1
|
Amadi PU, Osuoha JO, Ekweogu CN, Jarad SJ, Efiong EE, Odika PC, Ejiofor C, Aloy-Amadi O, Gill GS, Adumekwe CW, Gaowa A, Zhang D, de Courten B, Agomuo EN. Phenolic acids from Anisopus mannii modulates phosphofructokinase 1 to improve glycemic control in patients with type 2 diabetes: A double-blind, randomized, clinical trial. Pharmacol Res 2025; 212:107602. [PMID: 39818261 DOI: 10.1016/j.phrs.2025.107602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/31/2024] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Phenolic acid-rich fraction from Anisopus mannii (PhAM) contains abundance of ferulic acid, gallic acid, protocatechuic acid, and syringic acid. Among other glycolytic enzymes, in vitro, PhAM counteracted the binding of sodium orthovanadate to phosphofructokinase 1 (PFK-1), improving its activities. In a rat model of diet-induced diabetes, PhAM monotherapy reduced HbA1c by an average of 0.63 % and fasting plasma glucose by 25 mg/dl. This herb rescued β-cells from streptozotocin-mediated destruction, thereby improving glycemic control. Supported by the preclinical trial, eighty-five patients with type 2 diabetes (T2D) receiving first-line medications were enrolled in a double-blind, randomized, placebo-controlled trial with a 90 % power level. Patients were randomized into a placebo group or either of the following two treatment groups: oral administration of 12 mg or 20 mg/kg body weight of PhAM once every 48 h for 6 months. Both treatments were well tolerated. At the endpoint, more than 70 % of patients achieved a 0.5 - 2.0 decrease in HbA1c levels and a > 20 mg/dl decrease in fasting blood glucose, meeting the pre-specified primary outcome. 66 % of patients treated with 20 mg PhAM achieved the < 7 % HbA1c and HOMA-IR of > 1.0 goal. respectively. Our study shows that PhAM can supplement first-line medications to achieve target glycemic control within 6 months.
Collapse
Affiliation(s)
- Peter U Amadi
- Department of Pediatrics, Group on the Molecular and Cell Biology of Lipids, University of Alberta, Canada; Department of Biochemistry, Imo State University, Owerri, Nigeria.
| | | | - Chidi N Ekweogu
- Department of Medical Biochemistry, Imo State University, Owerri, Nigeria
| | - Suha J Jarad
- Department of Pediatrics, Group on the Molecular and Cell Biology of Lipids, University of Alberta, Canada; Department of Biochemistry, Group on the Molecular and Cell Biology of Lipids, University of Alberta, Canada
| | - Esienanwan E Efiong
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Prince C Odika
- Department of Biology, St. Mary's University, Halifax, Canada
| | - Chioma Ejiofor
- Department of Biochemistry, Federal University of Technology, Owerri, Nigeria
| | - Oluchi Aloy-Amadi
- Department of Medical Laboratory Science, Imo State University, Owerri, Nigeria
| | - Govind S Gill
- Department of Pediatrics, Group on the Molecular and Cell Biology of Lipids, University of Alberta, Canada
| | | | - Ailun Gaowa
- Department of Pediatrics, Group on the Molecular and Cell Biology of Lipids, University of Alberta, Canada
| | - Dawei Zhang
- Department of Pediatrics, Group on the Molecular and Cell Biology of Lipids, University of Alberta, Canada
| | - Barbora de Courten
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia; Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, Australia
| | | |
Collapse
|
2
|
Rojas-Pirela M, Andrade-Alviárez D, Rojas V, Marcos M, Salete-Granado D, Chacón-Arnaude M, Pérez-Nieto MÁ, Kemmerling U, Concepción JL, Michels PAM, Quiñones W. Exploring glycolytic enzymes in disease: potential biomarkers and therapeutic targets in neurodegeneration, cancer and parasitic infections. Open Biol 2025; 15:240239. [PMID: 39904372 PMCID: PMC11793985 DOI: 10.1098/rsob.240239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 02/06/2025] Open
Abstract
Glycolysis, present in most organisms, is evolutionarily one of the oldest metabolic pathways. It has great relevance at a physiological level because it is responsible for generating ATP in the cell through the conversion of glucose into pyruvate and reducing nicotinamide adenine dinucleotide (NADH) (that may be fed into the electron chain in the mitochondria to produce additional ATP by oxidative phosphorylation), as well as for producing intermediates that can serve as substrates for other metabolic processes. Glycolysis takes place through 10 consecutive chemical reactions, each of which is catalysed by a specific enzyme. Although energy transduction by glucose metabolism is the main function of this pathway, involvement in virulence, growth, pathogen-host interactions, immunomodulation and adaptation to environmental conditions are other functions attributed to this metabolic pathway. In humans, where glycolysis occurs mainly in the cytosol, the mislocalization of some glycolytic enzymes in various other subcellular locations, as well as alterations in their expression and regulation, has been associated with the development and progression of various diseases. In this review, we describe the role of glycolytic enzymes in the pathogenesis of diseases of clinical interest. In addition, the potential role of these enzymes as targets for drug development and their potential for use as diagnostic and prognostic markers of some pathologies are also discussed.
Collapse
Affiliation(s)
- Maura Rojas-Pirela
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Servicio de Medicina Interna, Hospital Universitario de Salamanca, Salamanca37007, Spain
| | - Diego Andrade-Alviárez
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - Verónica Rojas
- Instituto de Biología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso2373223, Chile
| | - Miguel Marcos
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Servicio de Medicina Interna, Hospital Universitario de Salamanca, Salamanca37007, Spain
| | - Daniel Salete-Granado
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
| | - Marirene Chacón-Arnaude
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - María Á. Pérez-Nieto
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León, Soria42002, Spain
| | - Ulrike Kemmerling
- Instituto de Ciencias Biomédicas, Universidad de Chile, Facultad de Medicina, Santiago de Chile8380453, Chile
| | - Juan Luis Concepción
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - Paul A. M. Michels
- School of Biological Sciences, University of Edinburgh, The King’s Buildings, EdinburghEH9 3FL, UK
| | - Wilfredo Quiñones
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| |
Collapse
|
3
|
Yang P, Rong X, Gao Z, Wang J, Liu Z. Metabolic and epigenetic regulation of macrophage polarization in atherosclerosis: Molecular mechanisms and targeted therapies. Pharmacol Res 2025; 212:107588. [PMID: 39778637 DOI: 10.1016/j.phrs.2025.107588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/05/2024] [Accepted: 01/04/2025] [Indexed: 01/11/2025]
Abstract
Atherosclerosis, a multifactorial progressive inflammatory disease, is the common pathology underlying cardiovascular and cerebrovascular diseases. The macrophage plasticity is involved in the pathogenesis of atherosclerosis. With the advance of metabolomics and epigenetics, metabolites/metabolic and epigenetic modification such as DNA methylation, histone modification and noncoding RNA, play a crucial role in macrophage polarization and the progression of atherosclerosis. Herein, we provide a comprehensive review of the essential role of metabolic and epigenetic regulation, as well as the crosstalk between the two in regulating macrophage polarization in atherosclerosis. We also highlight the potential therapeutic strategies of regulating macrophage polarization via epigenetic and metabolic modifications for atherosclerosis, and offer recommendations to advance our knowledge of the roles of metabolic-epigenetic crosstalk in macrophage polarization in the context of atherosclerosis. Fundamental studies that elucidate the mechanisms by which metabolic and epigenetic regulation of macrophage polarization influence atherosclerosis will pave the way for novel therapeutic approaches.
Collapse
Affiliation(s)
- Pinglian Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Xiaoling Rong
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhechang Gao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jiaojiao Wang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Zhiping Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
4
|
Xu Q, He X, Mou Y, Sun D, Zhang X, Han J, Liu X, Liu X, Ren X, Wang D, Wang J, Ma C, Zhang Q, Li A. Magnesium ions regulate the Warburg effect to promote the differentiation of enteric neural crest cells into neurons. Stem Cell Res Ther 2025; 16:19. [PMID: 39849616 PMCID: PMC11755793 DOI: 10.1186/s13287-024-04121-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/18/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Understanding how enteric neural crest cells (ENCCs) differentiate into neurons is crucial for neurogenesis therapy and gastrointestinal disease research. This study explores how magnesium ions regulate the glycolytic pathway to enhance ENCCs differentiation into neurons. MATERIALS AND METHODS We used polymerase chain reaction, western blot, immunofluorescence, and multielectrode array techniques to assess magnesium ions' impact on ENCCs differentiation. Non-targeted metabolomic sequencing, cellular acidification rate, oxygen consumption, and western blot analyzed sugar metabolism changes. D-glucose-13C6 isotope tracing identified key glucose flux changes. Surface plasmon resonance was used to detect the binding affinity of magnesium ions with key glycolysis genes. The elastic modulus of the hydrogel was measured using a universal testing machine, while pore size and porosity were assessed with scanning electron microscopy. Swelling ratios were determined using gravimetric analysis. In vivo, ENCCs in hydrogels were transplanted into renal capsule and subcutaneously, and magnesium ions' effects on ENCCs differentiation were evaluated. RESULTS Magnesium ions increased glycolysis levels during ENCCs differentiation into neurons, along with significant upregulation of neuronal markers β-Tubulin and ubiquitin C-terminal hydrolase L1, and enhanced functional neuronal properties. D-glucose-13C6 tracing results showed increased carbon flux in the glycolytic pathway after magnesium supplementation. The binding affinity of magnesium ions with the glycolytic key enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 was found to be 1.08 μM. Inhibiting glycolysis suppressed ENCCs differentiation into neurons, emphasizing its crucial role. The double-cross-linked hydrogel gelatin methacryloyl-alginate (gelMA-ALMA), cross-linked with magnesium ions, showed promise in enhancing ENCCs differentiation in vivo without causing systemic hypermagnesemia. CONCLUSION Magnesium ions promote ENCCs differentiation into neurons by activating the Warburg effect. The GelMA-ALMA hydrogel serves as an effective localized magnesium delivery system, supporting neuronal differentiation in vivo.
Collapse
Affiliation(s)
- Qiongqian Xu
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xixi He
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Yaru Mou
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Dong Sun
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xintao Zhang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Jichang Han
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoyang Liu
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xingjian Liu
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xue Ren
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Dongming Wang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Jian Wang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Chuncan Ma
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
- Department of Pediatric Surgery, Xiangxi Tujia and Miao Autonomous Prefecture People's Hospital, Xiangxi, China
| | - Qiangye Zhang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Aiwu Li
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
5
|
Chen N, Xing S, Song J, Lu S, Ling L, Qu L. Transcriptome Reveals the Differential Regulation of Sugar Metabolism to Saline-Alkali Stress in Different Resistant Oats. Genes (Basel) 2025; 16:105. [PMID: 39858652 PMCID: PMC11765123 DOI: 10.3390/genes16010105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Saline-alkali stress is a major factor limiting the growth of oats. Sugar is the primary carbon and energy source in plants which regulates plant development and growth by regulating enzyme activity and gene expression. Sucrose, glucose, and fructose are ubiquitous plant-soluble sugars that act as signalling molecules in the transcriptional regulation of various metabolic and defence-related genes. METHODS In this study, soluble sugars, fructose, sucrose, and starch contents were measured, and transcriptomics was used to determine the differentially expressed genes (DEGs) in saline-sensitive and saline-tolerant oats after 6, 12, 24, and 48 h. DEGs annotated to carbohydrates were selected using the Kyoto Encyclopedia of Genes and Genomes. RESULTS DEGs involved in carbohydrate metabolism were mainly enriched in the glycolysis/gluconeogenesis and pentose phosphate pathways, fructose and mannose metabolism, and starch and sucrose metabolism. GAPDH, SUPI, SUS2, ATP-PEK, HXK6, FBA4, TBA4, TKT, ISA3, PPDK1, and BAM2 were significantly expressed, and a quantitative reverse transcription polymerase chain reaction verified the transcriptome sequencing results. CONCLUSIONS In this study, oats with different salinity tolerances were used to determine sugar contents under four salinity stress durations, and transcriptome sequencing was used to explore the regulatory mechanism of sugars and provide a reference for elucidating the sugar signalling regulatory mechanism under abiotic stress.
Collapse
Affiliation(s)
- Naiyu Chen
- Heilongjiang Provincial Key Laboratory of Oilfield Applied Chemistry and Technology, Daqing 163712, China
- College of Bioengineering, Daqing Normal University, Daqing 163712, China
| | - Shuya Xing
- College of Bioengineering, Daqing Normal University, Daqing 163712, China
| | - Jiaxin Song
- College of Bioengineering, Daqing Normal University, Daqing 163712, China
| | - Shutong Lu
- College of Bioengineering, Daqing Normal University, Daqing 163712, China
| | - Lei Ling
- Heilongjiang Provincial Key Laboratory of Oilfield Applied Chemistry and Technology, Daqing 163712, China
- College of Bioengineering, Daqing Normal University, Daqing 163712, China
| | - Lina Qu
- Heilongjiang Provincial Key Laboratory of Oilfield Applied Chemistry and Technology, Daqing 163712, China
- College of Bioengineering, Daqing Normal University, Daqing 163712, China
| |
Collapse
|
6
|
Eyster C, Matsuzaki S, Pranay A, Giorgione JR, Faakye A, Ahmed M, Humphries KM. Mechanistic studies of PFKFB2 reveals a novel inhibitor of its kinase activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.25.630325. [PMID: 39763797 PMCID: PMC11703173 DOI: 10.1101/2024.12.25.630325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
The 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase (PFKFB) family of proteins are bifunctional enzymes that are of clinical relevance because of their roles in regulating glycolysis in insulin sensitive tissues and cancer. Here, we sought to express recombinant PFKFB2 and develop a robust protocol to measure its kinase activity. These studies resulted in the unexpected finding that bacterially expressed PFKFB2 is phosphorylated in situ on Ser483 but is not a result of autophosphorylation. Recombinant PFKFB2 was used to develop an enzymatic assay to test a library of molecules selected by the Atomwise AtomNet® AI platform. This resulted in the identification of a new inhibitor, B2, that inhibits PFKFB2 (IC50 3.29 μM) and PFKFB3 (IC50 11.89 μM). A-498 cells, which express both PFKFB2 and PFKFB3, were treated with B2. Seahorse XFe analysis revealed B2 inhibited cellular glycolysis and glycolytic capacity. Targeted LC/MS analysis showed B2 decreased fructose-1,6-bisphosphate and downstream glycolytic intermediates but increased fructose-6-phosphate levels, which is consistent with an inhibitory effect on PFK-1 activity. The LC/MS metabolic profile of A-498 cells treated under identical conditions with the known PFKFB3 inhibitor, PFK158, was distinct from that induced by B2. These results thus demonstrate the identification and validation of a new PFKFB kinase inhibitor.
Collapse
Affiliation(s)
- Craig Eyster
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Satoshi Matsuzaki
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Atul Pranay
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Jennifer R. Giorgione
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Anna Faakye
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Biochemistry and Molecular Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Mostafa Ahmed
- Atomwise Inc., 221 Main Street, Suite 1350, San Francisco, CA 94105
| | - Kenneth M. Humphries
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Biochemistry and Molecular Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
7
|
Lypova N, Dougherty SM, Clem BF, Feng J, Yin X, Zhang X, Li X, Chesney JA, Imbert-Fernandez Y. PFKFB3-dependent redox homeostasis and DNA repair support cell survival under EGFR-TKIs in non-small cell lung carcinoma. Cancer Metab 2024; 12:37. [PMID: 39696407 PMCID: PMC11658331 DOI: 10.1186/s40170-024-00366-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The efficacy of tyrosine kinase inhibitors (TKIs) targeting the EGFR is limited due to the persistence of drug-tolerant cell populations, leading to therapy resistance. Non-genetic mechanisms, such as metabolic rewiring, play a significant role in driving lung cancer cells into the drug-tolerant state, allowing them to persist under continuous drug treatment. METHODS Our study employed a comprehensive approach to examine the impact of the glycolytic regulator 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB3) on the adaptivity of lung cancer cells to EGFR TKI therapies. We conducted metabolomics to trace glucose rerouting in response to PFKFB3 inhibition during TKI treatment. Live cell imaging and DCFDA oxidation were used to quantify levels of oxidation stress. Immunocytochemistry and Neutral Comet assay were employed to evaluate DNA integrity in response to therapy-driven oxidative stress. RESULTS Our metabolic profiling revealed that PFKFB3 inhibition significantly alters the metabolic profile of TKI-treated cells. It limited glucose utilization in the polyol pathway, glycolysis, and TCA cycle, leading to a depletion of ATP levels. Furthermore, pharmacological inhibition of PFKFB3 overcome TKI-driven redox capacity by diminishing the expression of glutathione peroxidase 4 (GPX4), thereby exacerbating oxidative stress. Our study also unveiled a novel role of PFKFB3 in DNA oxidation and damage by controlling the expression of DNA-glycosylases involved in base excision repair. Consequently, PFKFB3 inhibition improved the cytotoxicity of EGFR-TKIs by facilitating ROS-dependent cell death. CONCLUSIONS Our results suggest that PFKFB3 inhibition reduces glucose utilization and DNA damage repair, limiting the adaptivity of the cells to therapy-driven oxidative stress and DNA integrity insults. Inhibiting PFKFB3 can be an effective strategy to eradicate cancer cells surviving under EGFR TKI therapy before they enter the drug-resistant state. These findings may have potential implications in the development of new therapies for drug-resistant cancer treatment.
Collapse
Affiliation(s)
- Nadiia Lypova
- Department of Medicine, School of Medicine, University of Louisville, Louisville, KY, 40202, USA.
| | - Susan M Dougherty
- Department of Medicine, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | - Brian F Clem
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
- UofL Health-Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Jing Feng
- Center for Regulatory Environmental Analytical Metabolomics, University of Louisville, Louisville, KY, 40208, USA
- Department of Chemistry, University of Louisville, Louisville, KY, 40208, USA
| | - Xinmin Yin
- Center for Regulatory Environmental Analytical Metabolomics, University of Louisville, Louisville, KY, 40208, USA
- Department of Chemistry, University of Louisville, Louisville, KY, 40208, USA
| | - Xiang Zhang
- Center for Regulatory Environmental Analytical Metabolomics, University of Louisville, Louisville, KY, 40208, USA
- Department of Chemistry, University of Louisville, Louisville, KY, 40208, USA
| | - Xiaohong Li
- Department of Anatomical Sciences and Neurobiology, Bioinformatics Core, University of Louisville, Louisville, KY, 40202, USA
| | - Jason A Chesney
- UofL Health-Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Yoannis Imbert-Fernandez
- Department of Medicine, School of Medicine, University of Louisville, Louisville, KY, 40202, USA.
- UofL Health-Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
8
|
He R, She Z, Zhang Y, Yao S, Wei J, Du M, An S. PKC phospho-activated PFK1 is required for PBAN regulated sex pheromone biosynthesis in Helicoverpa armigera. JOURNAL OF INSECT PHYSIOLOGY 2024:104739. [PMID: 39674363 DOI: 10.1016/j.jinsphys.2024.104739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/16/2024] [Accepted: 12/04/2024] [Indexed: 12/16/2024]
Abstract
The enzyme 6-phosphofructokinase-1 (PFK1) acts as the primary rate-limiting enzyme in glycolysis, catalyzing the conversion of fructose-6-phosphate to fructose-1,6-bisphosphate. This glycolytic process provides essential substrates for the synthesis of sex pheromones. However, the specific function of PFK1 in sex pheromone biosynthesis remains unidentified. This study aimed to investigate the detailed mechanism by which PFK1 influences pheromone biosynthesis activating neuropeptide (PBAN)-regulated sex pheromone biosynthesis in Hecoverpa armigera. Findings revealed the presence of two PFK genes in pheromone glands (PGs). Further investigation demonstrated that RNAi-mediated knockdown of PFK1 significantly reduced sex pheromone production, mating success and the female ability to attract males, whereas PFK2 did not influence sex pheromone biosynthesis. Importantly, PFK1 was activated by PBAN in both isolated PGs and Sf9 cells. However, PBAN-induced activation of PFK1 could be attenuated by chelerythrine chloride (CC), a specific inhibitor of protein kinase C (PKC). Furthermore, the phosphorylation levels of PFK1 significantly increased in response to PBAN challenge, while CC treatment significantly mitigated this phosphorylation. PFK1 activity was found to depend on phosphorylation at the S135 and S676 sites in response to PBAN stimulation. Mutants at these sites abolished PFK1 phosphorylation and its activity. Overall, our findings unveil a critical mechanism by which the PBAN signaling recruits PKC to phosphorylate PFK1 at S135 and S676 sites, thereby activating PFK1. This activation ensures the normal progression of the glycolysis pathway, ultimately facilitating sex pheromone biosynthesis.
Collapse
Affiliation(s)
- Ruolan He
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Zelong She
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Yao Zhang
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Shuangyan Yao
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Jizhen Wei
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Mengfang Du
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China.
| | - Shiheng An
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China.
| |
Collapse
|
9
|
Chen M, Li H, Li Y, Luo Y, He Y, Shui X, Lei W. Glycolysis modulation: New therapeutic strategies to improve pulmonary hypertension (Review). Int J Mol Med 2024; 54:115. [PMID: 39422043 PMCID: PMC11518579 DOI: 10.3892/ijmm.2024.5439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
Pulmonary hypertension (PH) is a progressive life‑threatening cardiopulmonary vascular disease involving various pathological mechanisms, including hypoxia, cellular metabolism, inflammation, abnormal proliferation and apoptosis. Specifically, metabolism has attracted the most attention. Glucose metabolism is essential to maintain the cardiopulmonary vascular function. However, once exposed to a noxious stimulus, intracellular glucose metabolism changes or switches to an alternative pathway more suitable for adaptation, which is known as metabolic reprogramming. By promoting the switch from oxidative phosphorylation to glycolysis, cellular metabolic reprogramming plays an important role in PH development. Suppression of glucose oxidation and secondary upregulation of glycolysis are responsible for various features of PH, including the proliferation and apoptosis resistance of pulmonary artery endothelial and smooth muscle cells. In the present review, the roles and importance of the glucose metabolism shift were discussed to aid in the development of new treatment approaches for PH.
Collapse
Affiliation(s)
- Meihong Chen
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Hui Li
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yun Li
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yangui Luo
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yuan He
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Xiaorong Shui
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Vascular Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Wei Lei
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Precision Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
10
|
Desterke C, Francés R, Monge C, Marchio A, Pineau P, Mata-Garrido J. Single-Cell RNA-Seq Analysis Links DNMT3B and PFKFB4 Transcriptional Profiles with Metastatic Traits in Hepatoblastoma. Biomolecules 2024; 14:1394. [PMID: 39595571 PMCID: PMC11591731 DOI: 10.3390/biom14111394] [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: 09/13/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Hepatoblastoma is the most common primary liver cancer in children. Poor outcomes are primarily associated with patients who have distant metastases. Using the Mammalian Metabolic Enzyme Database, we investigated the overexpression of metabolic enzymes in hepatoblastoma tumors compared to noncancerous liver tissue in the GSE131329 transcriptome dataset. For the overexpressed enzymes, we applied ElasticNet machine learning to assess their predictive value for metastasis. A metabolic expression score was then computed from the significant enzymes and integrated into a clinical-biological logistic regression model. Forty-one overexpressed enzymes distinguished hepatoblastoma tumors from noncancerous liver tissues. Eighteen of these enzymes predicted metastasis status with an AUC of 0.90, demonstrating 85.7% sensitivity and 92.3% specificity. ElasticNet machine learning identified DNMT3B and PFKFB4 as key predictors of metastasis. Univariate analyses confirmed the significance of these enzymes, with respective p-values of 0.0058 and 0.0091. A metabolic score based on DNMT3B and PFKFB4 expression discriminated metastasis status and high-risk CHIC scores (p-value = 0.005). The metabolic score was more sensitive than the C1/C2 classifier in predicting metastasis (accuracy: 0.72 vs. 0.55). In a regression model integrating the metabolic score with epidemiological parameters (gender, age at diagnosis, histological type, and clinical PRETEXT stage), the metabolic score was confirmed as an independent adverse predictor of metastasis (p-value = 0.003, odds ratio: 2.12). This study identified the dual overexpression of PFKFB4 and DNMT3B in hepatoblastoma patients at risk of metastasis (high-risk CHIC classification). The combined tumor expression of DNMT3B and PFKFB4 was used to compute a metabolic score, which was validated as an independent predictor of metastatic status in hepatoblastoma.
Collapse
Affiliation(s)
- Christophe Desterke
- Faculté de Médecine du Kremlin Bicêtre, Université Paris-Sud, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France;
| | - Raquel Francés
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, 75006 Paris, France;
| | - Claudia Monge
- INSERM U993, Unité Organisation Nucléaire et Oncogenèse, Institut Pasteur, Université Paris Cité, 75015 Paris, France; (C.M.); (A.M.); (P.P.)
| | - Agnès Marchio
- INSERM U993, Unité Organisation Nucléaire et Oncogenèse, Institut Pasteur, Université Paris Cité, 75015 Paris, France; (C.M.); (A.M.); (P.P.)
| | - Pascal Pineau
- INSERM U993, Unité Organisation Nucléaire et Oncogenèse, Institut Pasteur, Université Paris Cité, 75015 Paris, France; (C.M.); (A.M.); (P.P.)
| | - Jorge Mata-Garrido
- INSERM U993, Unité Organisation Nucléaire et Oncogenèse, Institut Pasteur, Université Paris Cité, 75015 Paris, France; (C.M.); (A.M.); (P.P.)
| |
Collapse
|
11
|
Kwon HJ, Hahn KR, Moon SM, Yoo DY, Kim DW, Hwang IK. PFKFB3 ameliorates ischemia-induced neuronal damage by reducing reactive oxygen species and inhibiting nuclear translocation of Cdk5. Sci Rep 2024; 14:24694. [PMID: 39433564 PMCID: PMC11494100 DOI: 10.1038/s41598-024-75031-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
The enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB) plays an essential role in glycolysis and in the antioxidant pathway associated with glutathione. Therefore, we investigated the effects of PFKFB3 on oxidative and ischemic damage. We synthesized a fusion protein of transactivator of transcription (Tat)-PFKFB3 to facilitate its passage into the intracellular space and examine its effects against oxidative stress induced by hydrogen peroxide (H2O2) treatment and ischemic damage caused by occlusion of the common carotid arteries for 5 min in gerbils. The Tat-PFKFB3 protein was efficiently delivered into HT22 cells in a concentration- and time-dependent manner, with higher levels observed 18 h after treatment. Furthermore, treatment with 6 µM Tat-PFKFB3 demonstrated intracellular delivery into HT22 cells, as analyzed through immunocytochemical staining. Moreover, it significantly ameliorated the reduction of cell viability induced by 200 µM H2O2 treatment. Tat-PFKFB3 treatment also alleviated H2O2-induced DNA fragmentation and reactive oxygen species formation in HT22 cells. In gerbils, the intraperitoneal administration of 2 mg/kg Tat-PFKFB3 efficiently delivered the substance to all hippocampal areas, including the hippocampal CA1 region. This administration significantly mitigated ischemia-induced hyperlocomotion, long-term memory deficits, and ischemic neuronal death in the hippocampal CA1 region after ischemia. Additionally, treatment with 2 mg/kg Tat-PFKFB3 significantly ameliorated the translocation of Cdk5 from the cytosol to the nucleus in the hippocampal CA1 region 24 h after ischemia, but not in other regions. The treatment also significantly reduced reactive oxygen species formation in the CA1 region. These findings suggest that Tat-PFKFB3 reduces neuronal damage in the hippocampal CA1 region after ischemia through the reduction of Cdk5 signaling and reactive oxygen species formation. Therefore, Tat-PFKFB3 may have potential applications in reducing ischemic damage.
Collapse
Affiliation(s)
- Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
- Department of Biomedical Sciences, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Seung Myung Moon
- Department of Neurosurgery, Kangnam Sacred Heart Hospital, College of Medicine, Hallym University, Seoul, 07441, South Korea
- Research Institute for Complementary & Alternative Medicine, Hallym University, Chuncheon, 24253, South Korea
| | - Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea.
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
12
|
Fujii J. Redox remodeling of central metabolism as a driving force for cellular protection, proliferation, differentiation, and dysfunction. Free Radic Res 2024; 58:606-629. [PMID: 39316831 DOI: 10.1080/10715762.2024.2407147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/03/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
The production of reactive oxygen species (ROS) is elevated via metabolic hyperactivation in response to a variety of stimuli such as growth factors and inflammation. Tolerable amounts of ROS moderately inactivate enzymes via oxidative modification, which can be reversed back to the native form in a redox-dependent manner. The excessive production of ROS, however, causes cell dysfunction and death. Redox-reactive enzymes are present in primary metabolic pathways such as glycolysis and the tricarboxylic acid cycle, and these act as floodgates for carbon flux. Oxidation of a specific form of cysteine inhibits glyceraldehyde-3-phosphate dehydrogenase, which is reversible, and causes an accumulation of upstream intermediary compounds that increases the flux of glucose-6-phosphate to the pentose phosphate pathway. These reactions increase the NADPH and ribose-5-phosphate that are available for reductive reactions and nucleotide synthesis, respectively. On the other hand, oxidative inactivation of mitochondrial aconitase increases citrate, which is then recruited to synthesize fatty acids in the cytoplasm. Decreases in the use of carbohydrate for ATP production can be compensated via amino acid catabolism, and this metabolic change makes nitrogen available for nucleic acid synthesis. Coupling of the urea cycle also converts nitrogen to urea and polyamine, the latter of which supports cell growth. This metabolic remodeling stimulates the proliferation of tumor cells and fibrosis in oxidatively damaged tissues. Oxidative modification of these enzymes is generally reversible in the early stages of oxidizing reactions, which suggests that early treatment with appropriate antioxidants promotes the maintenance of natural metabolism.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Japan
| |
Collapse
|
13
|
Pan Y, Hatano A, Ohno S, Morita K, Kokaji T, Bai Y, Sugimoto H, Egami R, Terakawa A, Li D, Uematsu S, Maehara H, Fujita S, Inoue H, Inaba Y, Nagano AJ, Hirayama A, Soga T, Kuroda S. Time and dose selective glucose metabolism for glucose homeostasis and energy conversion in the liver. NPJ Syst Biol Appl 2024; 10:107. [PMID: 39349490 PMCID: PMC11443093 DOI: 10.1038/s41540-024-00437-2] [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: 04/20/2024] [Accepted: 09/10/2024] [Indexed: 10/02/2024] Open
Abstract
Hepatic glucose metabolism serves dual purposes: maintaining glucose homeostasis and converting glucose into energy sources; however, the underlying mechanisms are unclear. We quantitatively measured liver metabolites, gene expression, and phosphorylated insulin signaling molecules in mice orally administered varying doses of glucose, and constructed a transomic network. Rapid phosphorylation of insulin signaling molecules in response to glucose intake was observed, in contrast to the more gradual changes in gene expression. Glycolytic and gluconeogenic metabolites and expression of genes involved in glucose metabolism including glucose-6-phosphate, G6pc, and Pck1, demonstrated high glucose dose sensitivity. Whereas, glucokinase expression and glycogen accumulation showed low glucose dose sensitivity. During the early phase after glucose intake, metabolic flux was geared towards glucose homeostasis regardless of the glucose dose but shifted towards energy conversion during the late phase at higher glucose doses. Our research provides a comprehensive view of time- and dose-dependent selective glucose metabolism.
Collapse
Affiliation(s)
- Yifei Pan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Atsushi Hatano
- Department of Omics and Systems Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Laboratory for Integrated Cellular Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Satoshi Ohno
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Department of AI Systems Medicine, M&D Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
- Molecular Genetics Research Laboratory, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Keigo Morita
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Molecular Genetics Research Laboratory, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Toshiya Kokaji
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Data Science Center, Nara Institute of Science and Technology, Ikoma, Japan
| | - Yunfan Bai
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Hikaru Sugimoto
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Riku Egami
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Akira Terakawa
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Dongzi Li
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Saori Uematsu
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Hideki Maehara
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Suguru Fujita
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Inoue
- Metabolism and Nutrition Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yuka Inaba
- Metabolism and Nutrition Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Atsushi J Nagano
- Faculty of Agriculture, Ryukoku University, Otsu, Shiga, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Tomoyoshi Soga
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, 108-8345, Tokyo, Japan
| | - Shinya Kuroda
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan.
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
14
|
Calì C, Cantando I, Veloz Castillo MF, Gonzalez L, Bezzi P. Metabolic Reprogramming of Astrocytes in Pathological Conditions: Implications for Neurodegenerative Diseases. Int J Mol Sci 2024; 25:8922. [PMID: 39201607 PMCID: PMC11354244 DOI: 10.3390/ijms25168922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Astrocytes play a pivotal role in maintaining brain energy homeostasis, supporting neuronal function through glycolysis and lipid metabolism. This review explores the metabolic intricacies of astrocytes in both physiological and pathological conditions, highlighting their adaptive plasticity and diverse functions. Under normal conditions, astrocytes modulate synaptic activity, recycle neurotransmitters, and maintain the blood-brain barrier, ensuring a balanced energy supply and protection against oxidative stress. However, in response to central nervous system pathologies such as neurotrauma, stroke, infections, and neurodegenerative diseases like Alzheimer's and Huntington's disease, astrocytes undergo significant morphological, molecular, and metabolic changes. Reactive astrocytes upregulate glycolysis and fatty acid oxidation to meet increased energy demands, which can be protective in acute settings but may exacerbate chronic inflammation and disease progression. This review emphasizes the need for advanced molecular, genetic, and physiological tools to further understand astrocyte heterogeneity and their metabolic reprogramming in disease states.
Collapse
Affiliation(s)
- Corrado Calì
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10124 Turin, Italy;
- Neuroscience Institute Cavalieri Ottolenghi, 10143 Orbassano, Italy
| | - Iva Cantando
- Department of Fundamental Neurosciences (DNF), University of Lausanne (UNIL), 1005 Lausanne, Switzerland; (I.C.); (L.G.)
| | - Maria Fernanda Veloz Castillo
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10124 Turin, Italy;
- Neuroscience Institute Cavalieri Ottolenghi, 10143 Orbassano, Italy
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Saudi Arabia
| | - Laurine Gonzalez
- Department of Fundamental Neurosciences (DNF), University of Lausanne (UNIL), 1005 Lausanne, Switzerland; (I.C.); (L.G.)
| | - Paola Bezzi
- Department of Fundamental Neurosciences (DNF), University of Lausanne (UNIL), 1005 Lausanne, Switzerland; (I.C.); (L.G.)
- Department of Physiology and Pharmacology, University of Rome Sapienza, 00185 Rome, Italy
| |
Collapse
|
15
|
Peart LA, Draper M, Tarasov AI. The impact of GLP-1 signalling on the energy metabolism of pancreatic islet β-cells and extrapancreatic tissues. Peptides 2024; 178:171243. [PMID: 38788902 DOI: 10.1016/j.peptides.2024.171243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/19/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
Glucagon-like peptide-1 signalling impacts glucose homeostasis and appetite thereby indirectly affecting substrate availability at the whole-body level. The incretin canonically produces an insulinotropic effect, thereby lowering blood glucose levels by promoting the uptake and inhibiting the production of the sugar by peripheral tissues. Likewise, GLP-1 signalling within the central nervous system reduces the appetite and food intake, whereas its gastric effect delays the absorption of nutrients, thus improving glycaemic control and reducing the risk of postprandial hyperglycaemia. We review the molecular aspects of the GLP-1 signalling, focusing on its impact on intracellular energy metabolism. Whilst the incretin exerts its effects predominantly via a Gs receptor, which decodes the incretin signal into the elevation of intracellular cAMP levels, the downstream signalling cascades within the cell, acting on fast and slow timescales, resulting in an enhancement or an attenuation of glucose catabolism, respectively.
Collapse
Affiliation(s)
- Leah A Peart
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine, Northern Ireland BT52 1SA, UK
| | - Matthew Draper
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine, Northern Ireland BT52 1SA, UK
| | - Andrei I Tarasov
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine, Northern Ireland BT52 1SA, UK.
| |
Collapse
|
16
|
Caturano A, Galiero R, Vetrano E, Sardu C, Rinaldi L, Russo V, Monda M, Marfella R, Sasso FC. Insulin-Heart Axis: Bridging Physiology to Insulin Resistance. Int J Mol Sci 2024; 25:8369. [PMID: 39125938 PMCID: PMC11313400 DOI: 10.3390/ijms25158369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Insulin signaling is vital for regulating cellular metabolism, growth, and survival pathways, particularly in tissues such as adipose, skeletal muscle, liver, and brain. Its role in the heart, however, is less well-explored. The heart, requiring significant ATP to fuel its contractile machinery, relies on insulin signaling to manage myocardial substrate supply and directly affect cardiac muscle metabolism. This review investigates the insulin-heart axis, focusing on insulin's multifaceted influence on cardiac function, from metabolic regulation to the development of physiological cardiac hypertrophy. A central theme of this review is the pathophysiology of insulin resistance and its profound implications for cardiac health. We discuss the intricate molecular mechanisms by which insulin signaling modulates glucose and fatty acid metabolism in cardiomyocytes, emphasizing its pivotal role in maintaining cardiac energy homeostasis. Insulin resistance disrupts these processes, leading to significant cardiac metabolic disturbances, autonomic dysfunction, subcellular signaling abnormalities, and activation of the renin-angiotensin-aldosterone system. These factors collectively contribute to the progression of diabetic cardiomyopathy and other cardiovascular diseases. Insulin resistance is linked to hypertrophy, fibrosis, diastolic dysfunction, and systolic heart failure, exacerbating the risk of coronary artery disease and heart failure. Understanding the insulin-heart axis is crucial for developing therapeutic strategies to mitigate the cardiovascular complications associated with insulin resistance and diabetes.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Luca Rinaldi
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy;
| | - Vincenzo Russo
- Department of Biology, College of Science and Technology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA;
- Division of Cardiology, Department of Medical Translational Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| |
Collapse
|
17
|
Cao Y, Wang S, Zhang M, Lai B, Liang Y. PFKFB3-mediated glycolysis in hepatic stellate cells promotes liver regeneration. Biochem Biophys Res Commun 2024; 712-713:149958. [PMID: 38640731 DOI: 10.1016/j.bbrc.2024.149958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
Hepatic stellate cells (HSCs) perform a significant function in liver regeneration (LR) by becoming active. We propose to investigate if activated HSCs enhance glycolysis via PFKFB3, an essential glycolytic regulator, and whether targeting this pathway could be beneficial for LR. The liver and isolated HSCs of mice subjected to 2/3 partial hepatectomy (PHx) exhibited a significant rise in PFKFB3 expression, as indicated by quantitative RT-PCR analyses and Western blotting. Also, the primary HSCs of mice subjected to PHx have a significant elevation of the glycolysis level. Knocking down PFKFB3 significantly diminished the enhancement of glycolysis by PDGF in human LX2 cells. The hepatocyte proliferation in mice treated with PHx was almost completely prevented when the PFKFB3 inhibitor 3PO was administered, emerging that PFKFB3 is essential in LR. Furthermore, there was a decline in mRNA expression of immediate early genes and proinflammatory cytokines. In terms of mechanism, both the p38 MAP kinase and ERK1/2 phosphorylation in LO2 cells and LO2 proliferation were significantly reduced by the conditioned medium (CM) obtained from LX2 cells with either PFKFB3 knockdown or inhibition. Compared to the control group, isolated hepatocytes from 3PO-treated mice showed decreased p38 MAP kinase and ERK1/2 phosphorylation and proliferation. Thus, LR after PHx involves the activation of PFKFB3 in HSCs, which enhances glycolysis and promotes lactate production, thereby facilitating hepatocyte proliferation via the p38/ERK MAPK signaling pathway.
Collapse
Affiliation(s)
- Yapeng Cao
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Siyu Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Min Zhang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Baochang Lai
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yanni Liang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xian Yang, 712046, China.
| |
Collapse
|
18
|
Vieira-Lara MA, Bakker BM. The paradox of fatty-acid β-oxidation in muscle insulin resistance: Metabolic control and muscle heterogeneity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167172. [PMID: 38631409 DOI: 10.1016/j.bbadis.2024.167172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/18/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
The skeletal muscle is a metabolically heterogeneous tissue that plays a key role in maintaining whole-body glucose homeostasis. It is well known that muscle insulin resistance (IR) precedes the development of type 2 diabetes. There is a consensus that the accumulation of specific lipid species in the tissue can drive IR. However, the role of the mitochondrial fatty-acid β-oxidation in IR and, consequently, in the control of glucose uptake remains paradoxical: interventions that either inhibit or activate fatty-acid β-oxidation have been shown to prevent IR. We here discuss the current theories and evidence for the interplay between β-oxidation and glucose uptake in IR. To address the underlying intricacies, we (1) dive into the control of glucose uptake fluxes into muscle tissues using the framework of Metabolic Control Analysis, and (2) disentangle concepts of flux and catalytic capacities taking into account skeletal muscle heterogeneity. Finally, we speculate about hitherto unexplored mechanisms that could bring contrasting evidence together. Elucidating how β-oxidation is connected to muscle IR and the underlying role of muscle heterogeneity enhances disease understanding and paves the way for new treatments for type 2 diabetes.
Collapse
Affiliation(s)
- Marcel A Vieira-Lara
- Laboratory of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Barbara M Bakker
- Laboratory of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
19
|
Johansen A, Thiede B, Anonsen JH, Nilsson GE. Phosphoproteomic changes in response to anoxia are tissue-specific in the anoxia-tolerant crucian carp ( Carassius carassius). Front Physiol 2024; 15:1407834. [PMID: 38872833 PMCID: PMC11170284 DOI: 10.3389/fphys.2024.1407834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/08/2024] [Indexed: 06/15/2024] Open
Abstract
Crucian carp (Carassius carassius), a freshwater fish, can survive chronic anoxia for several months at low temperatures. Consequently, anoxia-related physiological and biochemical adaptations in this species have been studied for more than half a century. Still, despite for the well-known role of protein phosphorylation in regulating cellular processes, no studies have comprehensively characterized the phosphoproteome in crucian carp. In this study, we report the global phosphoproteome in crucian carp brain and liver during anoxia and reoxygenation. By applying a bottom-up proteomic approach on enriched phosphopeptides we found that the brain phosphoproteome shows surprisingly few changes during anoxia-reoxygenation exposure with only 109 out of 4200 phosphopeptides being differentially changed compared to normoxic controls. By contrast, in the liver 395 out of 1287 phosphopeptides changed. Although most changes occurred in the liver phosphoproteome, the pattern of changes indicated metabolic depression and decreased translation in both brain and liver. We also found changes in phosphoproteins involved in apoptotic regulation and reactive oxygen species handling in both tissues. In the brain, some of the most changed phosphopeptides belonged to proteins involved in central nervous system development and neuronal activity at the synaptic cleft. Changed phosphoproteins specific for liver tissue were related to glucose metabolism, such as glycolytic flux and glycogenolysis. In conclusion, protein phosphorylation in response to anoxia and reoxygenation showed both common and tissue-specific changes related to the functional differences between brain and liver.
Collapse
Affiliation(s)
| | - Bernd Thiede
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jan Haug Anonsen
- Department of Biosciences, University of Oslo, Oslo, Norway
- Norwegian Research Centre AS, Climate and Environment Department, Stavanger, Norway
| | | |
Collapse
|
20
|
Laurent J, Diop M, Amara R, Fisson C, Armengaud J, Labadie P, Budzinski H, Couteau J, Maillet G, Le Floch S, Laroche J, Pichereau V. Relevance of flounder caging and proteomics to explore the impact of a major industrial accident caused by fire on the Seine estuarine water quality. MARINE POLLUTION BULLETIN 2024; 201:116178. [PMID: 38401391 DOI: 10.1016/j.marpolbul.2024.116178] [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: 09/07/2023] [Revised: 02/11/2024] [Accepted: 02/16/2024] [Indexed: 02/26/2024]
Abstract
On September 26th 2019, a major fire occurred in the Lubrizol factory located near the Seine estuary, in Rouen-France. Juvenile flounders were captured in the Canche estuary (a reference system) and caged one month in the Canche and in the Seine downstream the accident site. No significant increases of PAHs, PCBs and PFAS was detected in Seine vs Canche sediments after the accident, but a significant increase of dioxins and furans was observed in water and sewage sludge in the Rouen wastewater treatment plant. The proteomics approach highlighted a dysregulation of proteins associated with cholesterol synthesis and lipid metabolism, in fish caged in the Seine. The overall results suggested that the fire produced air borne dioxins and furans that got deposited on soil and subsequently entered in the Seine estuarine waters via runoff; thus contaminating fish preys and caged flounders in the Seine estuary.
Collapse
Affiliation(s)
- Jennifer Laurent
- Univ Brest - CNRS - IRD - Ifremer, UMR 6539 LEMAR, IUEM-Université de Bretagne Occidentale, Rue Dumont D'Urville, 29280 Plouzané, France; CEDRE, 715 rue Alain Colas, 29200 Brest, France.
| | - Mamadou Diop
- Univ. Littoral Côte d'Opale, Univ. Lille, CNRS, IRD, UMR 8187, LOG, Laboratoire d'Océanologie et de Géosciences, F-62930 Wimereux, France
| | - Rachid Amara
- Univ. Littoral Côte d'Opale, Univ. Lille, CNRS, IRD, UMR 8187, LOG, Laboratoire d'Océanologie et de Géosciences, F-62930 Wimereux, France
| | - Cédric Fisson
- GIP Seine-Aval, Hangar C - Espace des Marégraphes, CS 41174, 76176 Rouen Cedex 1, France
| | - Jean Armengaud
- Laboratoire Innovations Technologiques pour la Détection et le Diagnostic (Li2D), Service de Pharmacologie et Immunoanalyse (SPI), CEA, INRAe, F-30207 Bagnols-sur-Cèze, France
| | - Pierre Labadie
- Univ. Bordeaux, CNRS, Bordeaux INP, EPOC, UMR 5805, F-33600 Pessac, France
| | - Hélène Budzinski
- Univ. Bordeaux, CNRS, Bordeaux INP, EPOC, UMR 5805, F-33600 Pessac, France
| | - Jérôme Couteau
- TOXEM, 12 rue des 4 saisons, 76290 Montivilliers, France
| | | | | | - Jean Laroche
- Univ Brest - CNRS - IRD - Ifremer, UMR 6539 LEMAR, IUEM-Université de Bretagne Occidentale, Rue Dumont D'Urville, 29280 Plouzané, France
| | - Vianney Pichereau
- Univ Brest - CNRS - IRD - Ifremer, UMR 6539 LEMAR, IUEM-Université de Bretagne Occidentale, Rue Dumont D'Urville, 29280 Plouzané, France.
| |
Collapse
|
21
|
Belužić R, Šimunić E, Podgorski II, Pinterić M, Hadžija MP, Balog T, Sobočanec S. Gene Expression Profiling Reveals Fundamental Sex-Specific Differences in SIRT3-Mediated Redox and Metabolic Signaling in Mouse Embryonic Fibroblasts. Int J Mol Sci 2024; 25:3868. [PMID: 38612678 PMCID: PMC11012119 DOI: 10.3390/ijms25073868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Sirt-3 is an important regulator of mitochondrial function and cellular energy homeostasis, whose function is associated with aging and various pathologies such as Alzheimer's disease, Parkinson's disease, cardiovascular diseases, and cancers. Many of these conditions show differences in incidence, onset, and progression between the sexes. In search of hormone-independent, sex-specific roles of Sirt-3, we performed mRNA sequencing in male and female Sirt-3 WT and KO mouse embryonic fibroblasts (MEFs). The aim of this study was to investigate the sex-specific cellular responses to the loss of Sirt-3. By comparing WT and KO MEF of both sexes, the differences in global gene expression patterns as well as in metabolic and stress responses associated with the loss of Sirt-3 have been elucidated. Significant differences in the activities of basal metabolic pathways were found both between genotypes and between sexes. In-depth pathway analysis of metabolic pathways revealed several important sex-specific phenomena. Male cells mount an adaptive Hif-1a response, shifting their metabolism toward glycolysis and energy production from fatty acids. Furthermore, the loss of Sirt-3 in male MEFs leads to mitochondrial and endoplasmic reticulum stress. Since Sirt-3 knock-out is permanent, male cells are forced to function in a state of persistent oxidative and metabolic stress. Female MEFs are able to at least partially compensate for the loss of Sirt-3 by a higher expression of antioxidant enzymes. The activation of neither Hif-1a, mitochondrial stress response, nor oxidative stress response was observed in female cells lacking Sirt-3. These findings emphasize the sex-specific role of Sirt-3, which should be considered in future research.
Collapse
|
22
|
Kovářová J, Moos M, Barrett MP, Horn D, Zíková A. The bloodstream form of Trypanosoma brucei displays non-canonical gluconeogenesis. PLoS Negl Trop Dis 2024; 18:e0012007. [PMID: 38394337 PMCID: PMC10917290 DOI: 10.1371/journal.pntd.0012007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 03/06/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Trypanosoma brucei is a causative agent of the Human and Animal African Trypanosomiases. The mammalian stage parasites infect various tissues and organs including the bloodstream, central nervous system, skin, adipose tissue and lungs. They rely on ATP produced in glycolysis, consuming large amounts of glucose, which is readily available in the mammalian host. In addition to glucose, glycerol can also be used as a source of carbon and ATP and as a substrate for gluconeogenesis. However, the physiological relevance of glycerol-fed gluconeogenesis for the mammalian-infective life cycle forms remains elusive. To demonstrate its (in)dispensability, first we must identify the enzyme(s) of the pathway. Loss of the canonical gluconeogenic enzyme, fructose-1,6-bisphosphatase, does not abolish the process hence at least one other enzyme must participate in gluconeogenesis in trypanosomes. Using a combination of CRISPR/Cas9 gene editing and RNA interference, we generated mutants for four enzymes potentially capable of contributing to gluconeogenesis: fructose-1,6-bisphoshatase, sedoheptulose-1,7-bisphosphatase, phosphofructokinase and transaldolase, alone or in various combinations. Metabolomic analyses revealed that flux through gluconeogenesis was maintained irrespective of which of these genes were lost. Our data render unlikely a previously hypothesised role of a reverse phosphofructokinase reaction in gluconeogenesis and preclude the participation of a novel biochemical pathway involving transaldolase in the process. The sustained metabolic flux in gluconeogenesis in our mutants, including a triple-null strain, indicates the presence of a unique enzyme participating in gluconeogenesis. Additionally, the data provide new insights into gluconeogenesis and the pentose phosphate pathway, and improve the current understanding of carbon metabolism of the mammalian-infective stages of T. brucei.
Collapse
Affiliation(s)
- Julie Kovářová
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Martin Moos
- Institute of Entomology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Michael P. Barrett
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, United Kingdom
| | - David Horn
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, United Kingdom
| | - Alena Zíková
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
| |
Collapse
|
23
|
Sharma S, Sarkar O, Ghosh R. Exploring the Role of Unconventional Post-Translational Modifications in Cancer Diagnostics and Therapy. Curr Protein Pept Sci 2024; 25:780-796. [PMID: 38910429 DOI: 10.2174/0113892037274615240528113148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/22/2024] [Accepted: 04/29/2024] [Indexed: 06/25/2024]
Abstract
Unconventional Post-Translational Modifications (PTMs) have gained increasing attention as crucial players in cancer development and progression. Understanding the role of unconventional PTMs in cancer has the potential to revolutionize cancer diagnosis, prognosis, and therapeutic interventions. These modifications, which include O-GlcNAcylation, glutathionylation, crotonylation, including hundreds of others, have been implicated in the dysregulation of critical cellular processes and signaling pathways in cancer cells. This review paper aims to provide a comprehensive analysis of unconventional PTMs in cancer as diagnostic markers and therapeutic targets. The paper includes reviewing the current knowledge on the functional significance of various conventional and unconventional PTMs in cancer biology. Furthermore, the paper highlights the advancements in analytical techniques, such as biochemical analyses, mass spectrometry and bioinformatic tools etc., that have enabled the detection and characterization of unconventional PTMs in cancer. These techniques have contributed to the identification of specific PTMs associated with cancer subtypes. The potential use of Unconventional PTMs as biomarkers will further help in better diagnosis and aid in discovering potent therapeutics. The knowledge about the role of Unconventional PTMs in a vast and rapidly expanding field will help in detection and targeted therapy of cancer.
Collapse
Affiliation(s)
- Sayan Sharma
- Department of Biotechnology, Amity University Kolkata, AIBNK, Kolkata, West Bengal, India
| | - Oindrila Sarkar
- Department of Biotechnology, Amity University Kolkata, AIBNK, Kolkata, West Bengal, India
| | - Rajgourab Ghosh
- Department of Biotechnology, Amity University Kolkata, AIBNK, Kolkata, West Bengal, India
| |
Collapse
|
24
|
Luo G, Li H, Lu Q, Cao J, Lv H, Jiang Y. Effects of protoscoleces excretory-secretory products of Echinococcus granulosus on hepatocyte growth, function, and glucose metabolism. Acta Trop 2024; 249:107066. [PMID: 37944837 DOI: 10.1016/j.actatropica.2023.107066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/21/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023]
Abstract
Cystic echinococcosis (CE) is one of the most widespread and harmful zoonotic parasitic diseases, which most commonly affects the liver. In this study, we characterized multiple changes in mouse hepatocytes following treatment with excretory-secretory products (ESPs) of Echinococcus granulosus protoscoleces (Eg-PSCs) by a factorial experiment. The cell counting kit-8 assay (CCK-8), the 5-ethynyl-2'-deoxyuridine (EdU) assay, and flow cytometry were used to detect the growth of hepatocytes. Inverted microscopy, scanning electron microscopy (SEM), and transmission electron microscopy (TEM) were used to observe the morphology and ultrastructure of hepatocytes. An automatic biochemical analyzer and an ELISA detection kit were used to determine six conventional hepatocyte enzymatic indices, the levels of five hepatocyte-synthesized substances, and the contents of glucose and lactate. Western blot analysis was conducted to analyze the protein expression of three apoptosis-related proteins, Bax, Bcl-2, cleaved caspase-3, and six glucose metabolism pathways rate-limiting enzymes in hepatocytes. The results showed that ESPs inhibited hepatocyte proliferation and promoted hepatocyte apoptosis. The cell membrane and microvilli of hepatocytes changed, and the nucleus, mitochondria and rough endoplasmic reticulum were damaged to varying degrees. The contents of iron, albumin (ALB), uric acid (UA) and urea were increased, and the activities of six enzymes in hepatocytes were increased except for the decrease of transferrin (TRF). The expression levels of all six key enzymes in the glucose metabolism pathway in hepatocytes were reduced. Our characterization provides a basis for further research on the pathogenesis, prevention and treatment of CE.
Collapse
Affiliation(s)
- Guangyi Luo
- Section for Hepatopancreatobiliary Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Department of Hepatopancreatobiliary Surgery, Anyue County People's Hospital, Ziyang, 642350, Sichuan, China
| | - Haiwen Li
- School of Basic Medicine, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Qiong Lu
- Department of Infectious Diseases, Anyue County People's Hospital, Ziyang, 642350, Sichuan, China
| | - Jiangtao Cao
- School of Basic Medicine, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Hailong Lv
- Section for Hepatopancreatobiliary Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
| | - Yufeng Jiang
- School of Basic Medicine, Chengdu Medical College, Chengdu, 610500, Sichuan, China.
| |
Collapse
|
25
|
Satarug S. Is Environmental Cadmium Exposure Causally Related to Diabetes and Obesity? Cells 2023; 13:83. [PMID: 38201287 PMCID: PMC10778334 DOI: 10.3390/cells13010083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Cadmium (Cd) is a pervasive toxic metal, present in most food types, cigarette smoke, and air. Most cells in the body will assimilate Cd, as its charge and ionic radius are similar to the essential metals, iron, zinc, and calcium (Fe, Zn, and Ca). Cd preferentially accumulates in the proximal tubular epithelium of the kidney, and is excreted in urine when these cells die. Thus, excretion of Cd reflects renal accumulation (body burden) and the current toxicity of Cd. The kidney is the only organ other than liver that produces and releases glucose into the circulation. Also, the kidney is responsible for filtration and the re-absorption of glucose. Cd is the least recognized diabetogenic substance although research performed in the 1980s demonstrated the diabetogenic effects of chronic oral Cd administration in neonatal rats. Approximately 10% of the global population are now living with diabetes and over 80% of these are overweight or obese. This association has fueled an intense search for any exogenous chemicals and lifestyle factors that could induce excessive weight gain. However, whilst epidemiological studies have clearly linked diabetes to Cd exposure, this appears to be independent of adiposity. This review highlights Cd exposure sources and levels associated with diabetes type 2 and the mechanisms by which Cd disrupts glucose metabolism. Special emphasis is on roles of the liver and kidney, and cellular stress responses and defenses, involving heme oxygenase-1 and -2 (HO-1 and HO-2). From heme degradation, both HO-1 and HO-2 release Fe, carbon monoxide, and a precursor substrate for producing a potent antioxidant, bilirubin. HO-2 appears to have also anti-diabetic and anti-obese actions. In old age, HO-2 deficient mice display a symptomatic spectrum of human diabetes, including hyperglycemia, insulin resistance, increased fat deposition, and hypertension.
Collapse
Affiliation(s)
- Soisungwan Satarug
- Kidney Disease Research Collaborative, Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| |
Collapse
|
26
|
Chen M, Shin M, Ware TB, Donvito G, Muchhala KH, Mischel R, Mustafa MA, Serbulea V, Upchurch CM, Leitinger N, Akbarali HI, Lichtman AH, Hsu KL. Endocannabinoid biosynthetic enzymes regulate pain response via LKB1-AMPK signaling. Proc Natl Acad Sci U S A 2023; 120:e2304900120. [PMID: 38109529 PMCID: PMC10756258 DOI: 10.1073/pnas.2304900120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 11/08/2023] [Indexed: 12/20/2023] Open
Abstract
Diacylglycerol lipase-beta (DAGLβ) serves as a principal 2-arachidonoylglycerol (2-AG) biosynthetic enzyme regulating endocannabinoid and eicosanoid metabolism in immune cells including macrophages and dendritic cells. Genetic or pharmacological inactivation of DAGLβ ameliorates inflammation and hyper-nociception in preclinical models of pathogenic pain. These beneficial effects have been assigned principally to reductions in downstream proinflammatory lipid signaling, leaving alternative mechanisms of regulation largely underexplored. Here, we apply quantitative chemical- and phospho-proteomics to find that disruption of DAGLβ in primary macrophages leads to LKB1-AMPK signaling activation, resulting in reprogramming of the phosphoproteome and bioenergetics. Notably, AMPK inhibition reversed the antinociceptive effects of DAGLβ blockade, thereby directly supporting DAGLβ-AMPK crosstalk in vivo. Our findings uncover signaling between endocannabinoid biosynthetic enzymes and ancient energy-sensing kinases to mediate cell biological and pain responses.
Collapse
Affiliation(s)
- Miaomiao Chen
- Department of Chemistry, University of Virginia, Charlottesville, VA22904
| | - Myungsun Shin
- Department of Chemistry, University of Virginia, Charlottesville, VA22904
| | - Timothy B. Ware
- Department of Chemistry, University of Virginia, Charlottesville, VA22904
| | - Giulia Donvito
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA23298
| | - Karan H. Muchhala
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA23298
| | - Ryan Mischel
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA23298
| | - Mohammed A. Mustafa
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA23298
| | - Vlad Serbulea
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA22908
| | - Clint M. Upchurch
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA22908
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA22908
| | - Hamid I. Akbarali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA23298
| | - Aron H. Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA23298
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA23298
| | - Ku-Lung Hsu
- Department of Chemistry, University of Virginia, Charlottesville, VA22904
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA22908
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA22908
- University of Virginia Cancer Center, Cancer Biology Program, University of Virginia, Charlottesville, VA22903
| |
Collapse
|
27
|
Wang B, Pu R. Association between glycolysis markers and prognosis of liver cancer: a systematic review and meta-analysis. World J Surg Oncol 2023; 21:390. [PMID: 38114977 PMCID: PMC10731852 DOI: 10.1186/s12957-023-03275-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND In recent years, the capacity of tumor cells to maintain high levels of glycolysis, even in the presence of oxygen, has emerged as one of the main metabolic traits and garnered considerable attention. The purpose of this meta-analysis is to investigate the prognostic value of glycolysis markers in liver cancer. METHODS PubMed, Embase, and Cochrane Library databases were searched for articles on glycolytic marker expression levels associated with the prognosis of liver cancer until April 2023. Stata SE14.0 was used to calculate the aggregate hazard ratios and 95% confidence intervals. RESULTS Thirty-five studies were included. The worse overall survival (OS) (P < 0.001), disease-free survival (DFS) (P = 0.001), recurrence-free survival (RFS) (P = 0.004), and time to recurrence (TTR) (P < 0.001) were significantly associated with elevated expression of glycolysis markers. Higher expression of PKM2 (P < 0.001), STMN1 (P = 0.002), MCT4 (P < 0.001), GLUT1 (P = 0.025), HK-2 (P < 0.001), and CA9 (P < 0.001) were significantly related to shorter OS. Increased levels of PKM2 (P < 0.001), CA9 (P = 0.005), and MCT4 (P < 0.001) were associated with worse DFS. Elevated PKM2 expression (P = 0.002) was also associated with poorer RFS in hepatocellular carcinoma patients. GLUT2 expression was not correlated with the prognosis of liver cancer (P = 0.134). CONCLUSIONS Elevated expression of glycolysis markers was associated with worse OS, DFS, RFS, and TTR in patients with liver cancer. Therefore, these glycolysis markers could serve as potential prognostic markers and therapeutic targets in liver cancer. TRIAL REGISTRATION PROSPERO registration: CRD42023469645.
Collapse
Affiliation(s)
- Boqin Wang
- Department of Clinical Laboratory, SSL Central Hospital of Dongguan City, Dongguan, Guangdong, China
| | - Rong Pu
- Department of Clinical Laboratory, SSL Central Hospital of Dongguan City, Dongguan, Guangdong, China.
| |
Collapse
|
28
|
Centola CL, Dasso ME, Soria JD, Riera MF, Meroni SB, Galardo MN. Glycolysis as key regulatory step in FSH-induced rat Sertoli cell proliferation: Role of the mTORC1 pathway. Biochimie 2023; 214:145-156. [PMID: 37442535 DOI: 10.1016/j.biochi.2023.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/09/2023] [Accepted: 07/08/2023] [Indexed: 07/15/2023]
Abstract
The definitive number of Sertoli cells (SCs), achieved during the proliferative periods, defines the spermatogenic capacity in adulthood. It is recognized that FSH is the main mitogen targeting SC and that it exerts its action, at least partly, through the activation of the PI3K/Akt/mTORC1 pathway. mTORC1 controls a large number of cellular functions, including glycolysis and cell proliferation. Interestingly, recent evidence revealed that the glycolytic flux might modulate mTORC1 activity and, consequently, cell cycle progression. Although mature SC metabolism has been thoroughly studied, several aspects of metabolism regulation in proliferating SC are still to be elucidated. The objective of this study was to explore whether aerobic glycolysis is regulated by FSH through mTORC1 pathway in proliferating SC, and to assess the involvement of glycolysis in the regulation of SC proliferation. The present study was carried out utilizing 8-day-old rat SC cultures. The results obtained show that FSH enhances glycolytic flux through the induction of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) and lactate dehydrogenase A (LDHA) in an mTORC1 dependent manner. In addition, PFKFB3 and LDH inhibitors prevent FSH from activating mTORC1 and stimulating SC proliferation and glycolysis, presumably through mTORC1 pathway inhibition. In summary, FSH simultaneously regulates SC proliferation and glycolysis in an mTORC1 dependent manner, and glycolysis seems to cooperate with FSH in the stimulation of both cellular functions through the modulation of the same signalling pathway. Therefore, a positive feedback between the mTORC1 pathway and glycolysis triggered by FSH is hypothesized.
Collapse
Affiliation(s)
- Cecilia Lucia Centola
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD, Ciudad Autónoma de Buenos Aires, Argentina
| | - Marina Ercilia Dasso
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD, Ciudad Autónoma de Buenos Aires, Argentina
| | - Julio Daniel Soria
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD, Ciudad Autónoma de Buenos Aires, Argentina
| | - Maria Fernanda Riera
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD, Ciudad Autónoma de Buenos Aires, Argentina
| | - Silvina Beatriz Meroni
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD, Ciudad Autónoma de Buenos Aires, Argentina
| | - Maria Noel Galardo
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
29
|
Zhang Y, Barberis M. Exploring cell cycle-mediated regulations of glycolysis in budding yeast. Front Microbiol 2023; 14:1270487. [PMID: 37886071 PMCID: PMC10598772 DOI: 10.3389/fmicb.2023.1270487] [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: 07/31/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023] Open
Abstract
Coordination of cell cycle with metabolism exists in all cell types that grow by division. It serves to build a new cell, (i) fueling building blocks for the synthesis of proteins, nucleic acids, and membranes, and (ii) producing energy through glycolysis. Cyclin-dependent kinases (Cdks) play an essential role in this coordination, thereby in the regulation of cell division. Cdks are functional homologs across eukaryotes and are the engines that drive cell cycle events and the clocks that time them. Their function is counteracted by stoichiometric inhibitors; specifically, inhibitors of cyclin-cyclin dependent kinase (cyclin/Cdk) complexes allow for their activity at specific times. Here, we provide a new perspective about the yet unknown cell cycle mechanisms impacting on metabolism. We first investigated the effect of the mitotic cyclin/Cdk1 complex Cyclin B/Cdk1-functional homolog in mammalian cells of the budding yeast Clb2/Cdk1-on yeast metabolic enzymes of, or related to, the glycolysis pathway. Six glycolytic enzymes (Glk1, Hxk2, Pgi1, Fba1, Tdh1, and Pgk1) were subjected to in vitro Cdk-mediated phosphorylation assays. Glucose-6-phosphate dehydrogenase (Zwf1), the first enzyme in the pentose phosphate pathway that is important for NADPH production, and 6-phospho-fructo-2-kinase (Pfk27), which catalyzes fructose-2,6-bisphosphate synthesis, a key regulator of glycolysis, were also included in the study. We found that, among these metabolic enzymes, Fba1 and Pgk1 may be phosphorylated by Cdk1, in addition to the known Cdk1-mediated phosphorylation of Gph1. We then investigated the possible effect of Sic1, stoichiometric inhibitor of mitotic cyclin/Cdk1 complexes in budding yeast, on the activities of three most relevant glycolytic enzymes: Hxk2, Glk1, and Tdh1. We found that Sic1 may have a negative effect on Hxk2. Altogether, we reveal possible new routes, to be further explored, through which cell cycle may regulate cellular metabolism. Because of the functional homology of cyclin/Cdk complexes and their stoichiometric inhibitors across evolution, our findings may be relevant for the regulation of cell division in eukaryotes.
Collapse
Affiliation(s)
- Yanfei Zhang
- Molecular Systems Biology, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Matteo Barberis
- Molecular Systems Biology, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
30
|
Cimmino TP, Pagano E, Stornaiuolo M, Esposito G, Ammendola R, Cattaneo F. Formyl-peptide receptor 2 signalling triggers aerobic metabolism of glucose through Nox2-dependent modulation of pyruvate dehydrogenase activity. Open Biol 2023; 13:230336. [PMID: 37875162 PMCID: PMC10597678 DOI: 10.1098/rsob.230336] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/26/2023] Open
Abstract
The human formyl-peptide receptor 2 (FPR2) is activated by an array of ligands. By phospho-proteomic analysis we proved that FPR2 stimulation induces redox-regulated phosphorylation of many proteins involved in cellular metabolic processes. In this study, we investigated metabolic pathways activated in FPR2-stimulated CaLu-6 cells. The results showed an increased concentration of metabolites involved in glucose metabolism, and an enhanced uptake of glucose mediated by GLUT4, the insulin-regulated member of GLUT family. Accordingly, we observed that FPR2 transactivated IGF-IRβ/IRβ through a molecular mechanism that requires Nox2 activity. Since cancer cells support their metabolism via glycolysis, we analysed glucose oxidation and proved that FPR2 signalling promoted kinase activity of the bifunctional enzyme PFKFB2 through FGFR1/FRS2- and Akt-dependent phosphorylation. Furthermore, FPR2 stimulation induced IGF-IRβ/IRβ-, PI3K/Akt- and Nox-dependent inhibition of pyruvate dehydrogenase activity, thus preventing the entry of pyruvate in the tricarboxylic acid cycle. Consequently, we observed an enhanced FGFR-dependent lactate dehydrogenase (LDH) activity and lactate production in FPR2-stimulated cells. As LDH expression is transcriptionally regulated by c-Myc and HIF-1, we demonstrated that FPR2 signalling promoted c-Myc phosphorylation and Nox-dependent HIF-1α stabilization. These results strongly indicate that FPR2-dependent signalling can be explored as a new therapeutic target in treatment of human cancers.
Collapse
Affiliation(s)
- Tiziana Pecchillo Cimmino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Ester Pagano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Mariano Stornaiuolo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Gabriella Esposito
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Rosario Ammendola
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Fabio Cattaneo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
31
|
Gu X, Dai X, Huang Y, Zhang Y, Dong L, Gao C, Wang F. Differential roles of highly expressed PFKFB4 in colon adenocarcinoma patients. Sci Rep 2023; 13:16284. [PMID: 37770581 PMCID: PMC10539362 DOI: 10.1038/s41598-023-43619-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 09/26/2023] [Indexed: 09/30/2023] Open
Abstract
Colon adenocarcinoma (COAD) is a common malignant tumor, and the role of the protein PFKFB4 in glycolysis and pentose phosphate pathways is crucial. Researchers investigated the clinical significance of PFKFB4 in COAD by studying its expression in 79 tissue samples using immunohistochemistry. We found that PFKFB4 expression was significantly higher in COAD patients, particularly in the sigmoid colon. Interestingly, high PFKFB4 expression was associated with both improved overall survival (OS) and worse progression-free survival (PPS) in COAD patients. Further analysis revealed that genes associated with PFKFB4 were linked to various metabolic pathways, including amino acid biosynthesis, glycolysis, gluconeogenesis, glucose metabolism, and inflammatory response. PFKFB4 expression also showed correlations with the infiltration of different immune cell types in COAD patients, such as CD8+ T cells, CD4+ T cells, regulatory T cells (Tregs), macrophages, neutrophils, dendritic cells, active mast cells, and resting NK cells. Overall, the relationship between PFKFB4 expression and the prognosis of COAD is complex and diverse, possibly playing different roles at different stages of the disease. Moreover, its mechanism might involve interactions with various metabolic pathways and immune infiltration in the tumor microenvironment. These findings provide valuable insights into the potential role of PFKFB4 as a biomarker or therapeutic target in COAD.
Collapse
Affiliation(s)
- Xiaojing Gu
- Department of Gastroenterology, General Hospital, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xingchen Dai
- Department of Gastroenterology, General Hospital, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yongli Huang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yuhuan Zhang
- Department of Gastroenterology, General Hospital, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Lintao Dong
- Department of Gastroenterology, General Hospital, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Chanchan Gao
- Department of Oncology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China.
| | - Fang Wang
- Department of Gastroenterology, General Hospital, Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
32
|
Liu F, Wei X, Chen Z, Chen Y, Hu P, Jin Y. PFKFB2 is a favorable prognostic biomarker for colorectal cancer by suppressing metastasis and tumor glycolysis. J Cancer Res Clin Oncol 2023; 149:10737-10752. [PMID: 37311985 DOI: 10.1007/s00432-023-04946-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/26/2023] [Indexed: 06/15/2023]
Abstract
PURPOSE This study was to investigate the biological effect of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 2 (PFKFB2) in colorectal cancer (CRC). METHODS PFKFB2 was selected by metabolism polymerase chain reaction (PCR) array from CRC cells under alkaline culture medium (pH 7.4) and acidic culture medium (pH 6.8). The expression of PFKFB2 mRNA and protein was detected by quantitative real-time PCR and immunohistochemistry in 70 paired fresh and 268 paired paraffin-embedded human CRC tissues, respectively, and then the prognostic value of PFKFB2 was investigated. The effects of PFKFB2 on CRC cells were also verified in vitro, which were through detecting the change of migration, invasion, sphere formation, proliferation, colony formation, and extracellular acidification rate of CRC cells after PFKFB2 knockdown in alkaline culture medium (pH 7.4) and overexpression in acidic culture medium (pH 6.8). RESULTS PFKFB2 expression was downregulated in acidic culture medium (pH 6.8). In addition, we found PFKFB2 expression decreased in human CRC tissues compared with the adjacent normal tissues. Furthermore, the OS and DFS rate of CRC patients with low PFKFB2 expression was significantly shorter than those of patients with high PFKFB2 expression. Multivariate analysis indicated that low PFKFB2 expression was an independent prognostic factor for both OS and DFS in CRC patients. Moreover, the abilities of migration, invasion, spheroidizing ability, proliferation, and colony formation of CRC cells were significantly increased after depletion of PFKFB2 in alkaline culture medium (pH 7.4) and decreased after overexpression of PFKFB2 in acidic culture medium (pH 6.8) in vitro. Epithelial-mesenchymal transition (EMT) pathway was found and verified involved in the PFKFB2-mediated regulation of metastatic function in CRC cells. Further, glycolysis of CRC cells was significantly elevated after knockdown of PFKFB2 in alkaline culture medium (pH 7.4) and decreased after overexpression of PFKFB2 in acidic culture medium (pH 6.8). CONCLUSION PFKFB2 expression is downregulated in CRC tissues and associated with worse survival for CRC patients. PFKFB2 could inhibit metastasis and the malignant progression of CRC cells by suppressing EMT and glycolysis.
Collapse
Affiliation(s)
- Furong Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
- Department of Clinical Research, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Xiaoli Wei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Zhanhong Chen
- Department of Medical Oncology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Yanxing Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Peishan Hu
- Guangdong Institute of Gastroenterology, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital (Guangdong Gastrointestinal and Anal Hospital), Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China.
| | - Ying Jin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
| |
Collapse
|
33
|
Li X, Shen H, Zhang M, Teissier V, Huang EE, Gao Q, Tsubosaka M, Toya M, Kushioka J, Maduka CV, Contag CH, Chow SKH, Zhang N, Goodman SB. Glycolytic reprogramming in macrophages and MSCs during inflammation. Front Immunol 2023; 14:1199751. [PMID: 37675119 PMCID: PMC10477714 DOI: 10.3389/fimmu.2023.1199751] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/21/2023] [Indexed: 09/08/2023] Open
Abstract
Background Dysregulated inflammation is associated with many skeletal diseases and disorders, such as osteolysis, non-union of fractures, osteonecrosis, osteoarthritis and orthopaedic infections. We previously showed that continuous infusion of lipopolysaccharide (LPS) contaminated polyethylene particles (cPE) caused prolonged inflammation and impaired bone formation. However, the metabolic and bioenergetic processes associated with inflammation of bone are unknown. Mitochondria are highly dynamic organelles that modulate cell metabolism and orchestrate the inflammatory responses that involve both resident and recruited cells. Glycolytic reprogramming, the shift from oxidative phosphorylation (OXPHOS) to glycolysis causes inappropriate cell activation and function, resulting in dysfunctional cellular metabolism. We hypothesized that impaired immunoregulation and bone regeneration from inflammatory states are associated with glycolytic reprogramming and mitochondrial dysfunction in macrophages (Mφ) and mesenchymal stromal cells (MSCs). Methods We used the Seahorse XF96 analyzer and real-time qPCR to study the bioenergetics of Mφ and MSCs exposed to cPE. To understand the oxygen consumption rate (OCR), we used Seahorse XF Cell Mito Stress Test Kit with Seahorse XF96 analyzer. Similarly, Seahorse XF Glycolytic Rate Assay Kit was used to detect the extracellular acidification rate (ECAR) and Seahorse XF Real-Time ATP Rate Assay kit was used to detect the real-time ATP production rates from OXPHOS and glycolysis. Real-time qPCR was performed to analyze the gene expression of key enzymes in glycolysis and mitochondrial biogenesis. We further detected the gene expression of proinflammatory cytokines in Mφ and genes related to cell differentiation in MSC during the challenge of cPE. Results Our results demonstrated that the oxidative phosphorylation of Mφ exposed to cPE was significantly decreased when compared with the control group. We found reduced basal, maximal and ATP-production coupled respiration rates, and decreased proton leak in Mφ during challenge with cPE. Meanwhile, Mφ showed increased basal glycolysis and proton efflux rates (PER) when exposed to cPE. The percentage (%) of PER from glycolysis was higher in Mφ exposed to cPE, indicating that the contribution of the glycolytic pathway to total extracellular acidification was elevated during the challenge of cPE. In line with the results of OCR and ECAR, we found Mφ during cPE challenge showed higher glycolytic ATP (glycoATP) production rates and lower mitochondrial ATP (mitoATP) production rates which is mainly from OXPHOS. Interestingly, MSCs showed enhanced glycolysis during challenge with cPE, but no significant changes in oxygen consumption rates (OCR). In accordance, seahorse assay of real-time ATP revealed glycoATP rates were elevated while mitoATP rates showed no significant differences in MSC during challenge with cPE. Furthermore, Mφ and MSCs exposed to cPE showed upregulated gene expression levels of glycolytic regulators and Mφ exposed to cPE expressed higher levels of pro-inflammatory cytokines. Conclusion This study demonstrated the dysfunctional bioenergetic activity of bone marrow-derived Mφ and MSCs exposed to cPE, which could impair the immunoregulatory properties of cells in the bone niche. The underlying molecular defect related to disordered mitochondrial function could represent a potential therapeutic target during the resolution of inflammation.
Collapse
Affiliation(s)
- Xueping Li
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Orthopaedic Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mao Zhang
- Cardiovascular Institute Operations, Stanford University School of Medicine, Stanford, CA, United States
| | - Victoria Teissier
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Ejun Elijah Huang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Junichi Kushioka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Chima V. Maduka
- Departments of Biomedical Engineering and Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Christopher H. Contag
- Departments of Biomedical Engineering and Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
34
|
GÜZEL S, YALÇIN A, GÜRPINAR Y, GÜLER S. Expression of Pfkfb isoenzymes during in vitro differentiation of mouse embryonic stem cells into insulin-producing cells. Turk J Med Sci 2023; 53:1565-1573. [PMID: 38813509 PMCID: PMC10760535 DOI: 10.55730/1300-0144.5725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 12/12/2023] [Accepted: 08/11/2023] [Indexed: 05/31/2024] Open
Abstract
Background/aim Type 1 diabetes mellitus (T1DM) is caused by the autoimmune-mediated destruction of insulin-producing cells (IPCs) and still has no effective cure. Better understanding of the molecular mechanisms involved in the differentiation of embryonic stem cells (ESCs) into IPCs may help us improve the therapeutic strategies for treating T1DM. 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases (Pfkfb1-4) are key regulators of glucose metabolism. Although Pfkfb3 has been shown to be required for the growth of early differentiated mouse ESCs (mESCs), more studies are needed to further assess the roles of Pfkfb isoenzymes in embryonic development and differentiation, particularly into specific cell types. In this study, we aimed to elucidate the changes in the expression of Pfkfb isoenzymes on the differentiation of mESCs into IPCs. Materials and methods A 3-step protocol was used to differentiate R1 and J1 mESCs into IPCs. The changes in the gene expression of MafA, MafB, Ins2, and Nkx6.1 (IPC specific markers) and Pfkfb1-4 were analyzed using real-time quantitative polymerase chain reaction (qPCR). Insulin expression and secretion were determined by immunofluorescence (IF) staining and the enzyme linked immunosorbent assay (ELISA), respectively. Results Upon differentiation, the IPC specific markers in differentiated cells were upregulated. Continued differentiation was confirmed by the development of insulin-positive islet-like clusters that secreted insulin in response to glucose uptake. Expressions of the Pfkfb2 and Pfkfb3 isoenzymes were markedly increased in various stages of differentiation. Conclusion These findings suggest that Pfkfb2 and Pfkfb3 may impact the differentiation of mESCs into IPCs and the regulation of the insulin response to glucose levels. This study also lays a foundation for researchers to further probe the roles of Pfkfb isoenzymes on the differentiation of mESCs into IPCs and may open new avenues for regenerative medicine.
Collapse
Affiliation(s)
- Saime GÜZEL
- Department of Biochemistry, Faculty of Veterinary Medicine, Bursa Uludağ University, Bursa,
Turkiye
| | - Abdullah YALÇIN
- Department of Biochemistry, Faculty of Veterinary Medicine, Bursa Uludağ University, Bursa,
Turkiye
| | - Yunus GÜRPINAR
- Research Center for Translational Medicine, Koç University, İstanbul,
Turkiye
| | - Sabire GÜLER
- Department of Histology & Embryology, Faculty of Veterinary Medicine, Bursa Uludağ University, Bursa,
Turkiye
| |
Collapse
|
35
|
Wang Z, Wei D, Bin E, Li J, Jiang K, Lv T, Mao X, Wang F, Dai H, Tang N. Enhanced glycolysis-mediated energy production in alveolar stem cells is required for alveolar regeneration. Cell Stem Cell 2023; 30:1028-1042.e7. [PMID: 37541209 DOI: 10.1016/j.stem.2023.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 06/07/2023] [Accepted: 07/12/2023] [Indexed: 08/06/2023]
Abstract
Impaired differentiation of alveolar stem cells has been identified in a variety of acute and chronic lung diseases. In this study, we investigate the mechanisms that modulate alveolar regeneration and understand how aging impacts this process. We have discovered that the process of alveolar type II (AT2) cells differentiating into AT1 cells is an energetically costly process. During alveolar regeneration, activated AMPK-PFKFB2 signaling upregulates glycolysis, which is essential to support the intracellular energy expenditure that is required for cytoskeletal remodeling during AT2 cell differentiation. AT2 cells in aged lungs exhibit reduced AMPK-PFKFB2 signaling and ATP production, resulting in impaired alveolar regeneration. Activating AMPK-PFKFB2 signaling in aged AT2 cells can rescue defective alveolar regeneration in aged mice. Thus, beyond demonstrating that cellular energy metabolism orchestrates with stem cell differentiation during alveolar regeneration, our study suggests that modulating AMPK-PFKFB2 signaling promotes alveolar repair in aged lungs.
Collapse
Affiliation(s)
- Zheng Wang
- National Institute of Biological Sciences, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| | - Dongdong Wei
- National Institute of Biological Sciences, Beijing, China; Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Peking University, Beijing, China
| | - Ennan Bin
- National Institute of Biological Sciences, Beijing, China
| | - Jiao Li
- National Institute of Biological Sciences, Beijing, China
| | - Kewu Jiang
- National Institute of Biological Sciences, Beijing, China
| | - Tingting Lv
- Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoxu Mao
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Fengchao Wang
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Huaping Dai
- Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Nan Tang
- National Institute of Biological Sciences, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| |
Collapse
|
36
|
Sahoo B, Srivastava M, Katiyar A, Ecelbarger C, Tiwari S. Liver or kidney: Who has the oar in the gluconeogenesis boat and when? World J Diabetes 2023; 14:1049-1056. [PMID: 37547592 PMCID: PMC10401452 DOI: 10.4239/wjd.v14.i7.1049] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/20/2023] [Accepted: 04/11/2023] [Indexed: 07/12/2023] Open
Abstract
Gluconeogenesis is an endogenous process of glucose production from non-carbohydrate carbon substrates. Both the liver and kidneys express the key enzymes necessary for endogenous glucose production and its export into circulation. We would be remiss to add that more recently gluconeogenesis has been described in the small intestine, especially under high-protein, low-carbohydrate diets. The contribution of the liver glucose release, the net glucose flux, towards systemic glucose is already well known. The liver is, in most instances, the primary bulk contributor due to the sheer size of the organ (on average, over 1 kg). The contribution of the kidney (at just over 100 g each) to endogenous glucose production is often under-appreciated, especially on a weight basis. Glucose is released from the liver through the process of glycogenolysis and gluconeogenesis. Renal glucose release is almost exclusively due to gluconeogenesis, which occurs in only a fraction of the cells in that organ (proximal tubule cells). Thus, the efficiency of glucose production from other carbon sources may be superior in the kidney relative to the liver or at least on the level. In both these tissues, gluconeogenesis regulation is under tight hormonal control and depends on the availability of substrates. Liver and renal gluconeogenesis are differentially regulated under various pathological conditions. The impact of one source vs the other changes, based on post-prandial state, acid-base balance, hormonal status, and other less understood factors. Which organ has the oar (is more influential) in driving systemic glucose homeostasis is still in-conclusive and likely changes with the daily rhythms of life. We reviewed the literature on the differences in gluconeogenesis regulation between the kidneys and the liver to gain an insight into who drives the systemic glucose levels under various physiological and pathological conditions.
Collapse
Affiliation(s)
- Biswajit Sahoo
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Medha Srivastava
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Arpit Katiyar
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Carolyn Ecelbarger
- Department of Medicine, Georgetown University, Washington, DC 20057, United States
| | - Swasti Tiwari
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| |
Collapse
|
37
|
Zhuang P, Wang D, Zhang K, Wang J, Shen J. Sorafenib promotes hepatocellular carcinoma invasion via interleukin-6/HIF-1α/PFKFB3. J Cancer 2023; 14:1859-1874. [PMID: 37476196 PMCID: PMC10355201 DOI: 10.7150/jca.84451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/04/2023] [Indexed: 07/22/2023] Open
Abstract
Background: Although sorafenib is adopted as the first-line treatment for unresectable liver cancer, the antitumor efficacy is severely limited by the pro-invasive side effect. Methods: To explore the underlying mechanisms, various-dosage sorafenib was applied to survey its effect on cell invasion in HCCLM3 and PLC cell models. Results: Our results revealed that high-dosage sorafenib inhibited liver cancer cell invasion. By contrast, sorafenib with low and median dosages promoted the invasion. In vivo studies showed that sorafenib with a median dosage increased the intrahepatic metastasis (IHM) and lung metastasis (LM) of liver cancer cells, while sorafenib with a high dosage inhibited IHM and LM. Then, bioinformatics analysis indicated that HIF-1α, IL-6, and PFKFB3 were associated with the sorafenib resistance. In vitro models showed that the pro-invasive effect was mediated by IL-6/HIF-1α/PFKFB3 regulation in dosage- and time-dependent manners. PFKFB3 knockdown confirmed that PFKFB3 promoted HCCLM3 cell migration via modulating EMT-related markers. Furthermore, we found that sorafenib upregulated PFKFB3 by IL-6/HIF-1α in a time-dependent manner, without direct effect on PFKFB3 expression. Conclusions: In summary, these results demonstrated that sorafenib could dose-dependently promote cell invasion, intrahepatic and lung metastasis in hepatocellular carcinoma through IL-6/HIF-1α mediated PFKFB3 activation, providing novel insights to improve the therapeutic efficacy of sorafenib.
Collapse
Affiliation(s)
- Pengyuan Zhuang
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, Shanghai, 200092, China
| | - Dong Wang
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, Shanghai, 200092, China
| | - Kewei Zhang
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200050, China
| | - Jiandong Wang
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, Shanghai, 200092, China
| | - Jun Shen
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, Shanghai, 200092, China
| |
Collapse
|
38
|
Wang F, Yin X, Fan YM, Zhang X, Ma C, Jia K, Zhou W, Tang Z, Qi LW, Li J. Upregulation of glycolytic enzyme PFKFB3 by deubiquitinase OTUD4 promotes cardiac fibrosis post myocardial infarction. J Mol Med (Berl) 2023:10.1007/s00109-023-02323-6. [PMID: 37162556 DOI: 10.1007/s00109-023-02323-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/22/2023] [Accepted: 04/14/2023] [Indexed: 05/11/2023]
Abstract
Metabolic dysregulations have emerged as a major mediator of cardiovascular disorders and fibrotic diseases. Metabolic reprogramming contributes a lot to cardiac fibroblast activation and cardiac fibrosis post-myocardial infarction (MI), yet the mechanism remains incompletely understood. Our work aimed to determine whether or not glycolytic reprogramming, regulated by phosphofructokinase-2/fructose-2,6-bisphosphatase 3 (PFKFB3), is a therapeutic target for alleviating post-MI cardiac fibrosis. Here, we showed that cardiac fibroblasts displayed cell energy phenotype toward augmented glycolysis in response to transforming growth factor-beta 1 (TGF-β1), evidenced by significant extracellular acidification rate (ECAR) increase and lactate accumulation. The expression of glycolytic enzyme PFKFB3, a master activator of glycolysis, was up-regulated in TGF-β1-treated cardiac fibroblasts and in cardiac fibroblasts of post-MI mice. Pharmacological inhibition of PFKFB3 by 3PO diminished TGF-β1-mediated profibrotic phenotypes, attenuated cardiac fibrosis, and preserved cardiac functions in post-MI mice. Meanwhile, the genetic inhibition of PFKFB3 decreased the cardiac fibroblast activation and reversed the differentiated phenotypes in vitro and in vivo. Mechanistically, we identified deubiquitinase OTUD4 as a new binding protein of PFKFB3, and their interaction blocked PFKFB3 degradation via OTUD4-mediated deubiquitylation. Taken together, this work characterized a key role for PFKFB3 in cardiac fibroblast activation and suggested that inhibiting PFKFB3-involved glycolysis is an alternative way to alleviate post-MI cardiac fibrosis. KEY MESSAGES: PFKFB3, a master activator of glycolysis, was highly expressed in ischemic cardiac fibroblasts to enhance cardiac fibrosis The deubiquitinase OTUD4 was identified as a new binding protein of PFKFB3 TGF-β1 blunted the ubiquitination-mediated degradation of PFKFB3 via OTUD4-mediated deubiquitylation Blockade of PFKFB3 contributed to ameliorating ischemia-induced cardiac fibrosis.
Collapse
Affiliation(s)
- Feizuo Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing, 210023, Jiangsu, China
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Road, Nanjing, 210009, Jiangsu, China
| | - Xiaojian Yin
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Road, Nanjing, 210009, Jiangsu, China
| | - Yuan-Ming Fan
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Road, Nanjing, 210009, Jiangsu, China
| | - Xinyao Zhang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Road, Nanjing, 210009, Jiangsu, China
| | - Chao Ma
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing, 210023, Jiangsu, China
| | - Keke Jia
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing, 210023, Jiangsu, China
| | - Wei Zhou
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Road, Nanjing, 210009, Jiangsu, China
| | - Zongxiang Tang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing, 210023, Jiangsu, China.
| | - Lian-Wen Qi
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Road, Nanjing, 210009, Jiangsu, China.
| | - Jia Li
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing, 210023, Jiangsu, China.
| |
Collapse
|
39
|
Cabral-Costa JV, Vicente-Gutiérrez C, Agulla J, Lapresa R, Elrod JW, Almeida Á, Bolaños JP, Kowaltowski AJ. Mitochondrial sodium/calcium exchanger NCLX regulates glycolysis in astrocytes, impacting on cognitive performance. J Neurochem 2023; 165:521-535. [PMID: 36563047 PMCID: PMC10478152 DOI: 10.1111/jnc.15745] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
Intracellular Ca2+ concentrations are strictly controlled by plasma membrane transporters, the endoplasmic reticulum, and mitochondria, in which Ca2+ uptake is mediated by the mitochondrial calcium uniporter complex (MCUc), while efflux occurs mainly through the mitochondrial Na+ /Ca2+ exchanger (NCLX). RNAseq database repository searches led us to identify the Nclx transcript as highly enriched in astrocytes when compared with neurons. To assess the role of NCLX in mouse primary culture astrocytes, we inhibited its function both pharmacologically or genetically. This resulted in re-shaping of cytosolic Ca2+ signaling and a metabolic shift that increased glycolytic flux and lactate secretion in a Ca2+ -dependent manner. Interestingly, in vivo genetic deletion of NCLX in hippocampal astrocytes improved cognitive performance in behavioral tasks, whereas hippocampal neuron-specific deletion of NCLX impaired cognitive performance. These results unveil a role for NCLX as a novel modulator of astrocytic glucose metabolism, impacting on cognition.
Collapse
Affiliation(s)
- João Victor Cabral-Costa
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
- Institute of Functional Biology and Genomics, University of Salamanca-CSIC, Salamanca, Spain
| | - Carlos Vicente-Gutiérrez
- Institute of Functional Biology and Genomics, University of Salamanca-CSIC, Salamanca, Spain
- Centro de Investigación Biomédica en Red Sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca-CSIC, Salamanca, Spain
| | - Jesús Agulla
- Institute of Functional Biology and Genomics, University of Salamanca-CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca-CSIC, Salamanca, Spain
| | - Rebeca Lapresa
- Institute of Functional Biology and Genomics, University of Salamanca-CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca-CSIC, Salamanca, Spain
| | - John W. Elrod
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Ángeles Almeida
- Institute of Functional Biology and Genomics, University of Salamanca-CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca-CSIC, Salamanca, Spain
| | - Juan P. Bolaños
- Institute of Functional Biology and Genomics, University of Salamanca-CSIC, Salamanca, Spain
- Centro de Investigación Biomédica en Red Sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca-CSIC, Salamanca, Spain
| | - Alicia J. Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
40
|
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] [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
|
41
|
Harley G, Katerelos M, Gleich K, Lee M, Mount PF, Power DA. Mutation of regulatory phosphorylation sites in PFKFB2 does not affect the anti-fibrotic effect of metformin in the kidney. PLoS One 2023; 18:e0280792. [PMID: 36757995 PMCID: PMC9910667 DOI: 10.1371/journal.pone.0280792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/09/2023] [Indexed: 02/10/2023] Open
Abstract
The anti-fibrotic effect of metformin has been widely demonstrated. Fibrosis in the kidney after injury is associated with reduced expression of genes involved in both fatty acid and glycolytic energy metabolism. We have previously reported that the anti-fibrotic effect of metformin requires phosphoregulation of fatty acid oxidation by AMP-activated protein kinase (AMPK). To determine whether metformin also acts via regulation of glycolysis, we mutated regulatory phosphosites in the PFKFB2 isoform of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase (PFKFB2), a key regulator of glycolysis in the kidney. Mice with inactivating knockin (KI) mutations of the phosphorylation sites in PFKFB2 (PFKFB2 KI mice), which reduces the ability to increase the rate of glycolysis following stimulation, were used. Metformin was administered via drinking water to mice with a unilateral ureteric obstruction (UUO) model of renal fibrosis. In the PFKFB2 KI mice treated with metformin, there was decreased fibrosis and macrophage infiltration following UUO as assessed by Western blot for fibronectin and RT-PCR for α-smooth muscle actin, collagen 3, and F4.80, and confirmed by histology. Expression of the inducible PFKFB3 isoform was increased with metformin in UUO in both WT and PFKFB2 KI mice. There was no significant difference between WT and PFKFB2 KI mice treated with metformin in the degree of fibrosis following UUO in any of the Western blot or RT-PCR parameters that were measured. These data show that inhibition of the regulation of glycolysis by PFKFB2 does not diminish the anti-fibrotic effect of metformin in a model of renal fibrosis.
Collapse
Affiliation(s)
- Geoff Harley
- Kidney Laboratory, Department of Nephrology, Austin Health, Heidelberg, VIC, Australia
- * E-mail:
| | - Marina Katerelos
- Kidney Laboratory, Department of Nephrology, Austin Health, Heidelberg, VIC, Australia
| | - Kurt Gleich
- Kidney Laboratory, Department of Nephrology, Austin Health, Heidelberg, VIC, Australia
| | - Mardiana Lee
- Kidney Laboratory, Department of Nephrology, Austin Health, Heidelberg, VIC, Australia
| | - Peter F. Mount
- Kidney Laboratory, Department of Nephrology, Austin Health, Heidelberg, VIC, Australia
- Department of Medicine, The University of Melbourne, Heidelberg, VIC, Australia
| | - David A. Power
- Kidney Laboratory, Department of Nephrology, Austin Health, Heidelberg, VIC, Australia
- Department of Medicine, The University of Melbourne, Heidelberg, VIC, Australia
| |
Collapse
|
42
|
AMPK inhibits liver gluconeogenesis: fact or fiction? Biochem J 2023; 480:105-125. [PMID: 36637190 DOI: 10.1042/bcj20220582] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/14/2023]
Abstract
Is there a role for AMPK in the control of hepatic gluconeogenesis and could targeting AMPK in liver be a viable strategy for treating type 2 diabetes? These are frequently asked questions this review tries to answer. After describing properties of AMPK and different small-molecule AMPK activators, we briefly review the various mechanisms for controlling hepatic glucose production, mainly via gluconeogenesis. The different experimental and genetic models that have been used to draw conclusions about the role of AMPK in the control of liver gluconeogenesis are critically discussed. The effects of several anti-diabetic drugs, particularly metformin, on hepatic gluconeogenesis are also considered. We conclude that the main effect of AMPK activation pertinent to the control of hepatic gluconeogenesis is to antagonize glucagon signalling in the short-term and, in the long-term, to improve insulin sensitivity by reducing hepatic lipid content.
Collapse
|
43
|
Lu YA, Brien CMO, Mashek DG, Hu WS, Zhang Q. Kinetic-model-based pathway optimization with application to reverse glycolysis in mammalian cells. Biotechnol Bioeng 2023; 120:216-229. [PMID: 36184902 DOI: 10.1002/bit.28249] [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: 07/09/2022] [Revised: 09/19/2022] [Accepted: 09/28/2022] [Indexed: 12/13/2022]
Abstract
Over the last two decades, model-based metabolic pathway optimization tools have been developed for the design of microorganisms to produce desired metabolites. However, few have considered more complex cellular systems such as mammalian cells, which requires the use of nonlinear kinetic models to capture the effects of concentration changes and cross-regulatory interactions. In this study, we develop a new two-stage pathway optimization framework based on kinetic models that incorporate detailed kinetics and regulation information. In Stage 1, a set of optimization problems are solved to identify and rank the enzymes that contribute the most to achieving the metabolic objective. Stage 2 then determines the optimal enzyme interventions for specified desired numbers of enzyme adjustments. It also incorporates multi-scenario optimization, which allows the simultaneous consideration of multiple physiological conditions. We apply the proposed framework to find enzyme adjustments that enable a reverse glucose flow in cultured mammalian cells, thereby eliminating the need for glucose feed in the late culture stage and enhancing process robustness. The computational results demonstrate the efficacy of the proposed approach; it not only captures the important regulations and key enzymes for reverse glycolysis but also identifies differences and commonalities in the metabolic requirements for different carbon sources.
Collapse
Affiliation(s)
- Yen-An Lu
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Conor M O' Brien
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Wei-Shou Hu
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Qi Zhang
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
44
|
Salem K, Reese RM, Alarid ET, Fowler AM. Progesterone Receptor-Mediated Regulation of Cellular Glucose and 18F-Fluorodeoxyglucose Uptake in Breast Cancer. J Endocr Soc 2022; 7:bvac186. [PMID: 36601022 PMCID: PMC9795483 DOI: 10.1210/jendso/bvac186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Indexed: 12/05/2022] Open
Abstract
Context Positron emission tomography imaging with 2-deoxy-2-[18F]-fluoro-D-glucose (FDG) is used clinically for initial staging, restaging, and assessing therapy response in breast cancer. Tumor FDG uptake in steroid hormone receptor-positive breast cancer and physiologic FDG uptake in normal breast tissue can be affected by hormonal factors such as menstrual cycle phase, menopausal status, and hormone replacement therapy. Objective The purpose of this study was to determine the role of the progesterone receptor (PR) in regulating glucose and FDG uptake in breast cancer cells. Methods and Results PR-positive T47D breast cancer cells treated with PR agonists had increased FDG uptake compared with ethanol control. There was no significant change in FDG uptake in response to PR agonists in PR-negative MDA-MB-231 cells, MDA-MB-468 cells, or T47D PR knockout cells. Treatment of T47D cells with PR antagonists inhibited the effect of R5020 on FDG uptake. Using T47D cell lines that only express either the PR-A or the PR-B isoform, PR agonists increased FDG uptake in both cell types. Experiments using actinomycin D and cycloheximide demonstrated the requirement for both transcription and translation in PR regulation of FDG uptake. GLUT1 and PFKFB3 mRNA expression and the enzymatic activity of glucose-6-phosphate dehydrogenase and 6-phosphogluconate dehydrogenase were increased after progestin treatment of T47D cells. Conclusion Thus, progesterone and progestins increase FDG uptake in T47D breast cancer cells through the classical action of PR as a ligand-activated transcription factor. Ligand-activated PR ultimately increases expression and activity of proteins involved in glucose uptake, glycolysis, and the pentose phosphate pathway.
Collapse
Affiliation(s)
- Kelley Salem
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Rebecca M Reese
- McArdle Laboratory for Cancer Research, Department of Oncology and Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Elaine T Alarid
- McArdle Laboratory for Cancer Research, Department of Oncology and Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI 53792, USA
| | - Amy M Fowler
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI 53792, USA.,Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
45
|
Dai T, Rosario SR, Katsuta E, Dessai AS, Paterson EJ, Novickis AT, Cortes Gomez E, Zhu B, Liu S, Wang H, Abrams SI, Seshadri M, Bshara W, Dasgupta S. Hypoxic activation of PFKFB4 in breast tumor microenvironment shapes metabolic and cellular plasticity to accentuate metastatic competence. Cell Rep 2022; 41:111756. [PMID: 36476868 PMCID: PMC9807018 DOI: 10.1016/j.celrep.2022.111756] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 09/27/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer cells encounter a hostile tumor microenvironment (TME), and their adaptations to metabolic stresses determine metastatic competence. Here, we show that the metabolic enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase-4 (PFKFB4) is induced in hypoxic tumors acquiring metabolic plasticity and invasive phenotype. In mouse models of breast cancer, genetic ablation of PFKFB4 significantly delays distant organ metastasis, reducing local lymph node invasion by suppressing expression of invasive gene signature including integrin β3. Photoacoustic imaging followed by metabolomics analyses of hypoxic tumors show that PFKFB4 drives metabolic flexibility, enabling rapid detoxification of reactive oxygen species favoring survival under selective pressure. Mechanistically, hypoxic induction triggers nuclear translocation of PFKFB4 accentuating non-canonical transcriptional activation of HIF-1α, and breast cancer patients with increased nuclear PFKFB4 in their tumors are found to be significantly associated with poor prognosis. Our findings imply that PFKFB4 induction is crucial for tumor cell adaptation in the hypoxic TME that determines metastatic competence.
Collapse
Affiliation(s)
- Tao Dai
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Spencer R. Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Eriko Katsuta
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Abhisha Sawant Dessai
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Emily J. Paterson
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Aaron T. Novickis
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Eduardo Cortes Gomez
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Bokai Zhu
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Hai Wang
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Scott I. Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Mukund Seshadri
- Department of Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Wiam Bshara
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Subhamoy Dasgupta
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA,Lead contact,Correspondence:
| |
Collapse
|
46
|
Sittewelle M, Kappès V, Zhou C, Lécuyer D, Monsoro-Burq AH. PFKFB4 interacts with ICMT and activates RAS/AKT signaling-dependent cell migration in melanoma. Life Sci Alliance 2022; 5:5/12/e202201377. [PMID: 35914811 PMCID: PMC9348664 DOI: 10.26508/lsa.202201377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022] Open
Abstract
Glycolysis regulator PFKFB4 promotes cell migration in metastatic melanoma and normal melanocytes by a non-conventional glycolysis-independent function involving ICMT, RAS, and AKT signaling. Cell migration is a complex process, tightly regulated during embryonic development and abnormally activated during cancer metastasis. RAS-dependent signaling is a major nexus controlling essential cell parameters including proliferation, survival, and migration, utilizing downstream effectors such as the PI3K/AKT signaling pathway. In melanoma, oncogenic mutations frequently enhance RAS, PI3K/AKT, or MAP kinase signaling and trigger other cancer hallmarks among which the activation of metabolism regulators. PFKFB4 is one of these critical regulators of glycolysis and of the Warburg effect. Here, however, we explore a novel function of PFKFB4 in melanoma cell migration. We find that PFKFB4 interacts with ICMT, a posttranslational modifier of RAS. PFKFB4 promotes ICMT/RAS interaction, controls RAS localization at the plasma membrane, activates AKT signaling and enhances cell migration. We thus provide evidence of a novel and glycolysis-independent function of PFKFB4 in human cancer cells. This unconventional activity links the metabolic regulator PFKFB4 to RAS-AKT signaling and impacts melanoma cell migration.
Collapse
Affiliation(s)
- Méghane Sittewelle
- Université Paris-Saclay, Faculté des Sciences d'Orsay, CNRS UMR 3347, INSERM U1021, Orsay, France.,Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
| | - Vincent Kappès
- Université Paris-Saclay, Faculté des Sciences d'Orsay, CNRS UMR 3347, INSERM U1021, Orsay, France.,Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
| | - Chenxi Zhou
- Université Paris-Saclay, Faculté des Sciences d'Orsay, CNRS UMR 3347, INSERM U1021, Orsay, France.,Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
| | - Déborah Lécuyer
- Université Paris-Saclay, Faculté des Sciences d'Orsay, CNRS UMR 3347, INSERM U1021, Orsay, France.,Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
| | - Anne H Monsoro-Burq
- Université Paris-Saclay, Faculté des Sciences d'Orsay, CNRS UMR 3347, INSERM U1021, Orsay, France .,Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
| |
Collapse
|
47
|
Huang C, Lei H, Liu C, Wang Y. Acute and subchronic exposure of cyadox induced metabolic and transcriptomic disturbances in Wistar rats. Toxicology 2022; 482:153367. [DOI: 10.1016/j.tox.2022.153367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
|
48
|
Gu SH, Chang CH, Lin PL. Bombyxin-stimulated ecdysteroidogenesis in relation to sugar transporter/trehalase expressions in Bombyx prothoracic glands. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 151:103864. [PMID: 36336193 DOI: 10.1016/j.ibmb.2022.103864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 10/22/2022] [Accepted: 10/23/2022] [Indexed: 06/16/2023]
Abstract
Our previous studies showed that bombyxin stimulated ecdysteroidogenesis in Bombyx mori prothoracic glands (PGs) during a long-term incubation period in a phosphatidylinositol 3-kinase (PI3K)/Akt-dependent manner. In the present study, we further investigated the downstream signaling cascade in bombyxin-stimulated PGs. Our results showed that upon treatment with bombyxin, expression levels of the sugar transport 1 (St1) and St4 genes and trehalase 1 (Treh1) gene, but not ecdysteroid biosynthesis genes were greatly enhanced compared to the controls. Treatment with LY294002 (an inhibitor of PI3K) reduced the enhanced St1 and Treh1 expression levels, clearly indicating the involvement of PI3K. Treatment with 1 mM of mpV(pic) (a potent inhibitor of protein phosphotyrosine phosphatase and activator of insulin receptor (InR) kinase) also stimulated expression levels of the St1 and Treh1 genes, thus further confirming the involvement of the InR. Determining Treh enzyme activity showed that bombyxin treatment stimulated Treh enzyme activity in time- and PI3K-dependent manners. Validamycin A (a Treh inhibitor) blocked bombyxin-stimulated Treh enzyme activity and partly decreased bombyxin-stimulated ecdysteroidogenesis. A specific sugar transport inhibitor (cytochalasin B) and a glycolysis inhibitor (2-deoxy-D-glucose (2-DG)) also reduced bombyxin-stimulated ecdysteroidogenesis. Taken together, these results indicated that increased expressions of Sts and Treh1 and enhanced Treh enzyme activity downstream of InR/PI3K are involved in bombyxin-stimulated ecdysteroidogenesis in B. mori PGs.
Collapse
Affiliation(s)
- Shi-Hong Gu
- Department of Biology, National Museum of Natural Science, 1 Kuan-Chien Road, Taichung, 404, Taiwan, ROC.
| | - Chia-Hao Chang
- Department of Biology, National Museum of Natural Science, 1 Kuan-Chien Road, Taichung, 404, Taiwan, ROC
| | - Pei-Ling Lin
- Department of Biology, National Museum of Natural Science, 1 Kuan-Chien Road, Taichung, 404, Taiwan, ROC
| |
Collapse
|
49
|
Hu D, Zhang Y, Liu D, Wang D, Tian C. PFK2/FBPase-2 is a potential target for metabolic engineering in the filamentous fungus Myceliophthora thermophila. Front Microbiol 2022; 13:1056694. [PMID: 36478865 PMCID: PMC9721465 DOI: 10.3389/fmicb.2022.1056694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/07/2022] [Indexed: 09/05/2023] Open
Abstract
The key enzyme 6-phosphofructo-2-kinase (PFK2)/fructose-2,6-bisphosphatase (FBPase-2) is responsible for regulating the rates of glycolysis and gluconeogenesis in eukaryotes. However, its functions and mechanisms in filamentous fungi remain largely enigmatic. In this study, we systematically investigated the function of this enzyme in Myceliophthora thermophila, a thermophilic filamentous fungus with great capacity to produce industrial enzymes and organic acids. Our results showed that the M. thermophila genome encodes three isomers, all with the PFK2/FBPase-2 structure: pfk2-a, pfk2-b, and pfk2-c. Overexpression of each gene revealed that endogenous expression of pfk2-c (PFK2 activity) promoted glucose metabolism, while overexpression of pfk2-a (FBPase-2 activity) inhibited strain growth. Using knockouts, we found that each gene was individually non-essential, but the triple knockout led to significantly slower growth compared with the wild-type strain. Only the pfk2-a single knockout exhibited 22.15% faster sugar metabolism, exerted through activation of 6-phosphofructo-1-kinase (PFK1), thereby significantly promoting glycolysis and the tricarboxylic acid cycle. The FBPase-2 deletion mutant strain also exhibited overflow metabolism, and knocking out pfk2-a was proved to be able to improve the production and synthesis rate of various metabolites, such as glycerol and malate. This is the first study to systematically investigate the function of PFK2/FBPase-2 in a thermophilic fungus, providing an effective target for metabolic engineering in filamentous fungi.
Collapse
Affiliation(s)
- Die Hu
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Key Laboratory of Systems Microbial Biotechnology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Technology Innovation Center of Synthetic Biology, Tianjin, China
| | - Yongli Zhang
- Key Laboratory of Systems Microbial Biotechnology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Technology Innovation Center of Synthetic Biology, Tianjin, China
| | - Defei Liu
- Key Laboratory of Systems Microbial Biotechnology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Technology Innovation Center of Synthetic Biology, Tianjin, China
| | - Depei Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Chaoguang Tian
- Key Laboratory of Systems Microbial Biotechnology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Technology Innovation Center of Synthetic Biology, Tianjin, China
| |
Collapse
|
50
|
McGrail K, Granado-Martínez P, Esteve-Puig R, García-Ortega S, Ding Y, Sánchez-Redondo S, Ferrer B, Hernandez-Losa J, Canals F, Manzano A, Navarro-Sabaté A, Bartrons R, Yanes O, Pérez-Alea M, Muñoz-Couselo E, Garcia-Patos V, Recio JA. BRAF activation by metabolic stress promotes glycolysis sensitizing NRAS Q61-mutated melanomas to targeted therapy. Nat Commun 2022; 13:7113. [PMID: 36402789 PMCID: PMC9675737 DOI: 10.1038/s41467-022-34907-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/08/2022] [Indexed: 11/21/2022] Open
Abstract
NRAS-mutated melanoma lacks a specific line of treatment. Metabolic reprogramming is considered a novel target to control cancer; however, NRAS-oncogene contribution to this cancer hallmark is mostly unknown. Here, we show that NRASQ61-mutated melanomas specific metabolic settings mediate cell sensitivity to sorafenib upon metabolic stress. Mechanistically, these cells are dependent on glucose metabolism, in which glucose deprivation promotes a switch from CRAF to BRAF signaling. This scenario contributes to cell survival and sustains glucose metabolism through BRAF-mediated phosphorylation of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-2/3 (PFKFB2/PFKFB3). In turn, this favors the allosteric activation of phosphofructokinase-1 (PFK1), generating a feedback loop that couples glycolytic flux and the RAS signaling pathway. An in vivo treatment of NRASQ61 mutant melanomas, including patient-derived xenografts, with 2-deoxy-D-glucose (2-DG) and sorafenib effectively inhibits tumor growth. Thus, we provide evidence for NRAS-oncogene contributions to metabolic rewiring and a proof-of-principle for the treatment of NRASQ61-mutated melanoma combining metabolic stress (glycolysis inhibitors) and previously approved drugs, such as sorafenib.
Collapse
Affiliation(s)
- Kimberley McGrail
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Paula Granado-Martínez
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Rosaura Esteve-Puig
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,Present Address: MAJ3 Capital S.L, Barcelona, 08018 Spain
| | - Sara García-Ortega
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Yuxin Ding
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Sara Sánchez-Redondo
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,grid.7719.80000 0000 8700 1153Present Address: Microenvironment & Metastasis Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Berta Ferrer
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,grid.411083.f0000 0001 0675 8654Anatomy Pathology Department, Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Javier Hernandez-Losa
- grid.411083.f0000 0001 0675 8654Anatomy Pathology Department, Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Francesc Canals
- grid.411083.f0000 0001 0675 8654Proteomics Laboratory, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, 08035 Spain
| | - Anna Manzano
- grid.418284.30000 0004 0427 2257Department of Physiological Sciences, University of Barcelona, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Aura Navarro-Sabaté
- grid.418284.30000 0004 0427 2257Department of Physiological Sciences, University of Barcelona, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Ramón Bartrons
- grid.418284.30000 0004 0427 2257Department of Physiological Sciences, University of Barcelona, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Oscar Yanes
- grid.410367.70000 0001 2284 9230Universitat Rovira i Virgili, Department of Electronic Engineering, IISPV, Tarragona, Spain ,grid.413448.e0000 0000 9314 1427CIBER on Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Mileidys Pérez-Alea
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,Present Address: Advance Biodesign, 69800 Saint-Priest, France
| | - Eva Muñoz-Couselo
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,grid.411083.f0000 0001 0675 8654Clinical Oncology Program, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Vicenç Garcia-Patos
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,grid.411083.f0000 0001 0675 8654Dermatology Department, Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Juan A. Recio
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| |
Collapse
|