1
|
Arshadi-Bidgoli M, Mortazavian SMM. Polycross breeding enhances cumin quality and drought tolerance for sustainable agriculture. Sci Rep 2025; 15:18927. [PMID: 40442200 PMCID: PMC12122853 DOI: 10.1038/s41598-025-03668-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2025] [Accepted: 05/21/2025] [Indexed: 06/02/2025] Open
Abstract
The yield and quality of cumin (Cuminum cyminum L.) medicinal plant are significantly affected by water stress. Producing a synthetic variety of cumin using polycross breeding can be an appropriate method for optimizing the biosynthesis of metabolites to create high-yielding, drought-tolerant plants with higher metabolite content and antioxidant properties. Therefore, a comprehensive study was conducted to evaluate the impact of drought stress on seed yield, metabolite content, antioxidant properties of ethanolic, methanolic, and aqueous extracts, essential oil content and composition, fatty acid composition, and physical seed traits of the synthetic variety of cumin in comparison with several parental genotypes over two growing seasons under normal irrigation (100% field capacity) and drought stress (30% field capacity) conditions. The results showed a 79.58%, 12.78% and 12.89% increase in seed yield, protein and carbohydrate content of the synthetic cultivar compared to the average of the parental genotypes under drought stress conditions. The decreasing effect of drought stress on the physical traits of seeds including 1000-seed weight, area, circumference, length and width of the seed in the synthetic cultivar (6.31, 20.84, 17.56, 9.44 and 7.45% respectively) was much less than that of the parental genotypes (9.67, 31.82, 18.56, 19.76 and 11.99% respectively). Under drought stress conditions, the synthetic cultivar had the highest essential oil content (3.11%) and oil content (12.04%). Also, in the synthetic variety, the amount of the main active compound of the essential oil (cumin aldehyde) and oil (oleic acid) also increased more due to drought stress. The synthetic variety had higher content of secondary metabolites (phenol, flavonol and flavonoid) and antioxidant properties, especially under drought stress conditions. Ethanol was also identified as the most suitable solvent for the extraction of polyphenolic compounds and antioxidant power. These findings suggest that producing a synthetic variety can be a suitable option for breeding high-quality cumin in arid regions. The results of this study can be utilized in breeding programs to develop drought-tolerant cumin varieties and other related plants with high yield and quality.
Collapse
Affiliation(s)
- Mahdieh Arshadi-Bidgoli
- Department of Agronomy Sciences and Plant Breeding, College of Aburaihan, University of Tehran, Tehran, Iran
| | | |
Collapse
|
2
|
Jiang X, Wang T, Zhao B, Sun H, Dong Y, Ma Y, Li Z, Wu Y, Wang K, Guan X, Long B, Qin L, Shi W, Shi L, He Q, Liu W, Li M, Xiao L, Zhou C, Sun H, Yang J, Guan J, Zhou H, Yu Z, Jiao Z. KRAS G12D-driven pentose phosphate pathway remodeling imparts a targetable vulnerability synergizing with MRTX1133 for durable remissions in PDAC. Cell Rep Med 2025; 6:101966. [PMID: 39970873 PMCID: PMC11866490 DOI: 10.1016/j.xcrm.2025.101966] [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: 06/24/2024] [Revised: 11/04/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025]
Abstract
The KRASG12D inhibitor MRTX1133 shows the potential to revolutionize the treatment paradigm for pancreatic ductal adenocarcinoma (PDAC), yet presents challenges. Our findings indicate that KRASG12D remodels a pentose phosphate pathway (PPP)-dominant central carbon metabolism pattern, facilitating malignant progression and resistance to MRTX1133 in PDAC. Mechanistically, KRASG12D drives excessive degradation of p53 and glucose-6-phosphate dehydrogenase (G6PD)-mediated PPP reprogramming through retinoblastoma (Rb)/E2F1/p53 axis-regulated feedback loops that amplify ubiquitin-conjugating enzyme E2T (UBE2T) transcription. Genetic ablation or pharmacological inhibition of UBE2T significantly suppresses PDAC progression and potentiates MRTX1133 efficacy. Leveraging structure advantages of the UBE2T inhibitor pentagalloylglucose (PGG), we develop a self-assembling nano co-delivery system with F-127, PGG, and MRTX1133. This system enhances the efficacy of PGG and MRTX1133, achieving durable remissions (85% overall response rate) and long-term survival (100% progression-free survival) in patient-derived xenografts and spontaneous PDAC mice. This study reveals the role of KRASG12D-preferred PPP reprogramming in MRTX1133 resistance and proposes a potentially therapeutic strategy for KRASG12D-mutated PDAC.
Collapse
Affiliation(s)
- Xiangyan Jiang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Tao Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Bin Zhao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Haonan Sun
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Yuman Dong
- Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China; State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Yong Ma
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Zhigang Li
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Yuxia Wu
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Keshen Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Xiaoying Guan
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Bo Long
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Long Qin
- Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Wengui Shi
- Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Lei Shi
- School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Qichen He
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Wenbo Liu
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Mingdou Li
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Lixia Xiao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Chengliang Zhou
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Hui Sun
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Jing Yang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Junhong Guan
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Huinian Zhou
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Zeyuan Yu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Zuoyi Jiao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China.
| |
Collapse
|
3
|
Wu R, Zhu H, He Q, Yuan T, Yang B. Metabolic reprogramming in KRAS-mutant cancers: Proven targetable vulnerabilities and potential therapeutic strategies. Drug Discov Today 2024; 29:104220. [PMID: 39481592 DOI: 10.1016/j.drudis.2024.104220] [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: 06/04/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Kras (Ki-ras2 Kirsten rat sarcoma viral oncogene homolog), one of the most frequently mutated oncogenes in the human genome, is considered 'untargetable'. Although specific KRASG12C inhibitors have been developed, their overall impact is limited, highlighting the need for further research on targeting KRAS-mutant cancers. Metabolic abnormalities are key hallmarks of cancer, with KRAS-driven tumors exhibiting traits like glycolysis upregulation, glutamine addiction, lipid droplet accumulation, highly active macropinocytosis, and metabolic reprogramming-associated tumor microenvironment remodeling. Targeting these unique metabolic characteristics offers a promising strategy for new cancer treatments. This review summarizes recent advances in our understanding of the metabolic network in KRAS-mutated tumor cells, discusses potential targetable vulnerabilities, and outlines clinical developments in relevant therapies, while also addressing challenges to improve strategies against these aggressive cancers.
Collapse
Affiliation(s)
- Ruilin Wu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hong Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Yuan
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China.
| | - Bo Yang
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Pathak T, Benson JC, Johnson MT, Xin P, Abdelnaby AE, Walter V, Koltun WA, Yochum GS, Hempel N, Trebak M. Loss of STIM2 in colorectal cancer drives growth and metastasis through metabolic reprogramming and PERK-ATF4 endoplasmic reticulum stress pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560521. [PMID: 37873177 PMCID: PMC10592933 DOI: 10.1101/2023.10.02.560521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The endoplasmic reticulum (ER) stores large amounts of calcium (Ca2+), and the controlled release of ER Ca2+ regulates a myriad of cellular functions. Although altered ER Ca2+ homeostasis is known to induce ER stress, the mechanisms by which ER Ca2+ imbalance activate ER stress pathways are poorly understood. Stromal-interacting molecules STIM1 and STIM2 are two structurally homologous ER-resident Ca2+ sensors that synergistically regulate Ca2+ influx into the cytosol through Orai Ca2+ channels for subsequent signaling to transcription and ER Ca2+ refilling. Here, we demonstrate that reduced STIM2, but not STIM1, in colorectal cancer (CRC) is associated with poor patient prognosis. Loss of STIM2 causes SERCA2-dependent increase in ER Ca2+, increased protein translation and transcriptional and metabolic rewiring supporting increased tumor size, invasion, and metastasis. Mechanistically, STIM2 loss activates cMyc and the PERK/ATF4 branch of ER stress in an Orai-independent manner. Therefore, STIM2 and PERK/ATF4 could be exploited for prognosis or in targeted therapies to inhibit CRC tumor growth and metastasis.
Collapse
Affiliation(s)
- Trayambak Pathak
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - J. Cory Benson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- UPMC Hillman Cancer Center. University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Martin T. Johnson
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Ping Xin
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Ahmed Emam Abdelnaby
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Vonn Walter
- Department of Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States
- Penn State Cancer Institute. The Pennsylvania State University College of Medicine, Hershey, United States
| | - Walter A. Koltun
- Department of Surgery, Division of Colon and Rectal Surgery, The Pennsylvania State University College of Medicine, Hershey, United States
| | - Gregory S. Yochum
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, United States
- Department of Surgery, Division of Colon and Rectal Surgery, The Pennsylvania State University College of Medicine, Hershey, United States
| | - Nadine Hempel
- UPMC Hillman Cancer Center. University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Mohamed Trebak
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- UPMC Hillman Cancer Center. University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
5
|
TeSlaa T, Ralser M, Fan J, Rabinowitz JD. The pentose phosphate pathway in health and disease. Nat Metab 2023; 5:1275-1289. [PMID: 37612403 PMCID: PMC11251397 DOI: 10.1038/s42255-023-00863-2] [Citation(s) in RCA: 173] [Impact Index Per Article: 86.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 07/12/2023] [Indexed: 08/25/2023]
Abstract
The pentose phosphate pathway (PPP) is a glucose-oxidizing pathway that runs in parallel to upper glycolysis to produce ribose 5-phosphate and nicotinamide adenine dinucleotide phosphate (NADPH). Ribose 5-phosphate is used for nucleotide synthesis, while NADPH is involved in redox homoeostasis as well as in promoting biosynthetic processes, such as the synthesis of tetrahydrofolate, deoxyribonucleotides, proline, fatty acids and cholesterol. Through NADPH, the PPP plays a critical role in suppressing oxidative stress, including in certain cancers, in which PPP inhibition may be therapeutically useful. Conversely, PPP-derived NADPH also supports purposeful cellular generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) for signalling and pathogen killing. Genetic deficiencies in the PPP occur relatively commonly in the committed pathway enzyme glucose-6-phosphate dehydrogenase (G6PD). G6PD deficiency typically manifests as haemolytic anaemia due to red cell oxidative damage but, in severe cases, also results in infections due to lack of leucocyte oxidative burst, highlighting the dual redox roles of the pathway in free radical production and detoxification. This Review discusses the PPP in mammals, covering its roles in biochemistry, physiology and disease.
Collapse
Affiliation(s)
- Tara TeSlaa
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Markus Ralser
- Department of Biochemistry, Charité Universitätsmedizin, Berlin, Germany
- The Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Jing Fan
- Morgride Institute for Research, Madison, WI, USA
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua D Rabinowitz
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
- Department of Chemistry, Princeton University, Princeton, NJ, USA.
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA.
| |
Collapse
|
6
|
Role of SIRT5 in cancer. Friend or Foe? Biochimie 2023; 209:131-141. [PMID: 36813074 DOI: 10.1016/j.biochi.2023.02.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023]
Abstract
Cancer is one of the main diseases currently afflicting mankind, being difficult to treat and generating thousands of deaths per year. As a result, researchers around the world are constantly searching for new therapeutic strategies to increase the survival rate of patients. In this regard, SIRT5 may be a promising therapeutic target due to its involvement in many metabolic pathways. Notably, SIRT5 has a dual role in the context of cancer, being able to act as a tumor suppressor in some types of cancer and behaving as an oncogene in others. Interestingly, the performance of SIRT5 is not specific and is highly dependent on the cellular context. As a tumor suppressor, SIRT5 prevents the Warburg effect, increases protection against ROS and reduces cell proliferation and metastasis, while as an oncogene it has the opposite effects as well as increasing resistance to chemotherapeutics and/or radiation. In this way, the aim of this work was to identify in which cancers SIRT5 has beneficial effects and in which deleterious ones based on their molecular characteristics. Furthermore, it was analyzed whether it is feasible to use this protein as a therapeutic target, either enhancing its activity or inhibiting it as appropriate.
Collapse
|
7
|
Mert-Ozupek N, Calibasi-Kocal G, Olgun N, Basbinar Y, Cavas L, Ellidokuz H. In-silico molecular interactions among the secondary metabolites of Caulerpa spp. and colorectal cancer targets. Front Chem 2022; 10:1046313. [PMID: 36561138 PMCID: PMC9763605 DOI: 10.3389/fchem.2022.1046313] [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: 09/16/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Caulerpa spp. secrete more than thirty different bioactive chemicals which have already been used in cancer treatment research since they play a pivotal role in cancer metabolism. Colorectal cancer is one of the most common cancer types, thus using novel and effective chemicals for colorectal cancer treatment is crucial. In the cheminformatics pipeline of this study, ADME-Tox and drug-likeness tests were performed for filtering the secondary metabolites of Caulerpa spp. The ligands which were selected from the ADME test were used for in silico molecular docking studies against the enzymes of the oxidative branch of the pentose phosphate pathway (glucose-6-phosphate dehydrogenase and 6-phosphoglutarate dehydrogenase), which is of great importance for colorectal cancer, by using AutoDock Vina. Pharmacophore modeling was carried out to align the molecules. Molecular dynamic simulations were performed for each target to validate the molecular docking studies and binding free energies were calculated. According to the ADME test results, 13 different secondary metabolites were selected as potential ligands. Molecular docking studies revealed that vina scores of caulerpin and monomethyl caulerpinate for G6PDH were found as -10.6 kcal mol-1, -10.5 kcal mol-1, respectively. Also, the vina score of caulersin for 6PGD was found as -10.7 kcal mol-1. The highest and the lowest binding free energies were calculated for monomethyl caulerpinate and caulersin, respectively. This in silico study showed that caulerpin, monomethyl caulerpinate, and caulersin could be evaluated as promising marine phytochemicals against pentose phosphate pathway enzymes and further studies are recommended to investigate the detailed activity of these secondary metabolites on these targets.
Collapse
Affiliation(s)
- Nazli Mert-Ozupek
- Department of Basic Oncology, Institute of Health Sciences, Dokuz Eylül University, İzmir, Türkiye
| | - Gizem Calibasi-Kocal
- Department of Translational Oncology, Institute of Oncology, Dokuz Eylül University, İzmir, Türkiye
| | - Nur Olgun
- Department of Pediatric Oncology, Institute of Oncology, Dokuz Eylül University, İzmir, Türkiye
| | - Yasemin Basbinar
- Department of Translational Oncology, Institute of Oncology, Dokuz Eylül University, İzmir, Türkiye
| | - Levent Cavas
- Department of Chemistry, Faculty of Sciences, Dokuz Eylül University, İzmir, Türkiye
| | - Hulya Ellidokuz
- Department of Preventive Oncology, Institute of Oncology, Dokuz Eylül University, İzmir, Türkiye,*Correspondence: Hulya Ellidokuz,
| |
Collapse
|
8
|
Fu Y, Yu J, Li F, Ge S. Oncometabolites drive tumorigenesis by enhancing protein acylation: from chromosomal remodelling to nonhistone modification. J Exp Clin Cancer Res 2022; 41:144. [PMID: 35428309 PMCID: PMC9013066 DOI: 10.1186/s13046-022-02338-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/21/2022] [Indexed: 02/02/2023] Open
Abstract
AbstractMetabolites are intermediate products of cellular metabolism catalysed by various enzymes. Metabolic remodelling, as a biochemical fingerprint of cancer cells, causes abnormal metabolite accumulation. These metabolites mainly generate energy or serve as signal transduction mediators via noncovalent interactions. After the development of highly sensitive mass spectrometry technology, various metabolites were shown to covalently modify proteins via forms of lysine acylation, including lysine acetylation, crotonylation, lactylation, succinylation, propionylation, butyrylation, malonylation, glutarylation, 2-hydroxyisobutyrylation and β-hydroxybutyrylation. These modifications can regulate gene expression and intracellular signalling pathways, highlighting the extensive roles of metabolites. Lysine acetylation is not discussed in detail in this review since it has been broadly investigated. We focus on the nine aforementioned novel lysine acylations beyond acetylation, which can be classified into two categories: histone acylations and nonhistone acylations. We summarize the characteristics and common functions of these acylation types and, most importantly, provide a glimpse into their fine-tuned control of tumorigenesis and potential value in tumour diagnosis, monitoring and therapy.
Collapse
|
9
|
Papadaki S, Magklara A. Regulation of Metabolic Plasticity in Cancer Stem Cells and Implications in Cancer Therapy. Cancers (Basel) 2022; 14:5912. [PMID: 36497394 PMCID: PMC9741285 DOI: 10.3390/cancers14235912] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
Cancer stem cells (CSCs), a subpopulation of tumor cells with self-renewal capacity, have been associated with tumor initiation, progression, and therapy resistance. While the bulk of tumor cells mainly use glycolysis for energy production, CSCs have gained attention for their ability to switch between glycolysis and oxidative phosphorylation, depending on their energy needs and stimuli from their microenvironment. This metabolic plasticity is mediated by signaling pathways that are also implicated in the regulation of CSC properties, such as the Wnt/β-catenin, Notch, and Hippo networks. Two other stemness-associated processes, autophagy and hypoxia, seem to play a role in the metabolic switching of CSCs as well. Importantly, accumulating evidence has linked the metabolic plasticity of CSCs to their increased resistance to treatment. In this review, we summarize the metabolic signatures of CSCs and the pathways that regulate them; we especially highlight research data that demonstrate the metabolic adaptability of these cells and their role in stemness and therapy resistance. As the development of drug resistance is a major challenge for successful cancer treatment, the potential of specific elimination of CSCs through targeting their metabolism is of great interest and it is particularly examined.
Collapse
Affiliation(s)
- Styliani Papadaki
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
| | - Angeliki Magklara
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
- Biomedical Research Institute–Foundation for Research and Technology, 45110 Ioannina, Greece
- Institute of Biosciences, University Research Center of Ioannina (URCI), 45110 Ioannina, Greece
| |
Collapse
|
10
|
Exploring the Interplay between Metabolism and Tumor Microenvironment Based on Four Major Metabolism Pathways in Colon Adenocarcinoma. JOURNAL OF ONCOLOGY 2022; 2022:2159794. [PMID: 35747126 PMCID: PMC9213191 DOI: 10.1155/2022/2159794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/11/2022] [Accepted: 05/14/2022] [Indexed: 11/17/2022]
Abstract
Tumor metabolism plays a critical role in tumor progression. However, the interaction between metabolism and tumor microenvironment (TME) has not been comprehensively revealed in colon adenocarcinoma (COAD). We used unsupervised consensus clustering to establish three molecular subtypes (clusters) based on the enrichment score of four major metabolism pathways in TCGA-COAD dataset. GSE17536 was used as a validation dataset. Single-cell RNA sequencing data (GSE161277) was employed to further verify the reliability of subtyping and characterize the correlation between metabolism and TME. Three clusters were identified and they performed distinct prognosis and molecular features. Clust3 had the worst overall survival and the highest enrichment score of glycolysis. 86 differentially expressed genes (DEGs) were identified, in which 11 DEGs were associated with favorable prognosis and 75 DEGs were associated with poor prognosis. Striking correlations were observed between hypoxia and glycolysis, clust3 and hypoxia, and clust3 and angiogenesis (P < 0.001).We constructed a molecular subtyping system which was effective and reliable for predicting COAD prognosis. The 86 identified key DEGs may be greatly involved in COAD progression, and they provide new perspectives and directions for further understanding the mechanism of metabolism in promoting aggressive phenotype by interacting with TME.
Collapse
|
11
|
Liu L, Weng Y, Fang J, Zhao Z, Du S. Understanding the effect of GO on nitrogen assimilation in wheat through transcriptomics and metabolic process analysis. CHEMOSPHERE 2022; 296:134000. [PMID: 35192852 DOI: 10.1016/j.chemosphere.2022.134000] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/12/2022] [Accepted: 02/13/2022] [Indexed: 06/14/2023]
Abstract
The extensive use of graphene oxide (GO) has resulted in its inevitable entry into the environment. It has been established that GO is detrimental to nitrogen accumulation in plants, as nitrogen is one of the most important nutrient for plant growth. However, its influence on nitrogen assimilation has not yet been investigated comprehensively. Based on the analysis of transcriptomics and nitrogen metabolites, this study showed that 400 mg L-1 GO exposure downregulated most of the genes encoding nitrogen-assimilating enzymes, including nitrate reductase (NR), glutamine synthetase (GS), glutamate synthase (GOGAT), and glutamate dehydrogenase (GDH). The activities of the above enzymes in wheat roots were also reduced with GO addition, and the activities of NR and GS, the rate-limiting enzymes of nitrate and ammonium assimilation, were approximately 75% and 76% lower with 400 mg L-1 GO supply, respectively, compared to those upon control treatment. Correspondingly, GO appears to exert a negative effect on multiple nitrogen assimilation products, including nitrous nitrogen, ammonium nitrogen, glutamine, glutamate, and soluble protein. In summary, this study showed that GO has adverse effects on the nitrogen assimilation of plants, and NR and GS are the most affected sites. Our findings would provide deeper insights into the physiological and molecular mechanisms underlying GO phytotoxicity.
Collapse
Affiliation(s)
- Lijuan Liu
- Key Laboratory of Pollution Exposure and Health Intervention Technology of Zhejiang Province, Interdisciplinary Research Academy (IRA), Zhejiang Shuren University, Hangzhou, 310015, China
| | - Yineng Weng
- College of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Jin Fang
- College of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Zijing Zhao
- Key Laboratory of Pollution Exposure and Health Intervention Technology of Zhejiang Province, Interdisciplinary Research Academy (IRA), Zhejiang Shuren University, Hangzhou, 310015, China
| | - Shaoting Du
- Key Laboratory of Pollution Exposure and Health Intervention Technology of Zhejiang Province, Interdisciplinary Research Academy (IRA), Zhejiang Shuren University, Hangzhou, 310015, China.
| |
Collapse
|
12
|
Barata T, Vieira V, Rodrigues R, Neves RPD, Rocha M. Reconstruction of tissue-specific genome-scale metabolic models for human cancer stem cells. Comput Biol Med 2021; 142:105177. [PMID: 35026576 DOI: 10.1016/j.compbiomed.2021.105177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 02/07/2023]
Abstract
Cancer Stem Cells (CSCs) contribute to cancer aggressiveness, metastasis, chemo/radio-therapy resistance, and tumor recurrence. Recent studies emphasized the importance of metabolic reprogramming of CSCs for the maintenance and progression of the cancer phenotype through both the fulfillment of the energetic requirements and the supply of substrates fundamental for fast-cell growth, as well as through metabolite-induced epigenetic regulation. Therefore, it is of paramount importance to develop therapeutic strategies tailored to target the metabolism of CSCs. In this work, we built computational Genome-Scale Metabolic Models (GSMMs) for CSCs of different tissues. Flux simulations were then used to predict metabolic phenotypes, identify potential therapeutic targets, and spot already-known Transcription Factors (TFs), miRNAs and antimetabolites that could be used as part of drug repurposing strategies against cancer. Results were in accordance with experimental evidence, provided insights of new metabolic mechanisms for already known agents, and allowed for the identification of potential new targets and compounds that could be interesting for further in vitro and in vivo validation.
Collapse
Affiliation(s)
- Tânia Barata
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Vítor Vieira
- Centre of Biological Engineering, University of Minho - Campus de Gualtar, Braga, Portugal
| | - Rúben Rodrigues
- Centre of Biological Engineering, University of Minho - Campus de Gualtar, Braga, Portugal
| | - Ricardo Pires das Neves
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-517, Coimbra, Portugal; IIIUC-Institute of Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal.
| | - Miguel Rocha
- Centre of Biological Engineering, University of Minho - Campus de Gualtar, Braga, Portugal; Department of Informatics, University of Minho.
| |
Collapse
|
13
|
Paul B, Kysenius K, Hilton JB, Jones MWM, Hutchinson RW, Buchanan DD, Rosty C, Fryer F, Bush AI, Hergt JM, Woodhead JD, Bishop DP, Doble PA, Hill MM, Crouch PJ, Hare DJ. An integrated mass spectrometry imaging and digital pathology workflow for objective detection of colorectal tumours by unique atomic signatures. Chem Sci 2021; 12:10321-10333. [PMID: 34476052 PMCID: PMC8386113 DOI: 10.1039/d1sc02237g] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/29/2021] [Indexed: 12/21/2022] Open
Abstract
Tumours are abnormal growths of cells that reproduce by redirecting essential nutrients and resources from surrounding tissue. Changes to cell metabolism that trigger the growth of tumours are reflected in subtle differences between the chemical composition of healthy and malignant cells. We used LA-ICP-MS imaging to investigate whether these chemical differences can be used to spatially identify tumours and support detection of primary colorectal tumours in anatomical pathology. First, we generated quantitative LA-ICP-MS images of three colorectal surgical resections with case-matched normal intestinal wall tissue and used this data in a Monte Carlo optimisation experiment to develop an algorithm that can classify pixels as tumour positive or negative. Blinded testing and interrogation of LA-ICP-MS images with micrographs of haematoxylin and eosin stained and Ki67 immunolabelled sections revealed Monte Carlo optimisation accurately identified primary tumour cells, as well as returning false positive pixels in areas of high cell proliferation. We analysed an additional 11 surgical resections of primary colorectal tumours and re-developed our image processing method to include a random forest regression machine learning model to correctly identify heterogenous tumours and exclude false positive pixels in images of non-malignant tissue. Our final model used over 1.6 billion calculations to correctly discern healthy cells from various types and stages of invasive colorectal tumours. The imaging mass spectrometry and data analysis methods described, developed in partnership with clinical cancer researchers, have the potential to further support cancer detection as part of a comprehensive digital pathology approach to cancer care through validation of a new chemical biomarker of tumour cells.
Collapse
Affiliation(s)
- Bence Paul
- School of Geography, Earth and Atmospheric Sciences, The University of Melbourne Parkville Victoria 3010 Australia
| | - Kai Kysenius
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, The University of Melbourne Parkville Victoria 3010 Australia
| | - James B Hilton
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, The University of Melbourne Parkville Victoria 3010 Australia
| | - Michael W M Jones
- Central Analytical Research Facility, Queensland University of Technology Brisbane Queensland 4000 Australia
| | | | - Daniel D Buchanan
- Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne Parkville Victoria 3010 Australia
- University of Melbourne Centre for Cancer Research, The University of Melbourne Parkville Victoria 3010 Australia
- Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital Melbourne Victoria 3000 Australia
| | - Christophe Rosty
- Envoi Pathology Brisbane Queensland 4000 Australia
- Faculty of Medicine, The University of Queensland Brisbane Queensland 4000 Australia
- Department of Clinical Pathology, The University of Melbourne Parkville Victoria 3010 Australia
| | - Fred Fryer
- Agilent Technologies Australia Mulgrave Victoria 3170 Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre at the Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville Victoria 3010 Australia
| | - Janet M Hergt
- School of Geography, Earth and Atmospheric Sciences, The University of Melbourne Parkville Victoria 3010 Australia
| | - Jon D Woodhead
- School of Geography, Earth and Atmospheric Sciences, The University of Melbourne Parkville Victoria 3010 Australia
| | - David P Bishop
- Atomic Medicine Initiative, University of Technology Sydney Broadway NSW 2007 Australia
| | - Philip A Doble
- Atomic Medicine Initiative, University of Technology Sydney Broadway NSW 2007 Australia
| | - Michelle M Hill
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland Herston Qld 4006 Australia
- QIMR Berghofer Medical Research Institute Herston Queensland 4006 Australia
| | - Peter J Crouch
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, The University of Melbourne Parkville Victoria 3010 Australia
| | - Dominic J Hare
- Melbourne Dementia Research Centre at the Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville Victoria 3010 Australia
- Atomic Medicine Initiative, University of Technology Sydney Broadway NSW 2007 Australia
- School of BioSciences, The University of Melbourne Parkville Victoria 3010 Australia
- Monash eResearch Centre, Monash University Clayton Victoria 3800 Australia
| |
Collapse
|
14
|
Sustained oxidative stress instigates differentiation of cancer stem cells into tumor endothelial cells: Pentose phosphate pathway, reactive oxygen species and autophagy crosstalk. Biomed Pharmacother 2021; 139:111643. [PMID: 33945913 DOI: 10.1016/j.biopha.2021.111643] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/08/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor angiogenesis plays a vital role in tumor growth and metastasis. It is proven that in tumor vasculature, endothelial cells (ECs) originate from a small population of cancer cells introduced as cancer stem cells (CSCs). Autophagy has a vital role in ECs differentiation from CSCs and tumor angiogenesis. High levels of reactive oxygen species (ROS) increased autophagy by inhibition of glucose-6-phosphate dehydrogenase (G6PD) and inactivation of the pentose phosphate pathway (PPP). Previously, we suggested that cancer cells initially increase the glycolysis rate when encountering ROS, then the metabolic balance is changed from glycolysis to PPP, following the continuation of oxidative stress. In this study, we investigate the possible role of persistent oxidative stress in the differentiation of CSCs into tumor ECs by relying on the relationship between the ROS, PPP and autophagy. Because tumor angiogenesis plays an important role in the growth and development of cancer, understanding the mechanisms involved in differentiating ECs from CSCs can help find promising treatments for cancer.
Collapse
|
15
|
Khan T, Kryza T, Lyons NJ, He Y, Hooper JD. The CDCP1 Signaling Hub: A Target for Cancer Detection and Therapeutic Intervention. Cancer Res 2021; 81:2259-2269. [PMID: 33509939 DOI: 10.1158/0008-5472.can-20-2978] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/22/2020] [Accepted: 01/22/2021] [Indexed: 11/16/2022]
Abstract
CUB-domain containing protein 1 (CDCP1) is a type I transmembrane glycoprotein that is upregulated in malignancies of the breast, lung, colorectum, ovary, kidney, liver, pancreas, and hematopoietic system. Here, we discuss CDCP1 as an important hub for oncogenic signaling and its key roles in malignant transformation and summarize approaches focused on exploiting it for cancer diagnosis and therapy. Elevated levels of CDCP1 are associated with progressive disease and markedly poorer survival. Predominantly located on the cell surface, CDCP1 lies at the nexus of key tumorigenic and metastatic signaling cascades, including the SRC/PKCδ, PI3K/AKT, WNT, and RAS/ERK axes, the oxidative pentose phosphate pathway, and fatty acid oxidation, making important functional contributions to cancer cell survival and growth, metastasis, and treatment resistance. These findings have stimulated the development of agents that target CDCP1 for detection and treatment of a range of cancers, and results from preclinical models suggest that these approaches could be efficacious and have manageable toxicity profiles.
Collapse
Affiliation(s)
- Tashbib Khan
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Thomas Kryza
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Nicholas J Lyons
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Yaowu He
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - John D Hooper
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia.
| |
Collapse
|
16
|
Polat IH, Tarrado-Castellarnau M, Bharat R, Perarnau J, Benito A, Cortés R, Sabatier P, Cascante M. Oxidative Pentose Phosphate Pathway Enzyme 6-Phosphogluconate Dehydrogenase Plays a Key Role in Breast Cancer Metabolism. BIOLOGY 2021; 10:85. [PMID: 33498665 PMCID: PMC7911610 DOI: 10.3390/biology10020085] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023]
Abstract
The pentose phosphate pathway (PPP) plays an essential role in the metabolism of breast cancer cells for the management of oxidative stress and the synthesis of nucleotides. 6-phosphogluconate dehydrogenase (6PGD) is one of the key enzymes of the oxidative branch of PPP and is involved in nucleotide biosynthesis and redox maintenance status. Here, we aimed to analyze the functional importance of 6PGD in a breast cancer cell model. Inhibition of 6PGD in MCF7 reduced cell proliferation and showed a significant decrease in glucose consumption and an increase in glutamine consumption, resulting in an important alteration in the metabolism of these cells. No difference in reactive oxygen species (ROS) production levels was observed after 6PGD inhibition, indicating that 6PGD, in contrast to glucose 6-phosphate dehydrogenase, is not involved in redox balance. We found that 6PGD inhibition also altered the stem cell characteristics and mammosphere formation capabilities of MCF7 cells, opening new avenues to prevent cancer recurrance after surgery or chemotherapy. Moreover, inhibition of 6PGD via chemical inhibitor S3 resulted in an induction of senescence, which, together with the cell cycle arrest and apoptosis induction, might be orchestrated by p53 activation. Therefore, we postulate 6PGD as a novel therapeutic target to treat breast cancer.
Collapse
Affiliation(s)
- Ibrahim H. Polat
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain; (I.H.P.); (M.T.-C.); (R.B.); (J.P.); (A.B.); (R.C.)
- Equipe Environnement et Prédiction de la Santé des Populations, Laboratoire TIMC (UMR 5525), CHU de Grenoble, Université Grenoble Alpes, 38700 CEDEX La Tronche, France;
- Department of Medicine, Hematology/Oncology, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Míriam Tarrado-Castellarnau
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain; (I.H.P.); (M.T.-C.); (R.B.); (J.P.); (A.B.); (R.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), 28001 Madrid, Spain
| | - Rohit Bharat
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain; (I.H.P.); (M.T.-C.); (R.B.); (J.P.); (A.B.); (R.C.)
| | - Jordi Perarnau
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain; (I.H.P.); (M.T.-C.); (R.B.); (J.P.); (A.B.); (R.C.)
| | - Adrian Benito
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain; (I.H.P.); (M.T.-C.); (R.B.); (J.P.); (A.B.); (R.C.)
- Division of Cancer, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Roldán Cortés
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain; (I.H.P.); (M.T.-C.); (R.B.); (J.P.); (A.B.); (R.C.)
| | - Philippe Sabatier
- Equipe Environnement et Prédiction de la Santé des Populations, Laboratoire TIMC (UMR 5525), CHU de Grenoble, Université Grenoble Alpes, 38700 CEDEX La Tronche, France;
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain; (I.H.P.); (M.T.-C.); (R.B.); (J.P.); (A.B.); (R.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), 28001 Madrid, Spain
| |
Collapse
|
17
|
Ou T, Yang W, Li W, Lu Y, Dong Z, Zhu H, Sun X, Dong Z, Weng X, Chang S, Li H, Li Y, Qiu Z, Hu K, Sun A, Ge J. SIRT5 deficiency enhances the proliferative and therapeutic capacities of adipose-derived mesenchymal stem cells via metabolic switching. Clin Transl Med 2020; 10:e172. [PMID: 32997407 PMCID: PMC7510333 DOI: 10.1002/ctm2.172] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have therapeutic potential for multiple ischemic diseases. However, in vitro expansion of MSCs before clinical application leads to metabolic reprogramming from glycolysis to oxidative phosphorylation, drastically impairing their proliferative and therapeutic capacities. This study aimed to define the regulatory effects of Sirtuin 5 (SIRT5) on the proliferative and therapeutic functions of adipose-derived MSCs (ADMSCs) during in vitro expansion. METHODS ADMSCs were isolated from wild-type (WT) and Sirt5-knockout (Sirt5-/- ) mice. Cell counting assay was used to investigate the proliferative capacities of the ADMSCs. Dihydroethidium and senescence-associated β-galactosidase stainings were used to measure intracellular ROS and senescence levels. Mass spectrometry was used to analyze protein succinylation. Oxygen consumption rates and extra cellular acidification rates were measured as indicators of mitochondrial respiration and glycolysis. Metabolic-related genes expression were verified by quantitative PCR and western blot. Hind limb ischemia mouse model was used to evaluate the therapeutic potentials of WT and Sirt5-/- ADSMCs. RESULTS SIRT5 protein levels were upregulated in ADMCs during in vitro expansion. Sirt5-/- ADMSCs exhibited a higher proliferation rate, delayed senescence, and reduced ROS accumulation. Furthermore, elevated protein succinylation levels were observed in Sirt5-/- ADMSCs, leading to the reduced activity of tricarboxylic acid cycle-related enzymes and attenuated mitochondrial respiration. Glucose uptake, glycolysis, and pentose phosphate pathway were elevated in Sirt5-/- ADMSCs. Inhibition of succinylation by glycine or re-expression of Sirt5 reversed the metabolic alterations in Sirt5-/- ADMSCs, thus abolishing their enhanced proliferative capacities. In the hind limb ischemia mouse model, SIRT5-/- ADMSCs transplantation enhanced blood flow recovery and angiogenesis compared with WT ADMSCs. CONCLUSIONS Our results indicate that SIRT5 deficiency during ADMSC culture expansion leads to reversed metabolic pattern, enhanced proliferative capacities, and improved therapeutic outcomes. These data suggest SIRT5 as a potential target to enhance the functional properties of MSCs for clinical application.
Collapse
Affiliation(s)
- Tiantong Ou
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Wenlong Yang
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Wenjia Li
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Yijing Lu
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
| | - Zheng Dong
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Hongming Zhu
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Xiaolei Sun
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Zhen Dong
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Xinyu Weng
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Suchi Chang
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Hua Li
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Yufan Li
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Zhiwei Qiu
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Kai Hu
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Aijun Sun
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
| | - Junbo Ge
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
| |
Collapse
|