 2019; 160:313-330. [PMID: 30517626 PMCID: PMC6334269 DOI: 10.1210/en.2018-00887] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/16/2018] [Indexed: 12/15/2022]
Abstract
Glucokinase (GCK) is the principal hexokinase (HK) in the liver, operating as a glucose sensor to regulate glucose metabolism and lipid homeostasis. Recently, we proposed HK domain-containing 1 (HKDC1) to be a fifth HK with expression in the liver. Here, we reveal HKDC1 to have low glucose-phosphorylating ability and demonstrate its association with the mitochondria in hepatocytes. As we have shown previously that genetic deletion of HKDC1 leads to altered hepatic triglyceride levels, we also explored the influence of overexpression of HKDC1 in hepatocytes on cellular metabolism, observing reduced glycolytic capacity and maximal mitochondrial respiration with concurrent reductions in glucose oxidation and mitochondrial membrane potential. Furthermore, we found that acute in vivo overexpression of HKDC1 in the liver induced substantial changes in mitochondrial dynamics. Altogether, these findings suggest that overexpression of HKDC1 causes mitochondrial dysfunction in hepatocytes. However, its overexpression was not enough to alter energy storage in the liver but led to mild improvement in glucose tolerance. We next investigated the conditions necessary to induce HKDC1 expression, observing HKDC1 expression to be elevated in human patients whose livers were at more advanced stages of nonalcoholic fatty liver disease (NAFLD) and similarly, found high liver expression in mice on diets causing high levels of liver inflammation and fibrosis. Overall, our data suggest that HKDC1 expression in hepatocytes results in defective mitochondrial function and altered hepatocellular metabolism and speculate that its expression in the liver may play a role in the development of NAFLD.
Collapse
Affiliation(s)
- Carolina M Pusec
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Adam De Jesus
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Md Wasim Khan
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Alexander R Terry
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Anton E Ludvik
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Kai Xu
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Nicholas Giancola
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Haaris Pervaiz
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Xianzhong Ding
- Department of Pathology, Loyola University Chicago, Maywood, Illinois
| | | | - Navdeep S Chandel
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Thomas C Becker
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University Medical Center, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| | - Nissim Hay
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Hossein Ardehali
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jose Cordoba-Chacon
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Brian T Layden
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Jesse Brown VA Medical Center, Chicago, Illinois
| |
Collapse
|
36
|
Luo F, Li Y, Yuan F, Zuo J. Hexokinase II promotes the Warburg effect by phosphorylating alpha subunit of pyruvate dehydrogenase. Chin J Cancer Res 2019; 31:521-532. [PMID: 31354221 PMCID: PMC6613503 DOI: 10.21147/j.issn.1000-9604.2019.03.14] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Objective Tumor cells rely heavily on glycolysis regardless of oxygen tension, a phenomenon called the Warburg effect. Hexokinase II (HKII) catalyzes the first irreversible step of glycolysis and is often overexpressed in tumor cells. Mitochondrial HKII couples glycolysis and oxidative phosphorylation while maintaining mitochondrial membrane integrity. In this study, we investigated the role of HKII in promoting the Warburg effect in cancer cells. Methods HKII-mediated phosphorylation of the alpha subunit of pyruvate dehydrogenase (PDHA1) was tested in HEK293T cells and clear cell renal cell carcinoma (ccRCC) specimens using gene knockdown, western blotting, immunohistochemistry, and immunofluorescence. Results It was determined that HKII could not only transform glucose into glucose-6-phosphate, but also transfer the phosphate group of ATP onto PDHA1. In addition, it was found that HKII increased the phosphorylation of Ser293 on PDHA1, decreasing pyruvate dehydrogenase (PDH) complex activity and thus rerouting the metabolic pathway and promoting the Warburg effect. The overexpression of HKII correlated with the phosphorylation of PDHA1 and disease progression in ccRCC. Conclusions The data presented here suggest that HKII is an important biomarker in the evaluation and treatment of cancer.
Collapse
Affiliation(s)
- Fangxiu Luo
- Department of Pathology, Ruijin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai 201801, China
| | - You Li
- Department of General Surgery, Ruijin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai 201801, China
| | - Fei Yuan
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Junli Zuo
- Department of Geriatrics, Ruijin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai 201801, China
| |
Collapse
|
37
|
Zhang L, Sun Z, Zhao P, Huang L, Xu M, Yang Y, Chen X, Lu F, Zhang X, Wang H, Zhang S, Liu W, Jiang Z, Ma S, Chen R, Zhao C, Yang Z, Sui R, Zhu X. Whole-exome sequencing revealed HKDC1 as a candidate gene associated with autosomal-recessive retinitis pigmentosa. Hum Mol Genet 2018; 27:4157-4168. [PMID: 30085091 PMCID: PMC6240732 DOI: 10.1093/hmg/ddy281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/02/2018] [Accepted: 07/23/2018] [Indexed: 01/04/2023] Open
Abstract
Retinitis pigmentosa (RP) is an inheritable retina degenerative disease leading to blindness. Despite the identification of 70 genes associated with RP, the genetic cause of ∼40% of RP patients remains to be elucidated. Whole-exome sequencing was applied on the probands of a RP cohort of 68 unsolved cases to identify candidate genetic mutations. A homozygous missense variant (c.173C > T, p.T58 M) was found in HKDC1 in two unrelated families presenting late-onset retinal degeneration. This variant affects highly conserved amino acid residue and is very rare in several databases and absent in 4000 ethnic-matched controls. Mutant HKDC1 protein partially lost hexokinase activity. Hkdc1 is expressed in the mouse retina and localized to photoreceptor inner segments. To elucidate the in vivo roles of Hkdc1 in the retina, we generated Hkdc1 knockout (KO) mouse models using CRISPR/Cas9 technique. Two independent alleles were identified and backcrossed to C57BL/6 J for 6 generations. Absence of HKDC1 expression in the Hkdc1 KO retina was confirmed by western blot and immunostaning using HKDC1 antibody. Hkdc1 KO mice exhibited reduced scotopic electroretinogram response and thinner outer nuclear layer, similar to some of the human patient phenotypes. Loss of Hkdc1 led to mislocalization of rhodopsin to the inner segments and cell bodies of rods in some regions in the retina. Taken together, our data demonstrated that HKDC1 is associated with autosomal recessively inherited RP.
Collapse
Affiliation(s)
- Lin Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Center of Information in Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zixi Sun
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Peiquan Zhao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lulin Huang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Mingchu Xu
- Department of Molecular and Human Genetics
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Yeming Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xue Chen
- Department of Ophthalmology, Hospital of Nanjing Medical University, State Key Laboratory of Reproductive Medicine, Nanjing, China
| | - Fang Lu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xiang Zhang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hui Wang
- Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Shanshan Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Wenjing Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zhilin Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Institute of Laboratory Medicine, SichuanAcademy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
| | - Shi Ma
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Institute of Laboratory Medicine, SichuanAcademy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
| | - Rui Chen
- Department of Molecular and Human Genetics
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Chen Zhao
- Department of Ophthalmology, Hospital of Nanjing Medical University, State Key Laboratory of Reproductive Medicine, Nanjing, China
- Department of Ophthalmology and Vision Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Ophthalmology, Children’s Hospital of Zhengzhou, Zhengzhou, China
| | - Zhenglin Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Institute of Laboratory Medicine, SichuanAcademy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Hospital, Institute of Chengdu Biology Chengdu, China
| | - Ruifang Sui
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xianjun Zhu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Institute of Laboratory Animal Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
- Institute of Laboratory Medicine, SichuanAcademy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Hospital, Institute of Chengdu Biology Chengdu, China
- Center of Information in Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
38
|
Fuhr L, El-Athman R, Scrima R, Cela O, Carbone A, Knoop H, Li Y, Hoffmann K, Laukkanen MO, Corcione F, Steuer R, Meyer TF, Mazzoccoli G, Capitanio N, Relógio A. The Circadian Clock Regulates Metabolic Phenotype Rewiring Via HKDC1 and Modulates Tumor Progression and Drug Response in Colorectal Cancer. EBioMedicine 2018; 33:105-121. [PMID: 30005951 PMCID: PMC6085544 DOI: 10.1016/j.ebiom.2018.07.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/27/2018] [Accepted: 07/03/2018] [Indexed: 12/15/2022] Open
Abstract
An endogenous molecular clockwork drives various cellular pathways including metabolism and the cell cycle. Its dysregulation is able to prompt pathological phenotypes including cancer. Besides dramatic metabolic alterations, cancer cells display severe changes in the clock phenotype with likely consequences in tumor progression and treatment response. In this study, we use a comprehensive systems-driven approach to investigate the effect of clock disruption on metabolic pathways and its impact on drug response in a cellular model of colon cancer progression. We identified distinctive time-related transcriptomic and metabolic features of a primary tumor and its metastatic counterpart. A mapping of the expression data to a comprehensive genome-scale reconstruction of human metabolism allowed for the in-depth functional characterization of 24 h-oscillating transcripts and pointed to a clock-driven metabolic reprogramming in tumorigenesis. In particular, we identified a set of five clock-regulated glycolysis genes, ALDH3A2, ALDOC, HKDC1, PCK2, and PDHB with differential temporal expression patterns. These findings were validated in organoids and in primary fibroblasts isolated from normal colon and colon adenocarcinoma from the same patient. We further identified a reciprocal connection of HKDC1 to the clock in the primary tumor, which is lost in the metastatic cells. Interestingly, a disruption of the core-clock gene BMAL1 impacts on HKDC1 and leads to a time-dependent rewiring of metabolism, namely an increase in glycolytic activity, as well as changes in treatment response. This work provides novel evidence regarding the complex interplay between the circadian clock and metabolic alterations in carcinogenesis and identifies new connections between both systems with pivotal roles in cancer progression and response to therapy.
Collapse
Affiliation(s)
- Luise Fuhr
- Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Institute for Theoretical Biology, Germany; Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, Germany
| | - Rukeia El-Athman
- Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Institute for Theoretical Biology, Germany; Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, Germany
| | - Rosella Scrima
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Olga Cela
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Annalucia Carbone
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Unit, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo (FG), Italy
| | - Henning Knoop
- Institute for Theoretical Biology, Institut für Biologie, Humboldt-Universität zu Berlin, Germany
| | - Yin Li
- Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Institute for Theoretical Biology, Germany; Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, Germany
| | - Karen Hoffmann
- Department of Molecular Biology, Max Planck Institute for Infection Biology Berlin, Germany
| | | | - Francesco Corcione
- Department of General, Laparoscopic and Robotic Surgery, Azienda Ospedaliera Specialistica dei Colli, Monaldi Hospital, Via Leonardo Bianchi, 80131 Naples, Italy
| | - Ralf Steuer
- Institute for Theoretical Biology, Institut für Biologie, Humboldt-Universität zu Berlin, Germany
| | - Thomas F Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology Berlin, Germany
| | - Gianluigi Mazzoccoli
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Unit, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo (FG), Italy
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Angela Relógio
- Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Institute for Theoretical Biology, Germany; Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, Germany.
| |
Collapse
|
39
|
Multi-tissue transcriptomic study reveals the main role of liver in the chicken adaptive response to a switch in dietary energy source through the transcriptional regulation of lipogenesis. BMC Genomics 2018. [PMID: 29514634 PMCID: PMC5842524 DOI: 10.1186/s12864-018-4520-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Because the cost of cereals is unstable and represents a large part of production charges for meat-type chicken, there is an urge to formulate alternative diets from more cost-effective feedstuff. We have recently shown that meat-type chicken source is prone to adapt to dietary starch substitution with fat and fiber. The aim of this study was to better understand the molecular mechanisms of this adaptation to changes in dietary energy sources through the fine characterization of transcriptomic changes occurring in three major metabolic tissues – liver, adipose tissue and muscle – as well as in circulating blood cells. Results We revealed the fine-tuned regulation of many hepatic genes encoding key enzymes driving glycogenesis and de novo fatty acid synthesis pathways and of some genes participating in oxidation. Among the genes expressed upon consumption of a high-fat, high-fiber diet, we highlighted CPT1A, which encodes a key enzyme in the regulation of fatty acid oxidation. Conversely, the repression of lipogenic genes by the high-fat diet was clearly associated with the down-regulation of SREBF1 transcripts but was not associated with the transcript regulation of MLXIPL and NR1H3, which are both transcription factors. This result suggests a pivotal role for SREBF1 in lipogenesis regulation in response to a decrease in dietary starch and an increase in dietary PUFA. Other prospective regulators of de novo hepatic lipogenesis were suggested, such as PPARD, JUN, TADA2A and KAT2B, the last two genes belonging to the lysine acetyl transferase (KAT) complex family regulating histone and non-histone protein acetylation. Hepatic glycogenic genes were also down-regulated in chickens fed a high-fat, high-fiber diet compared to those in chickens fed a starch-based diet. No significant dietary-associated variations in gene expression profiles was observed in the other studied tissues, suggesting that the liver mainly contributed to the adaptation of birds to changes in energy source and nutrients in their diets, at least at the transcriptional level. Moreover, we showed that PUFA deposition observed in the different tissues may not rely on transcriptional changes. Conclusion We showed the major role of the liver, at the gene expression level, in the adaptive response of chicken to dietary starch substitution with fat and fiber. Electronic supplementary material The online version of this article (10.1186/s12864-018-4520-5) contains supplementary material, which is available to authorized users.
Collapse
|
40
|
Evstafieva AG, Kovaleva IE, Shoshinova MS, Budanov AV, Chumakov PM. Implication of KRT16, FAM129A and HKDC1 genes as ATF4 regulated components of the integrated stress response. PLoS One 2018; 13:e0191107. [PMID: 29420561 PMCID: PMC5805170 DOI: 10.1371/journal.pone.0191107] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/28/2017] [Indexed: 02/06/2023] Open
Abstract
The ATF4 transcription factor is a key regulator of the adaptive integrated stress response (ISR) induced by various stresses and pathologies. Identification of novel transcription targets of ATF4 during ISR would contribute to the understanding of adaptive networks and help to identify novel therapeutic targets. We were previously searching for genes that display an inverse regulation mode by the transcription factors ATF4 and p53 in response to mitochondrial respiration chain complex III inhibition. Among the selected candidates the human genes for cytokeratine 16 (KRT16), anti-apoptotic protein Niban (FAM129A) and hexokinase HKDC1 have been found highly responsive to ATF4 overexpression. Here we explored potential roles of the induction of KRT16, FAM129A and HKDC1 genes in ISR. As verified by RT-qPCR, a dysfunction of mitochondrial respiration chain and ER stress resulted in a partially ATF4-dependent stimulation of KRT16, FAM129A and HKDC1 expression in the HCT116 colon carcinoma cell line. ISRIB, a specific inhibitor of ISR, was able to downregulate the ER stress-induced levels of KRT16, FAM129A and HKDC1 transcripts. An inhibition of ATF4 by RNAi attenuated the induction of KRT16, FAM129A and HKDC1 mRNAs in response to ER stress or to a dysfunctional mitochondrial respiration. The similar induction of the three genes was observed in another tumor-derived cervical carcinoma cell line HeLa. However, in HaCaT and HEK293T cells that display transformed phenotypes, but do not originate from patient-derived tumors, the ER stress-inducing treatments resulted in an upregulation of FAM129A and HKDC1, but not KRT16 transcripts, By a luciferase reporter approach we identified a highly active ATF4-responsive element within the upstream region of the KRT16 gene. The results suggest a conditional regulation of KRT16 gene by ATF4 that may be inhibited in normal cells, but engaged during cancer progression. Potential roles of KRT16, FAM129A and HKDC1 genes upregulation in adaptive stress responses and pathologies are discussed.
Collapse
Affiliation(s)
- Alexandra G. Evstafieva
- Belozersky Institute of Physical and Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- Department of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
- * E-mail: (AGE); (PMC)
| | - Irina E. Kovaleva
- Belozersky Institute of Physical and Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Maria S. Shoshinova
- Department of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Andrei V. Budanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Dublin, Ireland
| | - Peter M. Chumakov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Chumakov Institute of Poliomyelitis and Viral Encephalitides, Federal Scientific Center for Research and Development of Immune-Biology Products, Russian Academy of Sciences, Moscow, Russia
- * E-mail: (AGE); (PMC)
| |
Collapse
|
41
|
Khan MW, Ding X, Cotler SJ, Clarke M, Layden BT. Studies on the Tissue Localization of HKDC1, a Putative Novel Fifth Hexokinase, in Humans. J Histochem Cytochem 2018; 66:385-392. [PMID: 29401404 DOI: 10.1369/0022155418756849] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hexokinase domain component 1 (HKDC1) is a recently discovered novel protein, which is being promoted as a putative fifth hexokinase. Although the exact role HKDC1 plays in physiology is still unclear, it has been shown to be important during pregnancy in the regulation of glucose homeostasis. In this study, we have comprehensively studied the expression pattern of HKDC1 in the human body. Using human tissue sample, immunohistochemistry imaging was performed. Our studies indicate that the tissues with highest HKDC1 expression were the brush border epithelium of the intestines, parts of the pancreas, and lung alveolar macrophages. Future directions will be to understand the role of this fifth hexokinase in these tissues, with a focus on its relative function as compared with other endogenously expressed hexokinases.
Collapse
Affiliation(s)
- Md Wasim Khan
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Xianzhong Ding
- Department of Pathology, Section of Hepatology, Loyola University Chicago, Maywood, Illinois
| | - Scott J Cotler
- Department of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Michael Clarke
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Brian T Layden
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| |
Collapse
|
42
|
Ali A, Wathes DC, Swali A, Burns H, Burns S. A novel mammalian glucokinase exhibiting exclusive inorganic polyphosphate dependence in the cell nucleus. Biochem Biophys Rep 2017; 12:151-157. [PMID: 29090276 PMCID: PMC5645163 DOI: 10.1016/j.bbrep.2017.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 09/13/2017] [Accepted: 09/15/2017] [Indexed: 11/19/2022] Open
Abstract
Background Hexokinase and glucokinase enzymes are ubiquitously expressed and use ATP and ADP as substrates in mammalian systems and a variety of polyphosphate substrates and/or ATP in some eukaryotic and microbial systems. Polyphosphate synthesising or utilizing enzymes are widely expressed in microbial systems but have not been reported in mammalian systems, despite the presence of polyphosphate in mammalian cells. Only two micro-organisms have previously been shown to express an enzyme that uses polyphosphate exclusively. Methods A variety of experimental approaches, including NMR and NAD-linked assay systems were used to conduct a biochemical investigation of polyphosphate dependent glucokinase activity in mammalian tissues. Results A novel mammalian glucokinase, highly responsive to hexametaphosphate (HMP) but not ATP or ADP as a phosphoryl donor is present in the nuclei of mammalian hepatocytes. The liver enzyme exhibited sigmoidal kinetics with respect to glucose with a S0.5 of 12 mM, similar to the known kinetics of mammalian ATP-glucokinase. The Km for HMP (0.5 mM) was also similar to that of phosphoryl donors for mammalian ATP-glucokinases. The new enzyme was inhibited by several nucleotide phosphates. Conclusions We report the discovery of a polyphosphate-dependent enzyme system in mammalian cells with kinetics similar to established ATP-dependent glucokinase, also known to have a nuclear location. The kinetics suggest possible regulatory or redox protective roles. General significance The role of polyphosphate in mammalian systems has remained an enigma for decades, and the present report describes progress on the significance of this compound in intracellular metabolism in mammals. The first mammalian enzyme activity using polyphosphate as a phosphorylation substrate. A polyphosphate dependent glucokinase with kinetics similar to human glucokinase. Further evidence of discreet substrate specificity of hexokinases. A possible evolutionary link between polyphosphate and ATP utilization.
Collapse
Affiliation(s)
- Antasar Ali
- Biological Sciences, University of Huddersfield, Huddersfield UK HD1 3DH, United Kingdom
| | - D. Claire Wathes
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, London UK NW1 0TU, United Kingdom
| | - Angelina Swali
- School of Biosciences, University of Nottingham, Nottingham UK LE12 5RD, United Kingdom
| | - Helena Burns
- Biological Sciences, University of Huddersfield, Huddersfield UK HD1 3DH, United Kingdom
| | - Shamus Burns
- Biological Sciences, University of Huddersfield, Huddersfield UK HD1 3DH, United Kingdom
- Corresponding author.
| |
Collapse
|
43
|
Molecular Cloning and Functional Characterization of a Hexokinase from the Oriental River Prawn Macrobrachium nipponense in Response to Hypoxia. Int J Mol Sci 2017; 18:ijms18061256. [PMID: 28608798 PMCID: PMC5486078 DOI: 10.3390/ijms18061256] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 06/09/2017] [Accepted: 06/10/2017] [Indexed: 01/02/2023] Open
Abstract
Metabolic adjustment to hypoxia in Macrobrachium nipponense (oriental river prawn) implies a shift to anaerobic metabolism. Hexokinase (HK) is a key glycolytic enzyme in prawns. The involvement of HK in the hypoxia inducible factors (HIFs) pathway is unclear in prawns. In this study, the full-length cDNA for HK (MnHK) was obtained from M. nipponense, and its properties were characterized. The full-length cDNA (2385 bp) with an open reading frame of 1350 bp, encoded a 450-amino acid protein. MnHK contained highly conserved amino acids in the glucose, glucose-6-phosphate, ATP, and Mg+2 binding sites. Quantitative real-time reverse transcription PCR assays revealed the tissue-specific expression pattern of MnHK, with abundant expression in the muscle, and gills. Kinetic studies validated the hexokinase activity of recombinant HK. Silencing of HIF-1α or HIF-1β subunit genes blocked the induction of HK and its enzyme activities during hypoxia in muscles. The results suggested that MnHK is a key factor that increases the anaerobic rate, and is probably involved in the HIF-1 pathway related to highly active metabolism during hypoxia.
Collapse
|
44
|
Kanthimathi S, Liju S, Laasya D, Anjana RM, Mohan V, Radha V. Hexokinase Domain Containing 1 (HKDC1) Gene Variants and their Association with Gestational Diabetes Mellitus in a South Indian Population. Ann Hum Genet 2017; 80:241-5. [PMID: 27346736 DOI: 10.1111/ahg.12155] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 04/06/2016] [Accepted: 04/12/2016] [Indexed: 02/04/2023]
Abstract
Hexokinase domain containing 1 (HKDC1), a novel human hexokinase gene, is known to affect glucose metabolism and was shown to have a strong association with 2-h plasma glucose in pregnant women in a recent genome wide association study. This study aimed to evaluate the association of these regulatory variants of HKDC1 (rs1076224, rs4746822, rs2394529 and rs9645501) with gestational diabetes mellitus (GDM) in a South Indian population. The regulatory variants of HKDC1 were genotyped in unrelated 500 women with GDM and 510 non-GDM individuals by using the MassARRAY system and by direct DNA sequencing. The minor alleles of the HKDC1 gene regulatory variants, namely rs10762264 and rs4746822, showed a significant association with GDM and these alleles conferred as much as 1.24 and 1.34 times higher risk for GDM, respectively. This is the first study to demonstrate the association of HKDC1 genetic variants with susceptibility to GDM.
Collapse
Affiliation(s)
| | - Samuel Liju
- Madras Diabetes Research Foundation, Chennai, India
| | | | - Ranjit Mohan Anjana
- Madras Diabetes Research Foundation, Chennai, India.,Dr.Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
| | - Viswanathan Mohan
- Madras Diabetes Research Foundation, Chennai, India.,Dr.Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
| | | |
Collapse
|
45
|
Karolak JA, Gambin T, Pitarque JA, Molinari A, Jhangiani S, Stankiewicz P, Lupski JR, Gajecka M. Variants in SKP1, PROB1, and IL17B genes at keratoconus 5q31.1-q35.3 susceptibility locus identified by whole-exome sequencing. Eur J Hum Genet 2016; 25:73-78. [PMID: 27703147 DOI: 10.1038/ejhg.2016.130] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 07/27/2016] [Accepted: 08/05/2016] [Indexed: 12/16/2022] Open
Abstract
Keratoconus (KTCN) is a protrusion and thinning of the cornea, resulting in impairment of visual function. The extreme genetic heterogeneity makes it difficult to discover factors unambiguously influencing the KTCN phenotype. In this study, we used whole-exome sequencing (WES) and Sanger sequencing to reduce the number of candidate genes at the 5q31.1-q35.3 locus and to prioritize other potentially relevant variants in an Ecuadorian family with KTCN. We applied WES in two affected KTCN individuals from the Ecuadorian family that showed a suggestive linkage between the KTCN phenotype and the 5q31.1-q35.3 locus. Putative variants identified by WES were further evaluated in this family using Sanger sequencing. Exome capture discovered a total of 173 rare (minor allele frequency <0.001 in control population) nonsynonymous variants in both affected individuals. Among them, 16 SNVs were selected for further evaluation. Segregation analysis revealed that variants c.475T>G in SKP1, c.671G>A in PROB1, and c.527G>A in IL17B in the 5q31.1-q35.3 linkage region, and c.850G>A in HKDC1 in the 10q22 locus completely segregated with the phenotype in the studied KTCN family. We demonstrate that a combination of various techniques significantly narrowed the studied genomic region and reduced the list of the putative exonic variants. Moreover, since this locus overlapped two other chromosomal regions previously recognized in distinct KTCN studies, our findings suggest that this 5q31.1-q35.3 locus might be linked with KTCN.
Collapse
Affiliation(s)
- Justyna A Karolak
- Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, Poznan, Poland.,Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Tomasz Gambin
- Institute of Computer Science, Warsaw University of Technology, Warsaw, Poland.,Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | - Jose A Pitarque
- Department of Ophthalmology, Hospital Metropolitano, Quito, Ecuador
| | - Andrea Molinari
- Department of Ophthalmology, Hospital Metropolitano, Quito, Ecuador
| | - Shalini Jhangiani
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Pawel Stankiewicz
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - James R Lupski
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Texas Children's Hospital, Houston, TX, USA
| | - Marzena Gajecka
- Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, Poznan, Poland. .,Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland.
| |
Collapse
|
46
|
Abstract
In recent years there has been a growing interest among cancer biologists in cancer metabolism. This Review summarizes past and recent advances in our understanding of the reprogramming of glucose metabolism in cancer cells, which is mediated by oncogenic drivers and by the undifferentiated character of cancer cells. The reprogrammed glucose metabolism in cancer cells is required to fulfil anabolic demands. This Review discusses the possibility of exploiting the reprogrammed glucose metabolism for therapeutic approaches that selectively target cancer cells.
Collapse
Affiliation(s)
- Nissim Hay
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois 60607 and Research and Development Section, Jesse Brown VA Medical Center, Chicago, Illinois 60612, USA
| |
Collapse
|
47
|
Ludvik AE, Pusec CM, Priyadarshini M, Angueira AR, Guo C, Lo A, Hershenhouse KS, Yang GY, Ding X, Reddy TE, Lowe WL, Layden BT. HKDC1 Is a Novel Hexokinase Involved in Whole-Body Glucose Use. Endocrinology 2016; 157:3452-61. [PMID: 27459389 PMCID: PMC5007896 DOI: 10.1210/en.2016-1288] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In a recent genome-wide association study, hexokinase domain-containing protein 1, or HKDC1, was found to be associated with gestational glucose levels during 2-hour glucose tolerance tests at 28 weeks of pregnancy. Because our understanding of the mediators of gestational glucose homeostasis is incomplete, we have generated the first transgenic mouse model to begin to understand the role of HKDC1 in whole-body glucose homeostasis. Interestingly, deletion of both HKDC1 alleles results in in utero embryonic lethality. Thus, in this study, we report the in vivo role of HKDC1 in whole-body glucose homeostasis using a heterozygous-deleted HKDC1 mouse model (HKDC1(+/-)) as compared with matched wild-type mice. First, we observed no weight, fasting or random glucose, or fasting insulin abnormalities with aging in male and female HKDC1(+/-) mice. However, during glucose tolerance tests, glucose levels were impaired in both female and male HKDC1(+/-) mice at 15, 30, and 120 minutes at a later age (28 wk of age). These glucose tolerance differences also existed in the female HKDC1(+/-) mice at earlier ages but only during pregnancy. And finally, the impaired glucose tolerance in HKDC1(+/-) mice was likely due to diminished whole-body glucose use, as indicated by the decreased hepatic energy storage and reduced peripheral tissue uptake of glucose in HKDC1(+/-) mice. Collectively, these data highlight that HKDC1 is needed to maintain whole-body glucose homeostasis during pregnancy but also with aging, possibly through its role in glucose use.
Collapse
Affiliation(s)
- Anton E Ludvik
- Division of Endocrinology, Metabolism, and Molecular Medicine (A.E.L., C.M.P., M.P., A.R.A., K.S.H., W.L.L., B.T.L.), Department of Pathology (A.L., G.-Y.Y.), Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; Department of Biostatistics and Bioinformatics (T.E.R.), and Center for Genomic and Computational Biology (C.G., T.E.R.), Duke University Medical School, and University Program in Genetics and Genomics (C.G.), Duke University, Durham, North Carolina 27710; Department of Pathology (X.D.), Loyola University Chicago, Maywood, Illinois 60153; and Jesse Brown Veterans Affairs Medical Center (B.T.L.), Chicago, Illinois 60612
| | - Carolina M Pusec
- Division of Endocrinology, Metabolism, and Molecular Medicine (A.E.L., C.M.P., M.P., A.R.A., K.S.H., W.L.L., B.T.L.), Department of Pathology (A.L., G.-Y.Y.), Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; Department of Biostatistics and Bioinformatics (T.E.R.), and Center for Genomic and Computational Biology (C.G., T.E.R.), Duke University Medical School, and University Program in Genetics and Genomics (C.G.), Duke University, Durham, North Carolina 27710; Department of Pathology (X.D.), Loyola University Chicago, Maywood, Illinois 60153; and Jesse Brown Veterans Affairs Medical Center (B.T.L.), Chicago, Illinois 60612
| | - Medha Priyadarshini
- Division of Endocrinology, Metabolism, and Molecular Medicine (A.E.L., C.M.P., M.P., A.R.A., K.S.H., W.L.L., B.T.L.), Department of Pathology (A.L., G.-Y.Y.), Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; Department of Biostatistics and Bioinformatics (T.E.R.), and Center for Genomic and Computational Biology (C.G., T.E.R.), Duke University Medical School, and University Program in Genetics and Genomics (C.G.), Duke University, Durham, North Carolina 27710; Department of Pathology (X.D.), Loyola University Chicago, Maywood, Illinois 60153; and Jesse Brown Veterans Affairs Medical Center (B.T.L.), Chicago, Illinois 60612
| | - Anthony R Angueira
- Division of Endocrinology, Metabolism, and Molecular Medicine (A.E.L., C.M.P., M.P., A.R.A., K.S.H., W.L.L., B.T.L.), Department of Pathology (A.L., G.-Y.Y.), Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; Department of Biostatistics and Bioinformatics (T.E.R.), and Center for Genomic and Computational Biology (C.G., T.E.R.), Duke University Medical School, and University Program in Genetics and Genomics (C.G.), Duke University, Durham, North Carolina 27710; Department of Pathology (X.D.), Loyola University Chicago, Maywood, Illinois 60153; and Jesse Brown Veterans Affairs Medical Center (B.T.L.), Chicago, Illinois 60612
| | - Cong Guo
- Division of Endocrinology, Metabolism, and Molecular Medicine (A.E.L., C.M.P., M.P., A.R.A., K.S.H., W.L.L., B.T.L.), Department of Pathology (A.L., G.-Y.Y.), Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; Department of Biostatistics and Bioinformatics (T.E.R.), and Center for Genomic and Computational Biology (C.G., T.E.R.), Duke University Medical School, and University Program in Genetics and Genomics (C.G.), Duke University, Durham, North Carolina 27710; Department of Pathology (X.D.), Loyola University Chicago, Maywood, Illinois 60153; and Jesse Brown Veterans Affairs Medical Center (B.T.L.), Chicago, Illinois 60612
| | - Amy Lo
- Division of Endocrinology, Metabolism, and Molecular Medicine (A.E.L., C.M.P., M.P., A.R.A., K.S.H., W.L.L., B.T.L.), Department of Pathology (A.L., G.-Y.Y.), Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; Department of Biostatistics and Bioinformatics (T.E.R.), and Center for Genomic and Computational Biology (C.G., T.E.R.), Duke University Medical School, and University Program in Genetics and Genomics (C.G.), Duke University, Durham, North Carolina 27710; Department of Pathology (X.D.), Loyola University Chicago, Maywood, Illinois 60153; and Jesse Brown Veterans Affairs Medical Center (B.T.L.), Chicago, Illinois 60612
| | - Korri S Hershenhouse
- Division of Endocrinology, Metabolism, and Molecular Medicine (A.E.L., C.M.P., M.P., A.R.A., K.S.H., W.L.L., B.T.L.), Department of Pathology (A.L., G.-Y.Y.), Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; Department of Biostatistics and Bioinformatics (T.E.R.), and Center for Genomic and Computational Biology (C.G., T.E.R.), Duke University Medical School, and University Program in Genetics and Genomics (C.G.), Duke University, Durham, North Carolina 27710; Department of Pathology (X.D.), Loyola University Chicago, Maywood, Illinois 60153; and Jesse Brown Veterans Affairs Medical Center (B.T.L.), Chicago, Illinois 60612
| | - Guang-Yu Yang
- Division of Endocrinology, Metabolism, and Molecular Medicine (A.E.L., C.M.P., M.P., A.R.A., K.S.H., W.L.L., B.T.L.), Department of Pathology (A.L., G.-Y.Y.), Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; Department of Biostatistics and Bioinformatics (T.E.R.), and Center for Genomic and Computational Biology (C.G., T.E.R.), Duke University Medical School, and University Program in Genetics and Genomics (C.G.), Duke University, Durham, North Carolina 27710; Department of Pathology (X.D.), Loyola University Chicago, Maywood, Illinois 60153; and Jesse Brown Veterans Affairs Medical Center (B.T.L.), Chicago, Illinois 60612
| | - Xianzhong Ding
- Division of Endocrinology, Metabolism, and Molecular Medicine (A.E.L., C.M.P., M.P., A.R.A., K.S.H., W.L.L., B.T.L.), Department of Pathology (A.L., G.-Y.Y.), Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; Department of Biostatistics and Bioinformatics (T.E.R.), and Center for Genomic and Computational Biology (C.G., T.E.R.), Duke University Medical School, and University Program in Genetics and Genomics (C.G.), Duke University, Durham, North Carolina 27710; Department of Pathology (X.D.), Loyola University Chicago, Maywood, Illinois 60153; and Jesse Brown Veterans Affairs Medical Center (B.T.L.), Chicago, Illinois 60612
| | - Timothy E Reddy
- Division of Endocrinology, Metabolism, and Molecular Medicine (A.E.L., C.M.P., M.P., A.R.A., K.S.H., W.L.L., B.T.L.), Department of Pathology (A.L., G.-Y.Y.), Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; Department of Biostatistics and Bioinformatics (T.E.R.), and Center for Genomic and Computational Biology (C.G., T.E.R.), Duke University Medical School, and University Program in Genetics and Genomics (C.G.), Duke University, Durham, North Carolina 27710; Department of Pathology (X.D.), Loyola University Chicago, Maywood, Illinois 60153; and Jesse Brown Veterans Affairs Medical Center (B.T.L.), Chicago, Illinois 60612
| | - William L Lowe
- Division of Endocrinology, Metabolism, and Molecular Medicine (A.E.L., C.M.P., M.P., A.R.A., K.S.H., W.L.L., B.T.L.), Department of Pathology (A.L., G.-Y.Y.), Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; Department of Biostatistics and Bioinformatics (T.E.R.), and Center for Genomic and Computational Biology (C.G., T.E.R.), Duke University Medical School, and University Program in Genetics and Genomics (C.G.), Duke University, Durham, North Carolina 27710; Department of Pathology (X.D.), Loyola University Chicago, Maywood, Illinois 60153; and Jesse Brown Veterans Affairs Medical Center (B.T.L.), Chicago, Illinois 60612
| | - Brian T Layden
- Division of Endocrinology, Metabolism, and Molecular Medicine (A.E.L., C.M.P., M.P., A.R.A., K.S.H., W.L.L., B.T.L.), Department of Pathology (A.L., G.-Y.Y.), Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; Department of Biostatistics and Bioinformatics (T.E.R.), and Center for Genomic and Computational Biology (C.G., T.E.R.), Duke University Medical School, and University Program in Genetics and Genomics (C.G.), Duke University, Durham, North Carolina 27710; Department of Pathology (X.D.), Loyola University Chicago, Maywood, Illinois 60153; and Jesse Brown Veterans Affairs Medical Center (B.T.L.), Chicago, Illinois 60612
| |
Collapse
|
48
|
Zhang Z, Huang S, Wang H, Wu J, Chen D, Peng B, Zhou Q. High expression of hexokinase domain containing 1 is associated with poor prognosis and aggressive phenotype in hepatocarcinoma. Biochem Biophys Res Commun 2016; 474:673-679. [PMID: 27155152 DOI: 10.1016/j.bbrc.2016.05.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 05/01/2016] [Indexed: 12/17/2022]
Abstract
Rapid progress and metastasis remain the major treatment failure modes of hepatocarcinoma (HCC). Unfortunately, the underlying molecular mechanisms of hepatoma cell proliferation and migration are poorly understood. Metabolic abnormalities play critical roles in tumorigenesis and progression. Hexokinase domain containing 1 (HKDC1) catalyzes the phosphorylation of glucose. However, the functions and mechanisms of HKDC1 in cancer remain unknown. In this study, real-time RT-PCR and Western blotting assays were used to detect the HKDC1 expression levels in HCC tissues and cell lines. The Oncomine™ Cancer Microarray Database was applied to analysis the correlations between HKDC1 expression and HCC clinical characteristics. MTT and Transwell migration assays were performed to determine the functions of HKDC1 in HCC cells. The effect of HKDC1 on Wnt/β-catenin signaling pathway was assessed using Western blotting assay. In this study, we found that HKDC1 expression levels were elevated in HCC tissues compared with the adjacent tissues. HCC patients with high expression levels of HKDC1 had poor overall survival (OS). Furthermore, higher HKDC1 levels also predicted a worse OS of patients within solitary, elevated pre-operated serum alpha fetoprotein (AFP) level and higher tumor diameter. Moreover, silencing HKDC1 suppressed HCC cells proliferation and migration in vitro. Downregulated HKDC1 expression repressed β-Catenin and c-Myc expression, which indicates that silencing HKDC1 may reduce proliferation and migration via inhibiting the Wnt/β-catenin signaling pathway in HCC. In summary, HKDC1 provides further insight into HCC tumor progression and may provide a novel prognostic biomarker and therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Zijian Zhang
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Shanzhou Huang
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Huanyu Wang
- Department of Thyroid and Breast Surgery, Nanshan District People's Hospital, Shenzhen, 518000, China
| | - Jian Wu
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Dong Chen
- Department of Biliopancreatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Baogang Peng
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Qi Zhou
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
49
|
Guo C, Ludvik AE, Arlotto ME, Hayes MG, Armstrong LL, Scholtens DM, Brown CD, Newgard CB, Becker TC, Layden BT, Lowe WL, Reddy TE. Coordinated regulatory variation associated with gestational hyperglycaemia regulates expression of the novel hexokinase HKDC1. Nat Commun 2015; 6:6069. [PMID: 25648650 PMCID: PMC4318120 DOI: 10.1038/ncomms7069] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 12/10/2014] [Indexed: 02/06/2023] Open
Abstract
Maternal glucose levels during pregnancy impact the developing fetus, affecting metabolic health early and later in life. Both genetic and environmental factors influence maternal metabolism, but little is known about the genetic mechanisms that alter glucose metabolism during pregnancy. Here we report that haplotypes previously associated with gestational hyperglycemia in the third trimester disrupt regulatory element activity and reduce expression of the nearby HKDC1 gene. We further find that experimentally reducing or increasing HKDC1 expression reduces or increases hexokinase activity, respectively, in multiple cellular models; and that purified HKDC1 protein has hexokinase activity in vitro. Together, these results suggest a novel mechanism of gestational glucose regulation in which the effects of genetic variants in multiple regulatory elements alter glucose homeostasis by coordinately reducing expression of the novel hexokinase HKDC1.
Collapse
Affiliation(s)
- Cong Guo
- 1] Duke University Program in Genetics &Genomics, Durham, North Carolina 27708, USA [2] Center for Genomic &Computational Biology, Duke University School of Medicine, Durham, North Carolina 27708, USA
| | - Anton E Ludvik
- Division of Endocrinology, Metabolism &Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Michelle E Arlotto
- Sarah W. Stedman Nutrition &Metabolism Center, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - M Geoffrey Hayes
- Division of Endocrinology, Metabolism &Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Loren L Armstrong
- Division of Endocrinology, Metabolism &Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Denise M Scholtens
- Department of Preventive Medicine, Division of Biostatistics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Christopher D Brown
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Christopher B Newgard
- 1] Sarah W. Stedman Nutrition &Metabolism Center, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina 27710, USA [2] Department of Pharmacology &Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA [3] Division of Endocrinology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Thomas C Becker
- 1] Sarah W. Stedman Nutrition &Metabolism Center, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina 27710, USA [2] Division of Endocrinology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Brian T Layden
- 1] Division of Endocrinology, Metabolism &Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA [2] Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612, USA
| | - William L Lowe
- Division of Endocrinology, Metabolism &Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Timothy E Reddy
- 1] Center for Genomic &Computational Biology, Duke University School of Medicine, Durham, North Carolina 27708, USA [2] Department of Biostatistics &Bioinformatics, Duke University Medical School, Durham, North Carolina 27710, USA
| |
Collapse
|
50
|
Angueira AR, Ludvik AE, Reddy TE, Wicksteed B, Lowe WL, Layden BT. New insights into gestational glucose metabolism: lessons learned from 21st century approaches. Diabetes 2015; 64:327-34. [PMID: 25614666 PMCID: PMC4876793 DOI: 10.2337/db14-0877] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pregnancy presents a unique physiological challenge that requires changes coordinated by placentally and non-placentally derived hormones to prepare the mother for the metabolic stress presented by fetal development and to ensure appropriate nutrient allocation between mother and fetus. Of particular importance is the maintenance of normal glucose metabolism during pregnancy. Here, we describe physiological changes in glucose metabolism during pregnancy and highlight new insights into these adaptations that have emerged over the past decade using novel methodologies, specifically genome-wide association studies (GWAS) and metabolomics. While GWAS have identified some novel associations with metabolic traits during pregnancy, the majority of the findings overlap with those observed in nonpregnant populations and individuals with type 2 diabetes (T2D). Metabolomics studies have provided new insight into key metabolites involved in gestational diabetes mellitus (GDM). Both of these approaches have suggested that a strong link exists between GDM and T2D. Most recently, a role of the gut microbiome in pregnancy has been observed, with changes in the microbiome during the third trimester having metabolic consequences for the mother. In this Perspectives in Diabetes article, we highlight how these new data have broadened our understanding of gestational metabolism, and emphasize the importance of future studies to elucidate differences between GDM and T2D.
Collapse
Affiliation(s)
- Anthony R Angueira
- Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Anton E Ludvik
- Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Timothy E Reddy
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC Institute of Genome Science and Policy, Duke University, Durham, NC
| | | | - William L Lowe
- Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Brian T Layden
- Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL Jesse Brown Veterans Affairs Medical Center, Chicago, IL
| |
Collapse
|