1
|
An S, Bi H, Luo X, Zhu C, Wang M, Pang A, Cui Y. Identification of key genes of diabetic cardiomyopathy in hiPSCs-CMs based on bioinformatics analysis. Mol Cell Biochem 2024; 479:3447-3458. [PMID: 38381273 DOI: 10.1007/s11010-023-04915-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/06/2023] [Indexed: 02/22/2024]
Abstract
Diabetic cardiomyopathy (DbCM) is one of the most common vascular complications of diabetes, and can cause heart failure and threaten the life of patients. The pathogenesis is complex, and key genes have not fully identified. In this study, bioinformatics analysis was used to predict DbCM-related gene targets. Published datasets from the NCBI Gene Expression Omnibus with accession numbers GSE62203 and GSE197850 were selected for analysis. Differentially expressed genes (DEGs) were identified by the online tool GEO2R. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed using the DAVID online database. Protein-protein interaction network construction and hub gene identification were performed using STRING and Cytoscape. We used 30 mM and 1 μM hydrocortisone-stimulated AC16 cells as an in vitro model of diabetic cardiomyopathy. Quantitative real-time PCR (qRT-PCR) was performed to validate the expression levels of hub genes. A total of 73 common DEGs were identified in both datasets, including 47 upregulated and 26 downregulated genes. GO and KEGG pathway enrichment analyses revealed that the DEGs were significantly enriched in metabolism, hypoxia response, apoptosis, cell proliferation regulation, and cytoplasmic and HIF signalling pathways. The top 10 hub genes were LDHA, PGK1, SLC2A1, ENO1, PFKFB3, EGLN1, MYC, PDK1, EGLN3 and BNIP3. In our in vitro study, we found that PGK1, SLC2A1, PFKFB3, EGLN1, MYC, EGLN3 and BNIP3 were upregulated, ENO1 was downregulated, and LDHA was unchanged. Except for PGK1 and ENO1, these hub genes have been previously reported to be involved in DbCM. In summary, we identified DEGs and hub genes and first reported PGK1 and ENO1 in DbCM, which may serve as potential candidate genes for DbCM targeted therapy.
Collapse
Affiliation(s)
- Shuo An
- School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, China
- Department of Clinical Laboratory, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, 300060, China
| | - Hongchen Bi
- School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, China
| | - Xiaoli Luo
- School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, China
| | - Caiying Zhu
- State Key Laboratory of Experimental Hematology, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Min Wang
- School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, China
| | - Aiming Pang
- State Key Laboratory of Experimental Hematology, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
| | - Yujie Cui
- School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, China.
| |
Collapse
|
2
|
Ahmad F, Sachdeva P, Sachdeva B, Singh G, Soni H, Tandon S, Rafeeq MM, Alam MZ, Baeissa HM, Khalid M. Dioxinodehydroeckol: A Potential Neuroprotective Marine Compound Identified by In Silico Screening for the Treatment and Management of Multiple Brain Disorders. Mol Biotechnol 2024; 66:663-686. [PMID: 36513873 DOI: 10.1007/s12033-022-00629-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022]
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD), Glioblastoma multiforme (GBM), Amyotrophic lateral sclerosis (ALS), and Parkinson's disease (PD) are some of the most prevalent neurodegenerative disorders in humans. Even after a variety of advanced therapies, prognosis of all these disorders is not favorable, with survival rates of 14-20 months only. To further improve the prognosis of these disorders, it is imperative to discover new compounds which will target effector proteins involved in these disorders. In this study, we have focused on in silico screening of marine compounds against multiple target proteins involved in AD, GBM, ALS, and PD. Fifty marine-origin compounds were selected from literature, out of which, thirty compounds passed ADMET parameters. Ligand docking was performed after ADMET analysis for AD, GBM, ALS, and PD-associated proteins in which four protein targets Keap1, Ephrin A2, JAK3 Kinase domain, and METTL3-METTL14 N6-methyladenosine methyltransferase (MTA70) were found to be binding strongly with the screened compound Dioxinodehydroeckol (DHE). Molecular dynamics simulations were performed at 100 ns with triplicate runs to validate the docking score and assess the dynamics of DHE interactions with each target protein. The results indicated Dioxinodehydroeckol, a novel marine compound, to be a putative inhibitor among all the screened molecules, which might be effective against multiple target proteins involved in neurological disorders, requiring further in vitro and in vivo validations.
Collapse
Affiliation(s)
- Faizan Ahmad
- Department of Medical Elementology and Toxicology, Jamia Hamdard University, Delhi, India.
| | - Punya Sachdeva
- Amity Institute of Neuropsychology and Neurosciences, Amity University, Noida, Uttar Pradesh, India
| | - Bhuvi Sachdeva
- Department of Physics and Astrophysics, University of Delhi, Delhi, India
| | - Gagandeep Singh
- Section of Microbiology, Central Ayurveda Research Institute, CCRAS, Ministry of AYUSH, Jhansi, India
- Kusuma School of Biological Sciences, India Institute of Technology, Delhi, India
| | - Hemant Soni
- Section of Microbiology, Central Ayurveda Research Institute, CCRAS, Ministry of AYUSH, Jhansi, India
| | - Smriti Tandon
- Section of Microbiology, Central Ayurveda Research Institute, CCRAS, Ministry of AYUSH, Jhansi, India
| | - Misbahuddin M Rafeeq
- Department of Pharmacology, Faculty of Medicine, Rabigh, King Abdulaziz University, Jeddah, 21589, Kingdom of Saudi Arabia
| | - Mohammad Zubair Alam
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hanadi M Baeissa
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj, 11942, Saudi Arabia
| |
Collapse
|
3
|
Orellana AM, Mazucanti CH, Dos Anjos LP, de Sá Lima L, Kawamoto EM, Scavone C. Klotho increases antioxidant defenses in astrocytes and ubiquitin-proteasome activity in neurons. Sci Rep 2023; 13:15080. [PMID: 37699938 PMCID: PMC10497516 DOI: 10.1038/s41598-023-41166-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/23/2023] [Indexed: 09/14/2023] Open
Abstract
Klotho is an antiaging protein, and its levels decline with age and chronic stress. The exogenous administration of Klotho can enhance cognitive performance in mice and negatively modulate the Insulin/IGF1/PI3K/AKT pathway in terms of metabolism. In humans, insulin sensitivity is a hallmark of healthy longevity. Therefore, this study aimed to determine if exogenous Klotho, when added to neuronal and astrocytic cell cultures, could reduce the phosphorylation levels of certain insulin signaling effectors and enhance antioxidant strategies in these cells. Primary cell cultures of cortical astrocytes and neurons from mice were exposed to 1 nM Klotho for 24 h, with or without glucose. Klotho decreased pAKT and mTOR levels. However, in astrocytes, Klotho increased FOXO-3a activity and catalase levels, shielding them from intermediate oxidative stress. In neurons, Klotho did not alter FOXO-3 phosphorylation levels but increased proteasome activity, maintaining lower levels of PFKFB3. This study offers new insights into the roles of Klotho in regulating energy metabolism and the redox state in the brain.
Collapse
Affiliation(s)
- Ana Maria Orellana
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences ICB-1, University of São Paulo, Avenida Professor Lineu Prestes, 1524, São Paulo, São Paulo, 05508-900, Brazil
| | - Caio Henrique Mazucanti
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences ICB-1, University of São Paulo, Avenida Professor Lineu Prestes, 1524, São Paulo, São Paulo, 05508-900, Brazil
- Laboratory of Clinical Investigation, Diabetes Section, National Institute on Aging (NIH/NIA), Baltimore, MD, USA
| | - Leticia Pavan Dos Anjos
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences ICB-1, University of São Paulo, Avenida Professor Lineu Prestes, 1524, São Paulo, São Paulo, 05508-900, Brazil
| | - Larissa de Sá Lima
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences ICB-1, University of São Paulo, Avenida Professor Lineu Prestes, 1524, São Paulo, São Paulo, 05508-900, Brazil
| | - Elisa Mitiko Kawamoto
- Laboratory of Molecular and Functional Neurobiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Cristoforo Scavone
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences ICB-1, University of São Paulo, Avenida Professor Lineu Prestes, 1524, São Paulo, São Paulo, 05508-900, Brazil.
| |
Collapse
|
4
|
Sneyers F, Kerkhofs M, Speelman-Rooms F, Welkenhuyzen K, La Rovere R, Shemy A, Voet A, Eelen G, Dewerchin M, Tait SWG, Ghesquière B, Bootman MD, Bultynck G. Intracellular BAPTA directly inhibits PFKFB3, thereby impeding mTORC1-driven Mcl-1 translation and killing MCL-1-addicted cancer cells. Cell Death Dis 2023; 14:600. [PMID: 37684238 PMCID: PMC10491774 DOI: 10.1038/s41419-023-06120-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 08/10/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023]
Abstract
Intracellular Ca2+ signals control several physiological and pathophysiological processes. The main tool to chelate intracellular Ca2+ is intracellular BAPTA (BAPTAi), usually introduced into cells as a membrane-permeant acetoxymethyl ester (BAPTA-AM). Previously, we demonstrated that BAPTAi enhanced apoptosis induced by venetoclax, a BCL-2 antagonist, in diffuse large B-cell lymphoma (DLBCL). This finding implied a novel interplay between intracellular Ca2+ signaling and anti-apoptotic BCL-2 function. Hence, we set out to identify the underlying mechanisms by which BAPTAi enhances cell death in B-cell cancers. In this study, we discovered that BAPTAi alone induced apoptosis in hematological cancer cell lines that were highly sensitive to S63845, an MCL-1 antagonist. BAPTAi provoked a rapid decline in MCL-1-protein levels by inhibiting mTORC1-driven Mcl-1 translation. These events were not a consequence of cell death, as BAX/BAK-deficient cancer cells exhibited similar downregulation of mTORC1 activity and MCL-1-protein levels. Next, we investigated how BAPTAi diminished mTORC1 activity and identified its ability to impair glycolysis by directly inhibiting 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) activity, a previously unknown effect of BAPTAi. Notably, these effects were also induced by a BAPTAi analog with low affinity for Ca2+. Consequently, our findings uncover PFKFB3 inhibition as an Ca2+-independent mechanism through which BAPTAi impairs cellular metabolism and ultimately compromises the survival of MCL-1-dependent cancer cells. These findings hold two important implications. Firstly, the direct inhibition of PFKFB3 emerges as a key regulator of mTORC1 activity and a promising target in MCL-1-dependent cancers. Secondly, cellular effects caused by BAPTAi are not necessarily related to Ca2+ signaling. Our data support the need for a reassessment of the role of Ca2+ in cellular processes when findings were based on the use of BAPTAi.
Collapse
Affiliation(s)
- Flore Sneyers
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O&N I, Herestraat 49 box 802, 3000, Leuven, Belgium
| | - Martijn Kerkhofs
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O&N I, Herestraat 49 box 802, 3000, Leuven, Belgium
| | - Femke Speelman-Rooms
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O&N I, Herestraat 49 box 802, 3000, Leuven, Belgium
- KU Leuven, Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O&N I bis, Herestraat 49 box 901, 3000, Leuven, Belgium
| | - Kirsten Welkenhuyzen
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O&N I, Herestraat 49 box 802, 3000, Leuven, Belgium
| | - Rita La Rovere
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O&N I, Herestraat 49 box 802, 3000, Leuven, Belgium
| | - Ahmed Shemy
- KU Leuven, Laboratory for Biomolecular Modelling and Design, Department of Chemistry, Celestijnenlaan 200G, 3001, Heverlee, Belgium
| | - Arnout Voet
- KU Leuven, Laboratory for Biomolecular Modelling and Design, Department of Chemistry, Celestijnenlaan 200G, 3001, Heverlee, Belgium
| | - Guy Eelen
- KU Leuven, Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Leuven Cancer Institute, Campus Gasthuisberg O&N4, Herestraat 49 box 912, Leuven, Belgium
- VIB-KU Leuven, Center for Cancer Biology, Laboratory of Angiogenesis and Vascular Metabolism, Campus Gasthuisberg O&N4, Herestraat 49 box 912, 3000, Leuven, Belgium
| | - Mieke Dewerchin
- KU Leuven, Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Leuven Cancer Institute, Campus Gasthuisberg O&N4, Herestraat 49 box 912, Leuven, Belgium
- VIB-KU Leuven, Center for Cancer Biology, Laboratory of Angiogenesis and Vascular Metabolism, Campus Gasthuisberg O&N4, Herestraat 49 box 912, 3000, Leuven, Belgium
| | - Stephen W G Tait
- Cancer Research UK Beatson Institute, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Bart Ghesquière
- KU Leuven, Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, Leuven, Belgium - VIB, Metabolomics Core Facility Leuven, Center for Cancer Biology, Leuven, Belgium, Herestraat 49 box 912, 3000, Leuven, Belgium
| | - Martin D Bootman
- School of Life, Health and Chemical Sciences, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK.
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O&N I, Herestraat 49 box 802, 3000, Leuven, Belgium.
| |
Collapse
|
5
|
Cheng X, Jia X, Wang C, Zhou S, Chen J, Chen L, Chen J. Hyperglycemia induces PFKFB3 overexpression and promotes malignant phenotype of breast cancer through RAS/MAPK activation. World J Surg Oncol 2023; 21:112. [PMID: 36973739 PMCID: PMC10044395 DOI: 10.1186/s12957-023-02990-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/18/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Breast cancer is the most common tumor in women worldwide. Diabetes mellitus is a global chronic metabolic disease with increasing incidence. Diabetes mellitus has been reported to positively regulate the development of many tumors. However, the specific mechanism of hyperglycemic environment regulating breast cancer remains unclear. PFKFB3 (6-phosphofructose-2-kinase/fructose-2, 6-bisphosphatase 3) is a key regulatory factor of the glycolysis process in diabetes mellitus, as well as a promoter of breast cancer. So, we want to explore the potential link between PFKFB3 and the poor prognosis of breast cancer patients with hyperglycemia in this study. METHODS Cell culture was utilized to construct different-glucose breast cancer cell lines. Immunohistochemistry was adopted to analyze the protein level of PFKFB3 in benign breast tissues, invasive ductal carcinoma with diabetes and invasive ductal carcinoma without diabetes. The Kaplan-Meier plotter database and GEO database (GSE61304) was adopted to analyze the survival of breast cancer patients with different PFKFB3 expression. Western blot was adopted to analyze the protein level of PFKFB3, epithelial-mesenchymal transition (EMT)-related protein and extracellular regulated protein kinases (ERK) in breast cancer cells. Gene Set Cancer Analysis (GSCA) was utilized to investigate the potential downstream signaling pathways of PFKFB3. TargetScan and OncomiR were utilized to explore the potential mechanism of PFKFB3 overexpression by hyperglycemia. Transfections (including siRNAs and miRNA transfection premiers) was utilized to restrain or mimic the expression of the corresponding RNA. Cell functional assays (including cell counting, MTT, colony formation, wound-healing, and cell migration assays) were utilized to explore the proliferation and migration of breast cancer cells. RESULTS In this study, we demonstrated that the expression of PFKFB3 in breast cancer complicated with hyperglycemia was higher than that in breast cancer with euglycemia through cell experiment in vitro and histological experiment. PFKFB3 overexpression decreased the survival period of breast cancer patients and was correlated with a number of clinicopathological parameters of breast cancer complicated with diabetes. PFKFB3 promoted the proliferation and migration of breast cancer in a hyperglycemic environment and might be regulated by miR-26. In addition, PFKFB3 stimulated epithelial-mesenchymal transition of breast cancer in a hyperglycemic environment. In terms of downstream mechanism exploration, we predicted and verified the cancer-promoting effect of PFKFB3 in breast cancer complicated with hyperglycemia through RAS/MAPK pathway. CONCLUSIONS In conclusion, PFKFB3 could be overexpressed by hyperglycemia and might be a potential therapeutic target for breast cancer complicated with diabetes.
Collapse
Affiliation(s)
- Xiao Cheng
- Department of Histopathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, 315000, Zhejiang, China
| | - Xiupeng Jia
- Department of Histopathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, 315000, Zhejiang, China
| | - Chunnian Wang
- Department of Histopathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, 315000, Zhejiang, China
| | - Shangyan Zhou
- Department of Histopathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, 315000, Zhejiang, China
| | - Jiayi Chen
- Department of Experimental Pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, 315000, Zhejiang, China
| | - Lei Chen
- Department of Cytopathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, 315000, Zhejiang, China
| | - Jinping Chen
- Department of Histopathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, 315000, Zhejiang, China.
| |
Collapse
|
6
|
Phosphohistidine signaling promotes FAK-RB1 interaction and growth factor-independent proliferation of esophageal squamous cell carcinoma. Oncogene 2023; 42:449-460. [PMID: 36513743 DOI: 10.1038/s41388-022-02568-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 11/08/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
Current clinical therapies targeting receptor tyrosine kinases including focal adhesion kinase (FAK) have had limited or no effect on esophageal squamous cell carcinoma (ESCC). Unlike esophageal adenocarcinomas, ESCC acquire glucose in excess of their anabolic need. We recently reported that glucose-induced growth factor-independent proliferation requires the phosphorylation of FAKHis58. Here, we confirm His58 phosphorylation in FAK immunoprecipitates of glucose-stimulated, serum-starved ESCC cells using antibodies specific for 3-phosphohistidine and mass spectrometry. We also confirm a role for the histidine kinase, NME1, in glucose-induced FAKpoHis58 and ESCC cell proliferation, correlating with increased levels of NME1 in ESCC tumors versus normal esophageal tissues. Unbiased screening identified glucose-induced retinoblastoma transcriptional corepressor 1 (RB1) binding to FAK, mediated through a "LxCxE" RB1-binding motif in FAK's FERM domain. Importantly, in the absence of growth factors, glucose increased FAK scaffolding of RB1 in the cytoplasm, correlating with increased ESCC G1→S phase transition. Our data strongly suggest that this glucose-mediated mitogenic pathway is novel and represents a unique targetable opportunity in ESCC.
Collapse
|
7
|
Kuzniak-Glanowska E, Glanowski M, Kurczab R, Bojarski AJ, Podgajny R. Mining anion-aromatic interactions in the Protein Data Bank. Chem Sci 2022; 13:3984-3998. [PMID: 35440982 PMCID: PMC8985504 DOI: 10.1039/d2sc00763k] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 02/28/2022] [Indexed: 12/01/2022] Open
Abstract
Mutual positioning and non-covalent interactions in anion–aromatic motifs are crucial for functional performance of biological systems. In this context, regular, comprehensive Protein Data Bank (PDB) screening that involves various scientific points of view and individual critical analysis is of utmost importance. Analysis of anions in spheres with radii of 5 Å around all 5- and 6-membered aromatic rings allowed us to distinguish 555 259 unique anion–aromatic motifs, including 92 660 structures out of the 171 588 structural files in the PDB. The use of a scarcely exploited (x, h) coordinate system led to (i) identification of three separate areas of motif accumulation: A – over the ring, B – over the ring-substituent bonds, and C – roughly in the plane of the aromatic ring, and (ii) unprecedented simultaneous comparative description of various anion–aromatic motifs located in these areas. Of the various residues considered, i.e. aminoacids, nucleotides, and ligands, the latter two exhibited a considerable tendency to locate in region Avia archetypal anion–π contacts. The applied model not only enabled statistical quantitative analysis of space around the ring, but also enabled discussion of local intermolecular arrangements, as well as detailed sequence and secondary structure analysis, e.g. anion–π interactions in the GNRA tetraloop in RNA and protein helical structures. As a purely practical issue of this work, the new code source for the PDB research was produced, tested and made freely available at https://github.com/chemiczny/PDB_supramolecular_search. The comprehensive analysis of non-redundant PDB macromolecular structures investigating anion distributions around all aromatic molecules in available biosystems is presented.![]()
Collapse
Affiliation(s)
| | - Michał Glanowski
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences Niezapominajek 8 30-239 Kraków Poland
| | - Rafał Kurczab
- Maj Institute of Pharmacology, Polish Academy of Sciences Smętna 12 31-343 Kraków Poland
| | - Andrzej J Bojarski
- Maj Institute of Pharmacology, Polish Academy of Sciences Smętna 12 31-343 Kraków Poland
| | - Robert Podgajny
- Faculty of Chemistry, Jagiellonian University Gronostajowa 2 30-387 Kraków Poland
| |
Collapse
|
8
|
Liu X, Li J, Wang Z, Meng J, Wang A, Zhao X, Xu Q, Cai Z, Hu Z. KDM2A Targets PFKFB3 for Ubiquitylation to Inhibit the Proliferation and Angiogenesis of Multiple Myeloma Cells. Front Oncol 2021; 11:653788. [PMID: 34079757 PMCID: PMC8165180 DOI: 10.3389/fonc.2021.653788] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/15/2021] [Indexed: 12/17/2022] Open
Abstract
The lysine demethylase KDM2A (also known as JHDM1A or FBXL11) demethylates histone H3 at lysine K36 which lead to epigenetic regulation of cell proliferation and tumorigenesis. However, many biological processes are mediated by KDM2A independently by its histone demethylation activity. In the present study, we aimed to characterize the functional significance of KDM2A in multiple myeloma (MM) disease progression. Specifically, we defined that one of the key enzymes of glycolysis PFKFB3 (6-phosphofructo-2-kinase) is ubiquitylated by KDM2A which suppresses MM cell proliferation. Previous study showed that KDM2A and PFKFB3 promoted angiogenesis in various tumor cells. We further reveal that KDM2A targets PFKFB3 for ubiquitination and degradation to inhibit angiogenesis. Several angiogenic cytokines are also downregulated in MM. Clinically, MM patients with low KDM2A and high PFKFB3 levels have shown worse prognosis. These results reveal a novel function of KDM2A through ubiquitin ligase activity by targeting PFKFB3 to induce proliferation, glycolysis and angiogenesis in MM cells. The data provides a new potential mechanism and strategy for MM treatment.
Collapse
Affiliation(s)
- Xinling Liu
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jiaqiu Li
- Department of Oncology, Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Zhanju Wang
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jie Meng
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Aihong Wang
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiaofei Zhao
- Department of Dermatology, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Qilu Xu
- Department of Hematology, The First Affiliated Hospital, Weifang Medical University, Weifang, China
| | - Zhen Cai
- Department of Hematology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhenbo Hu
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
9
|
Kotowski K, Rosik J, Machaj F, Supplitt S, Wiczew D, Jabłońska K, Wiechec E, Ghavami S, Dzięgiel P. Role of PFKFB3 and PFKFB4 in Cancer: Genetic Basis, Impact on Disease Development/Progression, and Potential as Therapeutic Targets. Cancers (Basel) 2021; 13:909. [PMID: 33671514 PMCID: PMC7926708 DOI: 10.3390/cancers13040909] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 12/11/2022] Open
Abstract
Glycolysis is a crucial metabolic process in rapidly proliferating cells such as cancer cells. Phosphofructokinase-1 (PFK-1) is a key rate-limiting enzyme of glycolysis. Its efficiency is allosterically regulated by numerous substances occurring in the cytoplasm. However, the most potent regulator of PFK-1 is fructose-2,6-bisphosphate (F-2,6-BP), the level of which is strongly associated with 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase activity (PFK-2/FBPase-2, PFKFB). PFK-2/FBPase-2 is a bifunctional enzyme responsible for F-2,6-BP synthesis and degradation. Four isozymes of PFKFB (PFKFB1, PFKFB2, PFKFB3, and PFKFB4) have been identified. Alterations in the levels of all PFK-2/FBPase-2 isozymes have been reported in different diseases. However, most recent studies have focused on an increased expression of PFKFB3 and PFKFB4 in cancer tissues and their role in carcinogenesis. In this review, we summarize our current knowledge on all PFKFB genes and protein structures, and emphasize important differences between the isoenzymes, which likely affect their kinase/phosphatase activities. The main focus is on the latest reports in this field of cancer research, and in particular the impact of PFKFB3 and PFKFB4 on tumor progression, metastasis, angiogenesis, and autophagy. We also present the most recent achievements in the development of new drugs targeting these isozymes. Finally, we discuss potential combination therapies using PFKFB3 inhibitors, which may represent important future cancer treatment options.
Collapse
Affiliation(s)
- Krzysztof Kotowski
- Department of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (K.K.); (K.J.)
| | - Jakub Rosik
- Department of Pathology, Pomeranian Medical University, 71-252 Szczecin, Poland; (J.R.); (F.M.)
| | - Filip Machaj
- Department of Pathology, Pomeranian Medical University, 71-252 Szczecin, Poland; (J.R.); (F.M.)
| | - Stanisław Supplitt
- Department of Genetics, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Daniel Wiczew
- Department of Biochemical Engineering, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland;
- Laboratoire de physique et chimie théoriques, Université de Lorraine, F-54000 Nancy, France
| | - Karolina Jabłońska
- Department of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (K.K.); (K.J.)
| | - Emilia Wiechec
- Department of Biomedical and Clinical Sciences (BKV), Division of Cell Biology, Linköping University, Region Östergötland, 581 85 Linköping, Sweden;
- Department of Otorhinolaryngology in Linköping, Anesthetics, Operations and Specialty Surgery Center, Region Östergötland, 581 85 Linköping, Sweden
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Research Institute in Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Piotr Dzięgiel
- Department of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (K.K.); (K.J.)
- Department of Physiotherapy, Wroclaw University School of Physical Education, 51-612 Wroclaw, Poland
| |
Collapse
|
10
|
Richardson DA, Sritangos P, James AD, Sultan A, Bruce JIE. Metabolic regulation of calcium pumps in pancreatic cancer: role of phosphofructokinase-fructose-bisphosphatase-3 (PFKFB3). Cancer Metab 2020; 8:2. [PMID: 32266066 PMCID: PMC7114799 DOI: 10.1186/s40170-020-0210-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 01/12/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND High glycolytic rate is a hallmark of cancer (Warburg effect). Glycolytic ATP is required for fuelling plasma membrane calcium ATPases (PMCAs), responsible for extrusion of cytosolic calcium, in pancreatic ductal adenocarcinoma (PDAC). Phosphofructokinase-fructose-bisphosphatase-3 (PFKFB3) is a glycolytic driver that activates key rate-limiting enzyme Phosphofructokinase-1; we investigated whether PFKFB3 is required for PMCA function in PDAC cells. METHODS PDAC cell-lines, MIA PaCa-2, BxPC-3, PANC1 and non-cancerous human pancreatic stellate cells (HPSCs) were used. Cell growth, death and metabolism were assessed using sulforhodamine-B/tetrazolium-based assays, poly-ADP-ribose-polymerase (PARP1) cleavage and seahorse XF analysis, respectively. ATP was measured using a luciferase-based assay, membrane proteins were isolated using a kit and intracellular calcium concentration and PMCA activity were measured using Fura-2 fluorescence imaging. RESULTS PFKFB3 was highly expressed in PDAC cells but not HPSCs. In MIA PaCa-2, a pool of PFKFB3 was identified at the plasma membrane. PFKFB3 inhibitor, PFK15, caused reduced cell growth and PMCA activity, leading to calcium overload and apoptosis in PDAC cells. PFK15 reduced glycolysis but had no effect on steady-state ATP concentration in MIA PaCa-2. CONCLUSIONS PFKFB3 is important for maintaining PMCA function in PDAC, independently of cytosolic ATP levels and may be involved in providing a localised ATP supply at the plasma membrane.
Collapse
Affiliation(s)
- D. A. Richardson
- Division of Cancer Sciences, School of Medical Sciences, University Of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT UK
| | - P. Sritangos
- Division of Cancer Sciences, School of Medical Sciences, University Of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT UK
| | - A. D. James
- Department of Biology, University of York, Heslington, York, UK
| | - A. Sultan
- Division of Cancer Sciences, School of Medical Sciences, University Of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT UK
| | - J. I. E. Bruce
- Division of Cancer Sciences, School of Medical Sciences, University Of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT UK
| |
Collapse
|
11
|
Macut H, Hu X, Tarantino D, Gilardoni E, Clerici F, Regazzoni L, Contini A, Pellegrino S, Luisa Gelmi M. Tuning PFKFB3 Bisphosphatase Activity Through Allosteric Interference. Sci Rep 2019; 9:20333. [PMID: 31889092 PMCID: PMC6937325 DOI: 10.1038/s41598-019-56708-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/28/2019] [Indexed: 12/05/2022] Open
Abstract
The human inducible phospho-fructokinase bisphosphatase isoform 3, PFKFB3, is a crucial regulatory node in the cellular metabolism. The enzyme is an important modulator regulating the intracellular fructose-2,6-bisphosphate level. PFKFB3 is a bifunctional enzyme with an exceptionally high kinase to phosphatase ratio around 740:1. Its kinase activity can be directly inhibited by small molecules acting directly on the kinase active site. On the other hand, here we propose an innovative and indirect strategy for the modulation of PFKFB3 activity, achieved through allosteric bisphosphatase activation. A library of small peptides targeting an allosteric site was discovered and synthesized. The binding affinity was evaluated by microscale thermophoresis (MST). Furthermore, a LC-MS/MS analytical method for assessing the bisphosphatase activity of PFKFB3 was developed. The new method was applied for measuring the activation on bisphosphatase activity with the PFKFB3-binding peptides. The molecular mechanical connection between the newly discovered allosteric site to the bisphosphatase activity was also investigated using both experimental and computational methods.
Collapse
Affiliation(s)
- Helena Macut
- DISFARM- Department of Pharmaceutical sciences, Via Mangiagalli 25, 20133, Milan, Italy
| | - Xiao Hu
- DISFARM- Department of Pharmaceutical sciences, Via Mangiagalli 25, 20133, Milan, Italy
| | - Delia Tarantino
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy
| | - Ettore Gilardoni
- DISFARM- Department of Pharmaceutical sciences, Via Mangiagalli 25, 20133, Milan, Italy
| | - Francesca Clerici
- DISFARM- Department of Pharmaceutical sciences, Via Mangiagalli 25, 20133, Milan, Italy
| | - Luca Regazzoni
- DISFARM- Department of Pharmaceutical sciences, Via Mangiagalli 25, 20133, Milan, Italy
| | - Alessandro Contini
- DISFARM- Department of Pharmaceutical sciences, Via Mangiagalli 25, 20133, Milan, Italy.
| | - Sara Pellegrino
- DISFARM- Department of Pharmaceutical sciences, Via Mangiagalli 25, 20133, Milan, Italy.
| | - Maria Luisa Gelmi
- DISFARM- Department of Pharmaceutical sciences, Via Mangiagalli 25, 20133, Milan, Italy
| |
Collapse
|
12
|
Zhang J, Gelman IH, Katsuta E, Liang Y, Wang X, Li J, Qu J, Yan L, Takabe K, Hochwald SN. Glucose Drives Growth Factor-Independent Esophageal Cancer Proliferation via Phosphohistidine-Focal Adhesion Kinase Signaling. Cell Mol Gastroenterol Hepatol 2019; 8:37-60. [PMID: 30836148 PMCID: PMC6518323 DOI: 10.1016/j.jcmgh.2019.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Most targeted therapies against cancer are designed to block growth factor-stimulated oncogenic growth. However, response rates are low, and resistance to therapy is high. One mechanism might relate to the ability of tumor cells to induce growth factor-independent proliferation (GFIP). This project aims to understand how (1) cancer cells preferentially derive a major growth advantage by using critical metabolic products of glucose, such as phosphoenolpyruvate (PEP), to drive proliferation and (2) esophageal squamous cell carcinoma (ESCC) cells, but not esophageal adenocarcinoma cells, can induce GFIP by using glycolysis to activate phosphohistidine (poHis)-mediated signaling through focal adhesion kinase (FAK). METHODS The hypothesis to be tested is that ESCC GFIP induced by glucose is facilitated by PEP-mediated histidine phosphorylation (poHis) of FAK, leading to the possibility that ESCC progression can be targeted by blocking poHis signaling. Biochemical, molecular biological, and in vivo experiments including bromodeoxyuridine/5-ethynyl-2'-deoxyuridine labeling, radioisotope tracing, CRISPR gene editing, and analysis of signaling gene sets in human cancer tissues and xenograft models were performed to define the mechanisms underlying ESCC GFIP. RESULTS Glucose promotes growth factor-independent DNA replication and accumulation of PEP in ESCC cells. PEP is the direct phospho-donor to poHis58-FAK within a known "HG" motif for histidine phosphorylation. Glucose-induced poHis58 promotes growth factor-independent FAK-mediated proliferation. Furthermore, glucose activates phosphatidylinositol-3'-kinase/AKT via poHis58-FAK signaling. Non-phosphorylatable His58A-FAK reduces xenograft growth. CONCLUSIONS Glucose induces ESCC, but not esophageal adenocarcinoma GFIP via PEP-His58-FAK-AKT signaling. ESCC progression is controlled by actionable growth factor-independent, glucose-induced pathways that regulate proliferation through novel histidine phosphorylation of FAK.
Collapse
Affiliation(s)
- Jianliang Zhang
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Irwin H. Gelman
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Eriko Katsuta
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Yuanzi Liang
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Xue Wang
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Jun Li
- University at Buffalo School of Medicine and Biomedical Sciences, Buffalo, New York
| | - Jun Qu
- University at Buffalo School of Medicine and Biomedical Sciences, Buffalo, New York
| | - Li Yan
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Steven N. Hochwald
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York,Correspondence Address correspondence to: Steven N. Hochwald, MD, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York 14263. fax: (716) 845-1060.
| |
Collapse
|
13
|
Bartrons R, Simon-Molas H, Rodríguez-García A, Castaño E, Navarro-Sabaté À, Manzano A, Martinez-Outschoorn UE. Fructose 2,6-Bisphosphate in Cancer Cell Metabolism. Front Oncol 2018; 8:331. [PMID: 30234009 PMCID: PMC6131595 DOI: 10.3389/fonc.2018.00331] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/01/2018] [Indexed: 01/28/2023] Open
Abstract
For a long time, pioneers in the field of cancer cell metabolism, such as Otto Warburg, have focused on the idea that tumor cells maintain high glycolytic rates even with adequate oxygen supply, in what is known as aerobic glycolysis or the Warburg effect. Recent studies have reported a more complex situation, where the tumor ecosystem plays a more critical role in cancer progression. Cancer cells display extraordinary plasticity in adapting to changes in their tumor microenvironment, developing strategies to survive and proliferate. The proliferation of cancer cells needs a high rate of energy and metabolic substrates for biosynthesis of biomolecules. These requirements are met by the metabolic reprogramming of cancer cells and others present in the tumor microenvironment, which is essential for tumor survival and spread. Metabolic reprogramming involves a complex interplay between oncogenes, tumor suppressors, growth factors and local factors in the tumor microenvironment. These factors can induce overexpression and increased activity of glycolytic isoenzymes and proteins in stromal and cancer cells which are different from those expressed in normal cells. The fructose-6-phosphate/fructose-1,6-bisphosphate cycle, catalyzed by 6-phosphofructo-1-kinase/fructose 1,6-bisphosphatase (PFK1/FBPase1) isoenzymes, plays a key role in controlling glycolytic rates. PFK1/FBpase1 activities are allosterically regulated by fructose-2,6-bisphosphate, the product of the enzymatic activity of the dual kinase/phosphatase family of enzymes: 6-phosphofructo-2-kinase/fructose 2,6-bisphosphatase (PFKFB1-4) and TP53-induced glycolysis and apoptosis regulator (TIGAR), which show increased expression in a significant number of tumor types. In this review, the function of these isoenzymes in the regulation of metabolism, as well as the regulatory factors modulating their expression and activity in the tumor ecosystem are discussed. Targeting these isoenzymes, either directly or by inhibiting their activating factors, could be a promising approach for treating cancers.
Collapse
Affiliation(s)
- Ramon Bartrons
- Unitat de Bioquímica, Departament de Ciències Fisiològiques, Universitat de Barcelona, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Catalunya, Spain
| | - Helga Simon-Molas
- Unitat de Bioquímica, Departament de Ciències Fisiològiques, Universitat de Barcelona, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Catalunya, Spain
| | - Ana Rodríguez-García
- Unitat de Bioquímica, Departament de Ciències Fisiològiques, Universitat de Barcelona, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Catalunya, Spain
| | - Esther Castaño
- Centres Científics i Tecnològics, Universitat de Barcelona, Catalunya, Spain
| | - Àurea Navarro-Sabaté
- Unitat de Bioquímica, Departament de Ciències Fisiològiques, Universitat de Barcelona, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Catalunya, Spain
| | - Anna Manzano
- Unitat de Bioquímica, Departament de Ciències Fisiològiques, Universitat de Barcelona, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Catalunya, Spain
| | | |
Collapse
|
14
|
Bartrons R, Rodríguez-García A, Simon-Molas H, Castaño E, Manzano A, Navarro-Sabaté À. The potential utility of PFKFB3 as a therapeutic target. Expert Opin Ther Targets 2018; 22:659-674. [PMID: 29985086 DOI: 10.1080/14728222.2018.1498082] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
INTRODUCTION It has been known for over half a century that tumors exhibit an increased demand for nutrients to fuel their rapid proliferation. Interest in targeting cancer metabolism to treat the disease has been renewed in recent years with the discovery that many cancer-related pathways have a profound effect on metabolism. Considering the recent increase in our understanding of cancer metabolism and the enzymes and pathways involved, the question arises as to whether metabolism is cancer's Achilles heel. Areas covered: This review summarizes the role of 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) in glycolysis, cell proliferation, and tumor growth, discussing PFKFB3 gene and isoenzyme regulation and the changes that occur in cancer and inflammatory diseases. Pharmacological options currently available for selective PFKFB3 inhibition are also reviewed. Expert opinion: PFKFB3 plays an important role in sustaining the development and progression of cancer and might represent an attractive target for therapeutic strategies. Nevertheless, clinical trials are needed to follow up on the promising results from preclinical studies with PFKFB3 inhibitors. Combination therapies with PFKFB3 inhibitors, chemotherapeutic drugs, or radiotherapy might improve the efficacy of cancer treatments targeting PFKFB3.
Collapse
Affiliation(s)
- Ramon Bartrons
- a Unitat de Bioquímica, Departament de Ciències Fisiològiques , Universitat de Barcelona, IDIBELL , Catalunya , Spain
| | - Ana Rodríguez-García
- a Unitat de Bioquímica, Departament de Ciències Fisiològiques , Universitat de Barcelona, IDIBELL , Catalunya , Spain
| | - Helga Simon-Molas
- a Unitat de Bioquímica, Departament de Ciències Fisiològiques , Universitat de Barcelona, IDIBELL , Catalunya , Spain
| | - Esther Castaño
- a Unitat de Bioquímica, Departament de Ciències Fisiològiques , Universitat de Barcelona, IDIBELL , Catalunya , Spain
| | - Anna Manzano
- a Unitat de Bioquímica, Departament de Ciències Fisiològiques , Universitat de Barcelona, IDIBELL , Catalunya , Spain
| | - Àurea Navarro-Sabaté
- a Unitat de Bioquímica, Departament de Ciències Fisiològiques , Universitat de Barcelona, IDIBELL , Catalunya , Spain
| |
Collapse
|
15
|
Non-canonical roles of PFKFB3 in regulation of cell cycle through binding to CDK4. Oncogene 2018; 37:1685-1698. [PMID: 29335521 DOI: 10.1038/s41388-017-0072-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 10/16/2017] [Accepted: 11/08/2017] [Indexed: 01/06/2023]
Abstract
There is growing interest in studying the molecular mechanisms of crosstalk between cancer metabolism and the cell cycle. 6-phosphate fructose-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3) is a well-known glycolytic activator that plays an important role in tumorigenesis. We investigated whether PFKFB3 was directly involved in oncogenic signaling networks. Mass Spectrometry showed that PFKFB3 interacts with cyclin-dependent kinase (CDK) 4, which controls the transition from G1 phase to S phase of the cell cycle. Further analysis indicated that lysine 147 was a key site for the binding of PFKBFB3 to CDK4. PFKFB3 binding resulted in the accumulation of CDK4 protein by inhibiting ubiquitin proteasome degradation mediated by the heat shock protein 90-Cdc37-CDK4 complex. The proteasome-dependent degradation of CDK4 was accelerated by disrupting the interaction of PFKFB3 with CDK4 by mutating lysine (147) to alanine. Blocking PFKFB3-CDK4 interaction improved the therapeutic effect of FDA-approved CDK4 inhibitor palbociclib on breast cancer. These findings suggest that PFKFB3 is a hub for coordinating cell cycle and glucose metabolism. Combined targeting of PFKFB3 and CDK4 may be new strategy for breast cancer treatment.
Collapse
|
16
|
Zhao YD, Yin L, Archer S, Lu C, Zhao G, Yao Y, Wu L, Hsin M, Waddell TK, Keshavjee S, Granton J, de Perrot M. Metabolic heterogeneity of idiopathic pulmonary fibrosis: a metabolomic study. BMJ Open Respir Res 2017; 4:e000183. [PMID: 28883924 PMCID: PMC5531310 DOI: 10.1136/bmjresp-2017-000183] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 11/28/2022] Open
Abstract
Introduction Idiopathic pulmonary fibrosis (IPF) is a chronic and fatal disease of unknown cause characterised by progressive fibrotic formation in lung tissue. We hypothesise that disrupted metabolic pathways in IPF contribute to disease pathogenesis. Methods Metabolomics of human IPF was performed using mass spectroscopy (IPF lung=8; donor lung=8). Gene expression of key metabolic enzymes was measured using microarrays. Of the 108 metabolites whose levels were found altered, 48 were significantly increased, whereas 60 were significantly decreased in IPF samples compared with normal controls. Results Specific metabolic pathways mediating the IPF remodelling were found with a downregulated sphingolipid metabolic pathway but an upregulated arginine pathway in IPF. In addition, disrupted glycolysis, mitochondrial beta-oxidation and tricarboxylic acid cycle, altered bile acid, haem and glutamate/aspartate metabolism were found in IPF samples compared with control. Conclusions Our results show alterations in metabolic pathways for energy consumption during lung structural remodelling, which may contribute to IPF pathogenesis. We believe that this is the first report of simultaneously and systemically measuring changes of metabolites involving nine metabolic pathways in human severe IPF lungs. The measurement of the metabolites may serve in the future diagnosis and prognosis of IPF.
Collapse
Affiliation(s)
- Yidan D Zhao
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Li Yin
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Stephen Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Catherine Lu
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - George Zhao
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Yan Yao
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Licun Wu
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Michael Hsin
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Thomas K Waddell
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - John Granton
- Division of Respirology, University Health Network, Toronto, Ontario, Canada
| | - Marc de Perrot
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Zhang Y, Borrel A, Ghemtio L, Regad L, Boije af Gennäs G, Camproux AC, Yli-Kauhaluoma J, Xhaard H. Structural Isosteres of Phosphate Groups in the Protein Data Bank. J Chem Inf Model 2017; 57:499-516. [DOI: 10.1021/acs.jcim.6b00519] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
| | - Alexandre Borrel
- Laboratoire
Molécules Thérapeutiques in silico (MTi), UMRS-973, Université Paris Diderot, Sorbonne Paris Cité, INSERM, F-75013 Paris, France
| | | | - Leslie Regad
- Laboratoire
Molécules Thérapeutiques in silico (MTi), UMRS-973, Université Paris Diderot, Sorbonne Paris Cité, INSERM, F-75013 Paris, France
| | | | - Anne-Claude Camproux
- Laboratoire
Molécules Thérapeutiques in silico (MTi), UMRS-973, Université Paris Diderot, Sorbonne Paris Cité, INSERM, F-75013 Paris, France
| | | | | |
Collapse
|
18
|
Reid MA, Lowman XH, Pan M, Tran TQ, Warmoes MO, Ishak Gabra MB, Yang Y, Locasale JW, Kong M. IKKβ promotes metabolic adaptation to glutamine deprivation via phosphorylation and inhibition of PFKFB3. Genes Dev 2016; 30:1837-51. [PMID: 27585591 PMCID: PMC5024682 DOI: 10.1101/gad.287235.116] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 07/28/2016] [Indexed: 02/06/2023]
Abstract
In this study, Reid et al. investigate how cancer cells adapt to low glutamine conditions, which is needed for cancer cell proliferation and survival. They show that IKKβ directly interacts with and phosphorylates PFKFB3, a major driver of aerobic glycolysis, at Ser269 upon glutamine deprivation to inhibit its activity, thereby down-regulating aerobic glycolysis when glutamine levels are low and thus providing new insights into cancer cell adaptation. Glutamine is an essential nutrient for cancer cell survival and proliferation. Enhanced utilization of glutamine often depletes its local supply, yet how cancer cells adapt to low glutamine conditions is largely unknown. Here, we report that IκB kinase β (IKKβ) is activated upon glutamine deprivation and is required for cell survival independently of NF-κB transcription. We demonstrate that IKKβ directly interacts with and phosphorylates 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase isoform 3 (PFKFB3), a major driver of aerobic glycolysis, at Ser269 upon glutamine deprivation to inhibit its activity, thereby down-regulating aerobic glycolysis when glutamine levels are low. Thus, due to lack of inhibition of PFKFB3, IKKβ-deficient cells exhibit elevated aerobic glycolysis and lactate production, leading to less glucose carbons contributing to tricarboxylic acid (TCA) cycle intermediates and the pentose phosphate pathway, which results in increased glutamine dependence for both TCA cycle intermediates and reactive oxygen species suppression. Therefore, coinhibition of IKKβ and glutamine metabolism results in dramatic synergistic killing of cancer cells both in vitro and in vivo. In all, our results uncover a previously unidentified role of IKKβ in regulating glycolysis, sensing low-glutamine-induced metabolic stress, and promoting cellular adaptation to nutrient availability.
Collapse
Affiliation(s)
- Michael A Reid
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, California 91010, USA
| | - Xazmin H Lowman
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, California 91010, USA
| | - Min Pan
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, California 91010, USA
| | - Thai Q Tran
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, California 91010, USA
| | - Marc O Warmoes
- Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Mari B Ishak Gabra
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, California 91010, USA
| | - Ying Yang
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, California 91010, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27708, USA
| | - Mei Kong
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, California 91010, USA
| |
Collapse
|
19
|
|
20
|
Pegoraro C, Maczkowiak F, Monsoro-Burq AH. Pfkfb (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase) isoforms display a tissue-specific and dynamic expression during Xenopus laevis development. Gene Expr Patterns 2013; 13:203-11. [DOI: 10.1016/j.gep.2013.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 04/04/2013] [Accepted: 04/13/2013] [Indexed: 12/13/2022]
|