1
|
Li S, Han H, Yang K, Li X, Ma L, Yang Z, Zhao YX. Emerging role of metabolic reprogramming in the immune microenvironment and immunotherapy of thyroid cancer. Int Immunopharmacol 2025; 144:113702. [PMID: 39602959 DOI: 10.1016/j.intimp.2024.113702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
The metabolic reprogramming of cancer cells is a hallmark of many malignancies. To meet the energy acquisition needs of tumor cells for rapid proliferation, tumor cells reprogram their nutrient metabolism, which is caused by the abnormal expression of transcription factors and signaling molecules related to energy metabolic pathways as well as the upregulation and downregulation of abnormal metabolic enzymes, receptors, and mediators. Thyroid cancer (TC) is the most common endocrine tumor, and immunotherapy has become the mainstream choice for clinical benefit after the failure of surgical, endocrine, and radioiodine therapies. TC change the tumor microenvironment (TME) through nutrient competition and metabolites, causing metabolic reprogramming of immune cells, profoundly changing immune cell function, and promoting immune evasion of tumor cells. A deeper understanding of how metabolic reprogramming alters the TME and controls immune cell fate and function will help improve the effectiveness of TC immunotherapy and patient outcomes. This paper aims to elucidate the metabolic communication that occurs between immune cells around TC and discusses how metabolic reprogramming in TC affects the immune microenvironment and the effectiveness of anti-cancer immunotherapy. Finally, targeting key metabolic checkpoints during metabolic reprogramming, combined with immunotherapy, is a promising strategy.
Collapse
Affiliation(s)
- Shouhua Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China.
| | - Hengtong Han
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Kaili Yang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China.
| | - Xiaoxiao Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China.
| | - Libin Ma
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Ze Yang
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Yong-Xun Zhao
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
2
|
Hassan MG, El-Sayyad GS, Abdel-Monem MO, Malash MN, Kishk MA, El Awady ME, El-Khonezy MI. Unravelling the outcome of L-glutaminase produced by Streptomyces sp. strain 5 M as an anti-neoplasm activity. Microb Cell Fact 2025; 24:4. [PMID: 39754133 PMCID: PMC11699688 DOI: 10.1186/s12934-024-02606-8] [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: 07/23/2024] [Accepted: 11/26/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Actinomycetes are a well-known example of a microbiological origin that may generate a wide variety of chemical structures. As excellent cell factories, these sources are able to manufacture medicines, agrochemicals, and enzymes that are crucial. RESULTS In this study, about 34 randomly selected Streptomyces isolates were discovered in soil, sediment, sea water, and other environments. Using a qualitative fast plate assay, they were tested for L-glutaminase production, and nine of them produced a significant amount of pink L-glutamine. Streptomyces sp. strain 5 M was identified by examining the 16S rRNA gene in the promising strain G8. A pH of 7.5, an incubation temperature of 40 °C, and the use of glucose and peptone as the carbon and nitrogen sources, respectively, produced the highest quantities of L-glutaminase. The molecular weight of the isolated L-glutaminase was estimated to be 52 kDa using SDS-PAGE analysis. At pH 7.5 and Temp., 40 °C, the isolated enzyme exhibited its highest levels of stability and activity. The isolated enzyme's Km and Vmax values were 2.62 mM and 10.20 U/ml, respectively. Strong toxicity against HepG-2, HeLa, and MCF-7 was observed due to the anticancer properties of the isolated L-glutaminase. CONCLUSION Our findings include the discovery of Streptomyces sp. strain 5 M, which yields a free L-glutaminase and maybe a possible applicant for extra pharmacological investigation as an antineoplastic drug.
Collapse
Affiliation(s)
- Mervat G Hassan
- Department of Botany and Microbiology, Faculty of Science, Benha University, Benha, 33516, Egypt
| | - Gharieb S El-Sayyad
- Medical Laboratory Technology Department, Faculty of Applied Health Sciences Technology, Badr University in Cairo (BUC), Cairo, Egypt.
- Department of Microbiology and Immunology, Faculty of Pharmacy, Galala University, Galala City, Suez, Egypt.
- Drug Microbiology Lab., Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt.
| | - Mohamed O Abdel-Monem
- Department of Botany and Microbiology, Faculty of Science, Benha University, Benha, 33516, Egypt
| | - Mohamed N Malash
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ahram Canadian University (ACU), Giza, Egypt
| | - Mona A Kishk
- Department of Botany and Microbiology, Faculty of Science, Benha University, Benha, 33516, Egypt
| | - Mohamed E El Awady
- Microbial Biotechnology Department, Biotechnology Research Institute, National Research Centre, El-Buhouth St. 33, Dokki, Cairo, Egypt
| | - Mohamed I El-Khonezy
- Molecular Biology Department, Biotechnology Research Institute, National Research Center, El-Buhouth St. 33, Dokki, P.O.12622, Giza, Egypt
| |
Collapse
|
3
|
Worral Wilfred Raj AS, Manoharan R. NUAKs promote mTOR/c-Myc-induced glucose and glutamine reprogramming for cell growth and metastasis in breast cancer cells. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167508. [PMID: 39270807 DOI: 10.1016/j.bbadis.2024.167508] [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: 06/23/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
Breast cancer progression and metastasis are closely connected to changes in glucose and glutamine metabolism. While Novel (nua) kinase family 1 (NUAK1) and Novel (nua) kinase family 2 (NUAK2), which are two members of the AMPK-related kinases, have been associated with breast tumorigenesis, their role in the metabolic reprogramming that occurs during breast cancer progression remains unclear. Our research uncovers that NUAKs expression is significantly higher in breast cancer tissues and cell lines, and it is positively related to glycolysis, the pentose phosphate pathway (PPP), glutamine metabolism, and a poor prognosis for breast cancer patients. We show that NUAKs significantly increase metabolic reprogramming, including aerobic glycolysis, PPP, and glutamine metabolism in triple negative breast cancer subtypes but only induce aerobic glycolysis and PPP in luminal breast cancer subtypes to meet the anabolic demands of rapidly dividing breast cancer cells. In contrast, the depletion of NUAKs has the opposite effect. Mechanistic insights reveal that NUAKs activate mammalian target of rapamycin (mTOR) signaling, which in turn upregulates the c-Myc transcription factor, a crucial regulator of glucose and glutamine metabolic gene expression. Moreover, we demonstrate that NUAKs enhance mTOR/c-Myc signaling pathways, leading to increased glucose and glutamine reprogramming, which supports rapid cell proliferation and metastatic potential in breast cancer cells. Importantly, pretreating breast cancer cells with mTOR inhibitors blocked the metabolic reprogramming and tumor-promoting effect of NUAK1/2. Therefore, targeting NUAKs may represent a novel therapeutic strategy for the treatment of breast cancer.
Collapse
Affiliation(s)
- Acily Skadon Worral Wilfred Raj
- Cell Signaling and Cancer Biology Laboratory, Department of Biochemistry, Guindy Campus, University of Madras, Chennai 600025, India
| | - Ravi Manoharan
- Cell Signaling and Cancer Biology Laboratory, Department of Biochemistry, Guindy Campus, University of Madras, Chennai 600025, India.
| |
Collapse
|
4
|
Xu X, Zhao H, Liu F, Xie Q, Yang J, Li S, Yang Z. Clinical Value of 18 F-(2S,4R)-4-Fluoroglutamine PET/CT in Glioma. Clin Nucl Med 2024:00003072-990000000-01458. [PMID: 39668484 DOI: 10.1097/rlu.0000000000005581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
PROPOSE The aim of this study was to evaluate the clinical application value of 18 F-FGln PET/CT in glioma. PATIENTS AND METHODS Patients with suspected gliomas by MRI were included in this study. Static and/or dynamic brain 18 F-FGln PET/CT was performed. The PET parameters SUV max , SUV mean , MTV, and TLG were evaluated. RESULTS Twenty-three patients were included in the analysis. Nineteen of 23 patients were positive for 18 F-FGln PET. The SUV max of high- and low-grade gliomas were 4.75 ± 2.21 and 1.00 ± 0.66 ( P < 0.001), respectively. FGln-PET SUV max , SUV mean , and TLG all showed statistically significant correlations with glioma grade, with correlation coefficients ( r ) of 0.667 ( P < 0.001), 0.693 ( P < 0.001), and 0.487 ( P = 0.021), respectively. Additionally, the SUV max , SUV mean , and TLG exhibited higher distinguishing performance for glioma grade by receiver operating characteristic curve analysis. The areas under the receiver operating characteristic curve of SUV max , SUV mean , and TLG were 0.976 (95% confidence interval [CI], 0.918-1) ( P = 0.002), 0.976 (95% CI, 0.918-1) ( P = 0.002), and 0.835 (95% CI, 0.628-1.000) ( P = 0.026), respectively. For glioma isocitrate dehydrogenase (IDH) mutation status, the SUV max of IDH wildtype and mutant glioma were 2.95 ± 1.99 and 6.13 ± 2.16 ( P = 0.005), respectively. The SUV mean and SUV max had good-to-satisfactory performance for IDH status with the area under the receiver operating characteristic curve of SUV max and SUV mean of 0.885 (95% CI, 0.734-1.000) ( P = 0.009) and 0.942 (95% CI, 0.828-1) ( P = 0.002). CONCLUSIONS Although we do not assert that 18 F-FGln PET/CT imaging is satisfactory in the differential diagnosis of glioma, we revealed its potential for identifying the stage of gliomas and the IDH mutation status and propose that glutamine-based PET imaging enables the assessment of metabolic nutrient uptake of gliomas to assist clinical diagnosis and treatment of patients.
Collapse
Affiliation(s)
- Xiaoxia Xu
- From the State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Heqian Zhao
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Futao Liu
- From the State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qing Xie
- From the State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jianhua Yang
- From the State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shouwei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Zhi Yang
- From the State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
5
|
Wu J, Qin T, Han W, Zhang C, Zhang X, Huang Z, Wu Y, Xu Y, Xu K, Ye W. GLS1-mediated glutamine metabolism mitigates oxidative stress-induced matrix degradation, ferroptosis, and senescence in nucleus pulposus cells by modulating Fe 2+ homeostasis. Free Radic Biol Med 2024:S0891-5849(24)01152-3. [PMID: 39710108 DOI: 10.1016/j.freeradbiomed.2024.12.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/26/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Intervertebral disc degeneration (IDD) is intricately linked to the pathogenesis of low back pain (LBP). The balance of nucleus pulposus (NP) cell and intervertebral disc (IVD) integrity is significantly supported by amino acid metabolism within an avascular milieu. However, the specific metabolic demands during the progression of IDD are not fully understood. Our study revealed that GLS1, a key enzyme that regulates glutamine metabolism, is key for mitigating NP cell ferroptosis, senescence, and IDD progression. Our findings show that GLS1 overexpression modulates glutamine metabolism, reducing NP cell matrix degradation, ferroptosis, and senescence. Mechanistically, GLS1 interacts with NFS1 and regulates ferrous ion (Fe2+) homeostasis. GLS1-driven glutamine metabolism facilitates acetyl-CoA production, which is important for the histone acetylation of NFS1. Thus, restoring GLS1 activity through gene overexpression to maintain Fe2+ homeostasis is a promising approach for mitigating matrix degradation, ferroptosis, and senescence and for rejuvenating intervertebral discs. Collectively, our data suggest a model in which GLS1-mediated glutamine metabolism is associated with NP cell matrix degradation, ferroptosis, and senescence and that NFS1 can be targeted to maintain Fe2+ homeostasis and ultimately revitalize intervertebral discs.
Collapse
Affiliation(s)
- Jiajun Wu
- Department of Orthopaedics, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510530; Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China
| | - Tianyu Qin
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China; Department of Spine Surgery, Orthopaedic, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University. The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Weitao Han
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China
| | - Chao Zhang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China
| | - Xiaohe Zhang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China
| | - Zhengqi Huang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China
| | - Yuliang Wu
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China
| | - Yichun Xu
- Department of Orthopaedics, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510530.
| | - Kang Xu
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China.
| | - Wei Ye
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China.
| |
Collapse
|
6
|
Hong J, Liu W, Xiao X, Gajendran B, Ben-David Y. Targeting pivotal amino acids metabolism for treatment of leukemia. Heliyon 2024; 10:e40492. [PMID: 39654725 PMCID: PMC11626780 DOI: 10.1016/j.heliyon.2024.e40492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024] Open
Abstract
Metabolic reprogramming is a crucial characteristic of cancer, allowing cancer cells to acquire metabolic properties that support their survival, immune evasion, and uncontrolled proliferation. Consequently, targeting cancer metabolism has become an essential therapeutic strategy. Abnormal amino acid metabolism is not only a key aspect of metabolic reprogramming but also plays a significant role in chemotherapy resistance and immune evasion, particularly in leukemia. Changes in amino acid metabolism in tumor cells are typically driven by a combination of signaling pathways and transcription factors. Current approaches to targeting amino acid metabolism in leukemia include inhibiting amino acid transporters, blocking amino acid biosynthesis, and depleting specific amino acids to induce apoptosis in leukemic cells. Different types of leukemic cells rely on the exogenous supply of specific amino acids, such as asparagine, glutamine, arginine, and tryptophan. Therefore, disrupting the supply of these amino acids may represent a vulnerability in leukemia. This review focuses on the pivotal role of amino acids in leukemia metabolism, their impact on leukemic stem cells, and their therapeutic potential.
Collapse
Affiliation(s)
- Jiankun Hong
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| | - Wuling Liu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| | - Xiao Xiao
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| | - Babu Gajendran
- Institute of Pharmacology and Biological Activity, Natural Products Research Center of Guizhou Province, Guiyang, Guizhou, 550014, PR China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550025, Guizhou Province, PR China
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| |
Collapse
|
7
|
Tang Q, Ren T, Bai P, Wang X, Zhao L, Zhong R, Sun G. Novel strategies to overcome chemoresistance in human glioblastoma. Biochem Pharmacol 2024; 230:116588. [PMID: 39461382 DOI: 10.1016/j.bcp.2024.116588] [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: 06/30/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Abstract
Temozolomide (TMZ) is currently the first-line chemotherapeutic agent for the treatment of glioblastoma multiforme (GBM). However, the inherent heterogeneity of GBM often results in suboptimal outcomes, particularly due to varying degrees of resistance to TMZ. Over the past several decades, O6-methylguanine-DNA methyltransferase (MGMT)-mediated DNA repair pathway has been extensively investigated as a target to overcome TMZ resistance. Nonetheless, the combination of small molecule covalent MGMT inhibitors with TMZ and other chemotherapeutic agents has frequently led to adverse clinical effects. Recently, additional mechanisms contributing to TMZ resistance have been identified, including epidermal growth factor receptor (EGFR) mutations, overactivation of intracellular signalling pathways, energy metabolism reprogramming or survival autophagy, and changes in tumor microenvironment (TME). These findings suggest that novel therapeutic strategies targeting these mechanisms hold promise for overcoming TMZ resistance in GBM patients. In this review, we summarize the latest advancements in understanding the mechanisms underlying intrinsic and acquired TMZ resistance. Additionally, we compile various small-molecule compounds with potential to mitigate chemoresistance in GBM. These mechanism-based compounds may enhance the sensitivity of GBM to TMZ and related chemotherapeutic agents, thereby improving overall survival rates in clinical practice.
Collapse
Affiliation(s)
- Qing Tang
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Ting Ren
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Peiying Bai
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Xin Wang
- Department of Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
| | - Lijiao Zhao
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Rugang Zhong
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Guohui Sun
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
8
|
Murcy F, Borowczyk C, Gourion-Arsiquaud S, Torrino S, Ouahrouche N, Barouillet T, Dussaud S, Couralet M, Vaillant N, Merlin J, Berquand A, Kaikkonen MU, McClelland RL, Tressel W, Stein J, Thorp EB, Bertero T, Barbry P, Bailly-Maitre B, Gautier EL, Karjalainen MK, Kettunen J, Duca L, Shea S, Yvan-Charvet L. GLS2 links glutamine metabolism and atherosclerosis by remodeling artery walls. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1454-1467. [PMID: 39562782 DOI: 10.1038/s44161-024-00566-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/23/2024] [Indexed: 11/21/2024]
Abstract
Metabolic dysregulation, including perturbed glutamine-glutamate homeostasis, is common among patients with cardiovascular diseases, but the underlying mechanisms remain largely unknown. Using the human MESA cohort, here we show that plasma glutamine-glutamate ratio is an independent risk factor for carotid plaque progression. Mice deficient in glutaminase-2 (Gls2), the enzyme that mediates hepatic glutaminolysis, developed accelerated atherosclerosis and susceptibility to catastrophic cardiac events, while Gls2 overexpression partially protected from disease progression. High-throughput transcriptional profiling and high-resolution structural biology imaging of aortas showed that Gls2 deficiency perturbed extracellular matrix composition and increased vessel stiffness. This results from an imbalance of glutamine- and glutamate-dependent cross-linked proteins within atherosclerotic lesions and cellular remodeling of plaques. Thus, hepatic glutaminolysis functions as a potent regulator of glutamine homeostasis, which affects the aortic wall structure during atherosclerotic plaque progression.
Collapse
Grants
- ERC2016COG724838 EC | EU Framework Programme for Research and Innovation H2020 | H2020 Priority Excellent Science | H2020 European Research Council (H2020 Excellent Science - European Research Council)
- 25-CE14-Glutacare Agence Nationale de la Recherche (French National Research Agency)
- 75N92020D00001 NHLBI NIH HHS
- 75N92020D00004 NHLBI NIH HHS
- N01-HC-95164 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 75N92020D00007 NHLBI NIH HHS
- N01-HC-95165 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- N01-HC-95166 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- N01-HC-95167 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- N01-HC-95169 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 75N92020D00001 NHLBI NIH HHS
- 75N92020D00004 NHLBI NIH HHS
- 75N92020D00007 NHLBI NIH HHS
Collapse
Affiliation(s)
- Florent Murcy
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | - Coraline Borowczyk
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | | | - Stéphanie Torrino
- Université Côte d'Azur, CNRS, Inserm, IPMC, IHU-RespirERA, Valbonne, France
| | | | - Thibault Barouillet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | | | - Marie Couralet
- Université Côte d'Azur, CNRS, Inserm, IPMC, IHU-RespirERA, Valbonne, France
| | - Nathalie Vaillant
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | - Johanna Merlin
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | | | - Minna U Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | - William Tressel
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - James Stein
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Edward B Thorp
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Thomas Bertero
- Université Côte d'Azur, CNRS, Inserm, IPMC, IHU-RespirERA, Valbonne, France
| | - Pascal Barbry
- Université Côte d'Azur, CNRS, Inserm, IPMC, IHU-RespirERA, Valbonne, France
| | - Béatrice Bailly-Maitre
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | | | - Minna K Karjalainen
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Northern Finland Birth Cohorts, Arctic Biobank, Infrastructure for Population Studies, Faculty of Medicine, University of Oulu, Oulu, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Johannes Kettunen
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | | | - Steven Shea
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France.
| |
Collapse
|
9
|
He Y, Cai P, Hu A, Li J, Li X, Dang Y. The role of 1400 plasma metabolites in gastric cancer: A bidirectional Mendelian randomization study and metabolic pathway analysis. Medicine (Baltimore) 2024; 103:e40612. [PMID: 39612432 DOI: 10.1097/md.0000000000040612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2024] Open
Abstract
While observational studies have illustrated correlations between plasma metabolites and gastric cancer (GC), the causal association between the 2 is still unclear. Our study aims to delineate the bidirectional relationship between plasma metabolites and GC and find potential metabolic pathways. We undertook a bidirectional 2-sample Mendelian randomization (MR) analysis to investigate the causal relationship, specificity, and direction of association between 1400 plasma metabolites and GC. The GWAS data for metabolites was obtained from a cohort of 8299 European individuals. And the GC's GWAS data was from FinnGen Consortium with 2384 European individuals, and the GWAS catalog with 1029 European ancestry cases for validation. Causal estimates were primarily calculated by the inverse-variance weighted (IVW) method. To ensure robustness, we performed comprehensive sensitivity analyses to assess heterogeneity and address concerns regarding horizontal pleiotropy. We validated the forward relationship between metabolites and GC from another database and implemented meta-analysis. Furthermore, we conducted metabolic enrichment and pathway analysis of these causal metabolites using MetaboAnalyst5.0/6.0 with the database of Kyoto Encyclopedia of Genes and Genomes. All statistical analysis was carried out using R software. Metabolites like 2s, 3R-dihydroxybutyrate, 4-acetamidobutanoate, ferulic acid 4-sulfate and methyl indole-3-acetate was proven positively linked with the development of GC. Asparagine, glucose to maltose ratio, glycohyocholate, Gulonate levels, linoleoyl ethanolamide and Spermidine to (N(1) + N(8))-acetylspermidine ratio was proven to be negatively associated with GC. Moreover, linoleic acid, histidine, glutamine, bilirubin, Succinate to proline ratio were found to be potentially linked to the development of GC. Furthermore, our analysis identified 18 significant metabolic pathways, including Arginine and proline metabolism (P < .009) and Valine, leucine, and isoleucine biosynthesis (P < .031). Our findings offer evidence supporting potential casual relations between multiple plasma metabolites and GC. These findings may offer great potential for future application of these biomarkers in GC screening and clinical prevention strategies.
Collapse
Affiliation(s)
- Yihao He
- Nanjing Medical University, Nanjing, China
| | | | - Anchi Hu
- Nanjing Medical University, Nanjing, China
| | - Jiali Li
- Nanjing Medical University, Nanjing, China
| | - Xuan Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yini Dang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Lyu J, Gu Z, Zhang Y, Vu HS, Lechauve C, Cai F, Cao H, Keith J, Brancaleoni V, Granata F, Motta I, Cappellini MD, Huang LJS, DeBerardinis RJ, Weiss MJ, Ni M, Xu J. A glutamine metabolic switch supports erythropoiesis. Science 2024; 386:eadh9215. [PMID: 39541460 DOI: 10.1126/science.adh9215] [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: 03/22/2023] [Revised: 02/18/2024] [Accepted: 09/05/2024] [Indexed: 11/16/2024]
Abstract
Metabolic requirements vary during development, and our understanding of how metabolic activity influences cell specialization is incomplete. Here, we describe a switch from glutamine catabolism to synthesis required for erythroid cell maturation. Glutamine synthetase (GS), one of the oldest functioning genes in evolution, is activated during erythroid maturation to detoxify ammonium generated from heme biosynthesis, which is up-regulated to support hemoglobin production. Loss of GS in mouse erythroid precursors caused ammonium accumulation and oxidative stress, impairing erythroid maturation and recovery from anemia. In β-thalassemia, GS activity is inhibited by protein oxidation, leading to glutamate and ammonium accumulation, whereas enhancing GS activity alleviates the metabolic and pathological defects. Our findings identify an evolutionarily conserved metabolic adaptation that could potentially be leveraged to treat common red blood cell disorders.
Collapse
Affiliation(s)
- Junhua Lyu
- Center of Excellence for Leukemia Studies, Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zhimin Gu
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yuannyu Zhang
- Center of Excellence for Leukemia Studies, Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Hieu S Vu
- Center of Excellence for Leukemia Studies, Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christophe Lechauve
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Feng Cai
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hui Cao
- Center of Excellence for Leukemia Studies, Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Julia Keith
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Valentina Brancaleoni
- Unit of Medicine and Metabolic Disease, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesca Granata
- Unit of Medicine and Metabolic Disease, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Irene Motta
- Unit of Medicine and Metabolic Disease, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Maria Domenica Cappellini
- Unit of Medicine and Metabolic Disease, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Lily Jun-Shen Huang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Min Ni
- Division of Molecular Oncology, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jian Xu
- Center of Excellence for Leukemia Studies, Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
11
|
Zhang S, He R, Zhang M, Zhang J, Wu M, Zhang G, Jiang T. Elucidation of the Role of SHMT2 in L-Serine Homeostasis in Hypoxic Hepa1-6 Cells. Int J Mol Sci 2024; 25:11786. [PMID: 39519335 PMCID: PMC11545941 DOI: 10.3390/ijms252111786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Hypoxia is a characteristic feature of malignancy; however, its effect on metabolism remains unclear. In this study, Hepa1-6 cells were cultured under hypoxic conditions and their metabolites were analyzed. Elevated levels of L-serine along with increased glycolytic activity are prominent features of hypoxia. Transcriptome sequencing revealed the downregulation of genes involved in L-serine synthesis and metabolism, which was confirmed by PCR analysis and comparison with public databases. Further experimental evidence indicates that the accumulation of L-serine under hypoxic conditions is attributable not only to enhanced glycolysis but also to a reduction in the catabolism of L-serine mediated by serine hydroxymethyltransferase 2 (SHMT2).
Collapse
Affiliation(s)
- Shuo Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (S.Z.); (R.H.); (J.Z.); (M.W.)
| | - Ruoxu He
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (S.Z.); (R.H.); (J.Z.); (M.W.)
| | - Mingsi Zhang
- Musculoskeletal Sport Science and Health, Loughborough University, Loughborough LE11 3TU, UK;
| | - Jingcheng Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (S.Z.); (R.H.); (J.Z.); (M.W.)
| | - Mengting Wu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (S.Z.); (R.H.); (J.Z.); (M.W.)
| | - Guangji Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (S.Z.); (R.H.); (J.Z.); (M.W.)
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Tao Jiang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (S.Z.); (R.H.); (J.Z.); (M.W.)
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
12
|
Chakraborty A, Patnaik J, Sinha A, Parida N, Parija T, Patnaik S. In silico analysis shows slc1a4 as a potential target of hsa-mir-133a for regulating glutamine metabolism in gastric cancer. Int J Biol Macromol 2024; 282:136974. [PMID: 39486714 DOI: 10.1016/j.ijbiomac.2024.136974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
Cutting-edge research has spotlighted glutamine metabolism as a promising therapeutic target in managing gastric cancer. This investigation highlights the upregulated glutamine transporters by leveraging clinical data from the TCGA Database and the expression analysis of the transcriptome profile of stomach adenocarcinoma (STAD) patients. Notably, it identifies SLC1A4 as a potential glutamine transporter in STAD. The screening of human miRNAs conducted using the TargetScan database, and the subsequent docking analysis present multiple miRNAs with the potential of being explored as therapeutic agents. By integrating transcriptome profiling, miRNA screening, and molecular docking, this study reveals, for the first time, the potential of hsa-mir-133a-1 in targeting slc1a4, along with its known target mTOR, in stomach cancer. The myriad interactions that can be regulated by this silencing mechanism are anticipated to ultimately reduce glutamine uptake in STAD. This study provides compelling evidence of glutamine transport via SLC1A4 in stomach cancer and delves into how it might impact mTOR and some of its pivotal downstream molecules. Considering these findings, novel therapeutic strategies can be devised to further enhance existing methods for combating gastric cancer.
Collapse
Affiliation(s)
- Averi Chakraborty
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India
| | - Jayasree Patnaik
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India
| | - Anupriya Sinha
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India
| | - Nandita Parida
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India
| | - Tithi Parija
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India.
| | - Srinivas Patnaik
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India.
| |
Collapse
|
13
|
Pérez SE, Gooz M, Maldonado EN. Mitochondrial Dysfunction and Metabolic Disturbances Induced by Viral Infections. Cells 2024; 13:1789. [PMID: 39513896 PMCID: PMC11545457 DOI: 10.3390/cells13211789] [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: 09/14/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Viruses are intracellular parasites that utilize organelles, signaling pathways, and the bioenergetics machinery of the cell to replicate the genome and synthesize proteins to build up new viral particles. Mitochondria are key to supporting the virus life cycle by sustaining energy production, metabolism, and synthesis of macromolecules. Mitochondria also contribute to the antiviral innate immune response. Here, we describe the different mechanisms involved in virus-mitochondria interactions. We analyze the effects of viral infections on the metabolism of glucose in the Warburg phenotype, glutamine, and fatty acids. We also describe how viruses directly regulate mitochondrial function through modulation of the activity of the electron transport chain, the generation of reactive oxygen species, the balance between fission and fusion, and the regulation of voltage-dependent anion channels. In addition, we discuss the evasion strategies used to avoid mitochondrial-associated mechanisms that inhibit viral replication. Overall, this review aims to provide a comprehensive view of how viruses modulate mitochondrial function to maintain their replicative capabilities.
Collapse
Affiliation(s)
- Sandra E. Pérez
- Centro de Investigación Veterinaria de Tandil (CIVETAN), UNCPBA-CICPBA-CONICET, Campus Universitario, Tandil CC7000, Buenos Aires, Argentina;
| | - Monika Gooz
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, DD 506 Drug Discovery Building, 70 President Street, MSC 139, Charleston, SC 29425, USA;
| | - Eduardo N. Maldonado
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, DD 506 Drug Discovery Building, 70 President Street, MSC 139, Charleston, SC 29425, USA;
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
14
|
Zhu Y, Liu W, Luo Z, Xiao F, Sun B. New insights into the roles of lactylation in cancer. Front Pharmacol 2024; 15:1412672. [PMID: 39502530 PMCID: PMC11534861 DOI: 10.3389/fphar.2024.1412672] [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: 04/05/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024] Open
Abstract
Lactylation, a novel discovered posttranslational modification, is a vital component of lactate function and is prevalent in a wide range of cells, interacting with both histone and non-histone proteins. Recent studies have confirmed that lactylation as a new contributor to epigenetic landscape is involved in multiple pathological processes. Accumulating evidence reveals that lactylation exists in different pathophysiological states and leads to inflammation and cancer; however, few mechanisms of lactylation have been elaborated. This review summarizes the biological processes and pathophysiological roles of lactylation in cancer, as well as discusses the relevant mechanisms and potential therapeutic targets, aiming to provide new insights for targeted cancer therapy.
Collapse
Affiliation(s)
- Yajun Zhu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Wenhui Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Zhiying Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Feiyan Xiao
- Center for Clinical Trial and Research, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bao Sun
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
15
|
Zhang P, Wu D, Zha X, Su S, Zhang Y, Wei Y, Xia L, Fan S, Peng X. Glutamine promotes the proliferation of intestinal stem cells via inhibition of TP53-induced glycolysis and apoptosis regulator promoter methylation in burned mice. BURNS & TRAUMA 2024; 12:tkae045. [PMID: 39328365 PMCID: PMC11427069 DOI: 10.1093/burnst/tkae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/08/2024] [Indexed: 09/28/2024]
Abstract
Background Intestinal stem cells (ISCs) play a pivotal role in maintaining intestinal homeostasis and facilitating the restoration of intestinal mucosal barrier integrity. Glutamine (Gln) is a crucial energy substrate in the intestine, promoting the proliferation of ISCs and mitigating damage to the intestinal mucosal barrier after burn injury. However, the underlying mechanism has not yet been fully elucidated. The objective of this study was to explore the mechanism by which Gln facilitates the proliferation of ISCs. Methods A mouse burn model was established to investigate the impact of Gln on intestinal function. Subsequently, crypts were isolated, and changes in TP53-induced glycolysis and apoptosis regulator (TIGAR) expression were assessed using real-time quantitative polymerase chain reaction (RT-qPCR), western blotting, immunohistochemistry, and immunofluorescence. The effects of TIGAR on cell proliferation were validated through CCK-8, EdU, and clonogenicity assays. Furthermore, the effect of TIGAR on Yes-associated protein (YAP) nuclear translocation and ferroptosis was examined by western blotting and immunofluorescence staining. Finally, dot blot analysis and methylation-specific PCR were performed to evaluate the effect of Gln on TIGAR promoter methylation. Results The mRNA and protein levels of TIGAR decreased after burn injury, and supplementation with Gln increased the expression of TIGAR. TIGAR accelerates the nuclear translocation of YAP, thereby increasing the proliferation of ISCs. Concurrently, TIGAR promotes the synthesis of nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione to suppress ferroptosis in ISCs. Subsequent investigations demonstrated that Gln inhibits TIGAR promoter methylation by increasing the expression of the demethylase ten-eleven translocation. This change increased TIGAR transcription, increased NADPH synthesis, and reduced oxidative stress, thereby facilitating the restoration of intestinal mucosal barrier integrity post-burn injury. Conclusions Our data confirmed the inhibitory effect of Gln on TIGAR promoter methylation, which facilitates YAP translocation into the nucleus and suppresses ferroptosis, ultimately promoting the proliferation of ISCs.
Collapse
Affiliation(s)
- Panyang Zhang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Dan Wu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xule Zha
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Sen Su
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yajuan Zhang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yan Wei
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Lin Xia
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Shijun Fan
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xi Peng
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| |
Collapse
|
16
|
Lobel GP, Han N, Arocho WAM, Silber M, Shoush J, Noji MC, Jerrick To TK, Zhai L, Lesner NP, Simon MC, Haldar M. Glutamine availability regulates cDC subsets in tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613574. [PMID: 39345449 PMCID: PMC11429688 DOI: 10.1101/2024.09.17.613574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Proliferating tumor cells take up glutamine for anabolic processes engendering glutamine deficiency in the tumor microenvironment. How this might impact immune cells is not well understood. Using multiple mouse models of soft tissue sarcomas, glutamine antagonists, as well as genetic and pharmacological inhibition of glutamine utilization, we found that the number and frequency of conventional dendritic cells (cDC) is dependent on microenvironmental glutamine levels. cDCs comprise two distinct subsets - cDC1 and cDC2, with the former subset playing a critical role in antigen cross-presentation and tumor immunity. While both subsets show dependence on Glutamine, cDC1s are particularly sensitive. Notably, glutamine antagonism did not reduce the frequency of DC precursors but decreased proliferation and survival of cDC1s. Further studies suggest a role of the nutrient sensing mTOR signaling pathway in this process. Taken together, these findings uncover glutamine dependence of cDC1s that is coopted by tumors to escape immune responses. One Sentence Summary Type 1 conventional dendritic cells require glutamine to maintain their number in non-lymphoid tissue. Significance Immune evasion is a key hallmark of cancer; however, the underlying pathways are diverse, tumor-specific and not fully elucidated. Many tumor cells avidly import glutamine to support their anabolic needs, creating a glutamine-deficient tumor microenvironment (TME). Herein, using mouse models of soft tissue sarcomas, we show that glutamine depletion in TME leads to reduced type 1 conventional dendritic cells - a cell type that is critical for adaptive immune responses. This work is a paradigm for how tumor cell metabolism can regulate anti-tumor immune responses and will be foundational to future efforts targeting glutamine metabolism for cancer immunotherapy.
Collapse
|
17
|
Chen Y, Yang S, Yu T, Zeng T, Wei L, You Y, Tang J, Dang T, Sun H, Zhang Y. KDM4A promotes malignant progression of breast cancer by down-regulating BMP9 inducing consequent enhancement of glutamine metabolism. Cancer Cell Int 2024; 24:322. [PMID: 39300582 DOI: 10.1186/s12935-024-03504-0] [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: 07/21/2024] [Accepted: 09/06/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Recent studies have found that histone-modified genes play an increasingly important role in tumor progression. Lysine(K) specific demethylase 4A (KDM4A) is a histone lysine-specific demethylase highly expressed in a variety of malignant tumors, data showed that KDM4A was negatively correlated with the Bone Morphogenetic Protein 9 (BMP9) in breast cancer. And previous experiments have demonstrated that exogenous BMP9 significantly inhibits breast cancer development. MATERIALS AND METHODS We detected the expression of KDM4A in breast cancer and the relationship between KDM4A and BMP9 using real-time quantitative PCR (RT-qPCR) and Western blot, and verified the interaction between KDM4A and BMP9 by ChIP experiments. At the same time, we also detected whether KDM4A had effects on the RNA and protein stability of BMP9 using actinomycin D and cycloheximide. Measurement of alpha-ketoglutarate (α-KG) level by ELISA to observe the effect of BMP9 on glutamine metabolism in breast cancer cells. Nucleoplasmic distribution of KDM4A after exogenous BMP9 treatment in breast cancer cells were observed by immunofluorescence staining and Western blot. A subcutaneous xenograft tumor model in nude mice was used to study the therapeutic effects of exogenous BMP9 and KDM4A inhibitor (JIB-04) in breast cancer. CCK-8, conoly formation, Transwell, wound healing, and immunohistochemistry were used to monitor the growth of tumor and cell function. RESULTS We found that KDM4A was abnormally highly expressed in breast cancer, and silenced BMP9 expression by removing histone methyl groups from the BMP9 gene region. Meanwhile, KDM4A could also reduce the stability of BMP9 protein. BMP9 inhibit glutamine metabolism in breast cancer, resulting in a decrease in its product α-KG, is confirmed by ELISA. Altered nucleoplasmic distribution of KDM4A due to decreased α-KG was confirmed by immunofluorescence staining and Western blot. Animal experiments confirm that the combination of exogenous BMP9 and JIB-04 shows significantly better results in breast cancer. CONCLUSIONS KDM4A silences BMP9 expression by removing histone methyl groups from the BMP9 gene region, leading to further enhancement of glutamine metabolism, which contributes to malignant tumor progression. In addition, using JIB-04 in combination with exogenous BMP9 could inhibit the malignant progression of breast cancer cells and the growth of tumors more significantly.
Collapse
Affiliation(s)
- Yuanxiang Chen
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Shiyu Yang
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Tao Yu
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Tao Zeng
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Lan Wei
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yiqing You
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jiafeng Tang
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Tingting Dang
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Haoli Sun
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yan Zhang
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
18
|
Maurya M, Liu CH, Bora K, Kushwah N, Pavlovich MC, Wang Z, Chen J. Animal Models of Retinopathy of Prematurity: Advances and Metabolic Regulators. Biomedicines 2024; 12:1937. [PMID: 39335451 PMCID: PMC11428941 DOI: 10.3390/biomedicines12091937] [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: 07/01/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 09/30/2024] Open
Abstract
Retinopathy of prematurity (ROP) is a primary cause of visual impairment and blindness in premature newborns, characterized by vascular abnormalities in the developing retina, with microvascular alteration, neovascularization, and in the most severe cases retinal detachment. To elucidate the pathophysiology and develop therapeutics for ROP, several pre-clinical experimental models of ROP were developed in different species. Among them, the oxygen-induced retinopathy (OIR) mouse model has gained the most popularity and critically contributed to our current understanding of pathological retinal angiogenesis and the discovery of potential anti-angiogenic therapies. A deeper comprehension of molecular regulators of OIR such as hypoxia-inducible growth factors including vascular endothelial growth factors as primary perpetrators and other new metabolic modulators such as lipids and amino acids influencing pathological retinal angiogenesis is also emerging, indicating possible targets for treatment strategies. This review delves into the historical progressions that gave rise to the modern OIR models with a focus on the mouse model. It also reviews the fundamental principles of OIR, recent advances in its automated assessment, and a selected summary of metabolic investigation enabled by OIR models including amino acid transport and metabolism.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jing Chen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
19
|
Chai X, Zhang Y, Zhang W, Feng K, Jiang Y, Zhu A, Chen X, Di L, Wang R. Tumor Metabolism: A New Field for the Treatment of Glioma. Bioconjug Chem 2024; 35:1116-1141. [PMID: 39013195 DOI: 10.1021/acs.bioconjchem.4c00287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
The clinical treatment of glioma remains relatively immature. Commonly used clinical treatments for gliomas are surgery combined with chemotherapy and radiotherapy, but there is a problem of drug resistance. In addition, immunotherapy and targeted therapies also suffer from the problem of immune evasion. The advent of metabolic therapy holds immense potential for advancing more efficacious and tolerable therapies against this aggressive disease. Metabolic therapy alters the metabolic processes of tumor cells at the molecular level to inhibit tumor growth and spread, and lead to better outcomes for patients with glioma that are insensitive to conventional treatments. Moreover, compared with conventional therapy, it has less impact on normal cells, less toxicity and side effects, and higher safety. The objective of this review is to examine the changes in metabolic characteristics throughout the development of glioma, enumerate the current methodologies employed for studying tumor metabolism, and highlight the metabolic reprogramming pathways of glioma along with their potential molecular mechanisms. Importantly, it seeks to elucidate potential metabolic targets for glioblastoma (GBM) therapy and summarize effective combination treatment strategies based on various studies.
Collapse
Affiliation(s)
- Xiaoqian Chai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Yingjie Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Wen Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Kuanhan Feng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Yingyu Jiang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Anran Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Xiaojin Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| |
Collapse
|
20
|
Erb HHH, Polishchuk N, Stasyk O, Kahya U, Weigel MM, Dubrovska A. Glutamine Metabolism and Prostate Cancer. Cancers (Basel) 2024; 16:2871. [PMID: 39199642 PMCID: PMC11352381 DOI: 10.3390/cancers16162871] [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: 06/29/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/01/2024] Open
Abstract
Glutamine (Gln) is a non-essential amino acid that is involved in the development and progression of several malignancies, including prostate cancer (PCa). While Gln is non-essential for non-malignant prostate epithelial cells, PCa cells become highly dependent on an exogenous source of Gln. The Gln metabolism in PCa is tightly controlled by well-described oncogenes such as MYC, AR, and mTOR. These oncogenes contribute to therapy resistance and progression to the aggressive castration-resistant PCa. Inhibition of Gln catabolism impedes PCa growth, survival, and tumor-initiating potential while sensitizing the cells to radiotherapy. Therefore, given its significant role in tumor growth, targeting Gln metabolism is a promising approach for developing new therapeutic strategies. Ongoing clinical trials evaluate the safety and efficacy of Gln catabolism inhibitors in combination with conventional and targeted therapies in patients with various solid tumors, including PCa. Further understanding of how PCa cells metabolically interact with their microenvironment will facilitate the clinical translation of Gln inhibitors and help improve therapeutic outcomes. This review focuses on the role of Gln in PCa progression and therapy resistance and provides insights into current clinical trials.
Collapse
Affiliation(s)
- Holger H. H. Erb
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany;
| | - Nikita Polishchuk
- Department of Cell Signaling, Institute of Cell Biology, National Academy of Sciences of Ukraine, 79000 Lviv, Ukraine; (N.P.); (O.S.)
| | - Oleh Stasyk
- Department of Cell Signaling, Institute of Cell Biology, National Academy of Sciences of Ukraine, 79000 Lviv, Ukraine; (N.P.); (O.S.)
| | - Uğur Kahya
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (M.M.W.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
| | - Matthias M. Weigel
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (M.M.W.)
| | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (M.M.W.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, 01309 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| |
Collapse
|
21
|
Qiu Y, Stamatatos OT, Hu Q, Ruiter Swain J, Russo S, Sann A, Costa ASH, Violante S, Spector DL, Cross JR, Lukey MJ. The unique catalytic properties of PSAT1 mediate metabolic adaptation to glutamine blockade. Nat Metab 2024; 6:1529-1548. [PMID: 39192144 PMCID: PMC11490312 DOI: 10.1038/s42255-024-01104-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 07/09/2024] [Indexed: 08/29/2024]
Abstract
Cultured cancer cells frequently rely on the consumption of glutamine and its subsequent hydrolysis by glutaminase (GLS). However, this metabolic addiction can be lost in the tumour microenvironment, rendering GLS inhibitors ineffective in the clinic. Here we show that glutamine-addicted breast cancer cells adapt to chronic glutamine starvation, or GLS inhibition, via AMPK-mediated upregulation of the serine synthesis pathway (SSP). In this context, the key product of the SSP is not serine, but α-ketoglutarate (α-KG). Mechanistically, we find that phosphoserine aminotransferase 1 (PSAT1) has a unique capacity for sustained α-KG production when glutamate is depleted. Breast cancer cells with resistance to glutamine starvation or GLS inhibition are highly dependent on SSP-supplied α-KG. Accordingly, inhibition of the SSP prevents adaptation to glutamine blockade, resulting in a potent drug synergism that suppresses breast tumour growth. These findings highlight how metabolic redundancy can be context dependent, with the catalytic properties of different metabolic enzymes that act on the same substrate determining which pathways can support tumour growth in a particular nutrient environment. This, in turn, has practical consequences for therapies targeting cancer metabolism.
Collapse
Affiliation(s)
- Yijian Qiu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Qingting Hu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY, USA
| | - Jed Ruiter Swain
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- CSHL School of Biological Sciences, Cold Spring Harbor, NY, USA
| | - Suzanne Russo
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ava Sann
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ana S H Costa
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Matterworks Inc., Somerville, MA, USA
| | - Sara Violante
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Justin R Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
22
|
Wu J, Han W, Zhang Y, Li S, Qin T, Huang Z, Zhang C, Shi M, Wu Y, Zheng W, Gao B, Xu K, Ye W. Glutamine Mitigates Oxidative Stress-Induced Matrix Degradation, Ferroptosis, and Pyroptosis in Nucleus Pulposus Cells via Deubiquitinating and Stabilizing Nrf2. Antioxid Redox Signal 2024; 41:278-295. [PMID: 38504579 DOI: 10.1089/ars.2023.0384] [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] [Indexed: 03/21/2024]
Abstract
Aims: Intervertebral disc degeneration (IDD) is closely related to low back pain, which is a prevalent age-related problem worldwide; however, the mechanism underlying IDD is unknown. Glutamine, a free amino acid prevalent in plasma, is recognized for its anti-inflammatory and antioxidant properties in various diseases, and the current study aims to clarify the effect and mechanism of glutamine in IDD. Results: A synergistic interplay was observed between pyroptosis and ferroptosis within degenerated human disc specimens. Glutamine significantly mitigated IDD in both ex vivo and in vivo experimental models. Moreover, glutamine protected nucleus pulposus (NP) cells after tert-butyl hydroperoxide (TBHP)-induced pyroptosis, ferroptosis, and extracellular matrix (ECM) degradation in vitro. Glutamine protected NP cells from TBHP-induced ferroptosis by promoting the nuclear factor erythroid 2-related factor 2 (Nrf2) accumulation by inhibiting its ubiquitin-proteasome degradation and inhibiting lipid oxidation. Innovation and Conclusions: A direct correlation is evident in the progression of IDD between the processes of pyroptosis and ferroptosis. Glutamine suppressed oxidative stress-induced cellular processes, including pyroptosis, ferroptosis, and ECM degradation through deubiquitinating Nrf2 and inhibiting lipid oxidation in NP cells. Glutamine is a promising novel therapeutic target for the management of IDD.
Collapse
Affiliation(s)
- Jiajun Wu
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weitao Han
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yangyang Zhang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuangxing Li
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Orthopedics, Sun Yat-sen Memorial Hospital Shenshan Central Hospital of Sun Yat-sen University, Shanwei, China
| | - Tianyu Qin
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Orthopedics, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhengqi Huang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chao Zhang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ming Shi
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Orthopedics, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yuliang Wu
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wanli Zheng
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Orthopedics, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Bo Gao
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kang Xu
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Ye
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Cai Y, Li L, Shao C, Chen Y, Wang Z. Therapeutic Strategies for Angiogenesis Based on Endothelial Cell Epigenetics. J Cardiovasc Transl Res 2024; 17:816-827. [PMID: 38294628 DOI: 10.1007/s12265-024-10485-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/17/2024] [Indexed: 02/01/2024]
Abstract
With the in-depth investigation of various diseases, angiogenesis has gained increasing attention. Among the contributing factors to angiogenesis research, endothelial epigenetics has emerged as an influential player. Endothelial epigenetic therapy exerts its regulatory effects on endothelial cells by controlling gene expression, RNA, and histone modification within these cells, which subsequently promotes or inhibits angiogenesis. As a result, this therapeutic approach offers potential strategies for disease treatment. The purpose of this review is to outline the pertinent mechanisms of endothelial cell epigenetics, encompassing glycolysis, lactation, amino acid metabolism, non-coding RNA, DNA methylation, histone modification, and their connections to specific diseases and clinical applications. We firmly believe that endothelial cell epigenetics has the potential to become an integral component of precision medicine therapy, unveiling novel therapeutic targets and providing new directions and opportunities for disease treatment.
Collapse
Affiliation(s)
- Yue Cai
- Department of Cardiology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China
| | - Yiliu Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China.
| |
Collapse
|
24
|
Liu X, Ren B, Ren J, Gu M, You L, Zhao Y. The significant role of amino acid metabolic reprogramming in cancer. Cell Commun Signal 2024; 22:380. [PMID: 39069612 DOI: 10.1186/s12964-024-01760-1] [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: 04/15/2024] [Accepted: 07/21/2024] [Indexed: 07/30/2024] Open
Abstract
Amino acid metabolism plays a pivotal role in tumor microenvironment, influencing various aspects of cancer progression. The metabolic reprogramming of amino acids in tumor cells is intricately linked to protein synthesis, nucleotide synthesis, modulation of signaling pathways, regulation of tumor cell metabolism, maintenance of oxidative stress homeostasis, and epigenetic modifications. Furthermore, the dysregulation of amino acid metabolism also impacts tumor microenvironment and tumor immunity. Amino acids can act as signaling molecules that modulate immune cell function and immune tolerance within the tumor microenvironment, reshaping the anti-tumor immune response and promoting immune evasion by cancer cells. Moreover, amino acid metabolism can influence the behavior of stromal cells, such as cancer-associated fibroblasts, regulate ECM remodeling and promote angiogenesis, thereby facilitating tumor growth and metastasis. Understanding the intricate interplay between amino acid metabolism and the tumor microenvironment is of crucial significance. Expanding our knowledge of the multifaceted roles of amino acid metabolism in tumor microenvironment holds significant promise for the development of more effective cancer therapies aimed at disrupting the metabolic dependencies of cancer cells and modulating the tumor microenvironment to enhance anti-tumor immune responses and inhibit tumor progression.
Collapse
Affiliation(s)
- Xiaohong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China
| | - Bo Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China
| | - Jie Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China
| | - Minzhi Gu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China.
| |
Collapse
|
25
|
Liu J, Li H, Wang L, Wang S, Tang Q. Spatial transcriptome and single-cell reveal the role of nucleotide metabolism in colorectal cancer progression and tumor microenvironment. J Transl Med 2024; 22:702. [PMID: 39075485 PMCID: PMC11288102 DOI: 10.1186/s12967-024-05495-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/10/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND The intricacies of nucleotide metabolism within tumor cells specific to colorectal cancer (CRC) remain insufficiently characterized. A nuanced examination of particular tumor clusters and their dynamic interplay with the tumor microenvironment (TME) may yield profound insights into these therapeutically auspicious communicative networks. METHODS By integrating ten types of single-cell enrichment scoring methods, we carried out enrichment analysis on CRC cell types, which was validated through four additional single-cell cohorts. Groups of tumor cells were determined using the average values of the scores. Using cellphonedb, monocle, inferCNV, SCENIC, and Cytotrace, functional analyses were performed. Utilizing the RCTD approach, single-cell groupings were mapped onto spatial transcriptomics, analyzing cell dependency and pathway activity to distinguish between tumor cell subtypes. Differential expression analysis identified core genes in nucleotide metabolism, with single-cell and spatial transcriptomics analyses elucidating the function of these genes in tumor cells and the immune microenvironment. Prognostic models were developed from bulk transcriptome cohorts to forecast responses to immune therapy. Laboratory experiments were conducted to verify the biological function of the core gene. RESULTS Nucleotide metabolism is significantly elevated in tumor cells, dividing them into two groups: NUhighepi and NUlowepi. The phenotype NUhighepi was discerned to exhibit pronounced malignant attributes. Utilizing the analytical tool stlearn for cell-to-cell communication assessment, it was ascertained that NUhighepi engages in intimate interactions with fibroblasts. Corroborating this observation, spatial transcriptome cell interaction assessment through MISTy unveiled a particular reliance of NUhighepi on fibroblasts. Subsequently, we pinpointed NME1, a key gene in nucleotide metabolism, affirming its role in thwarting metastasis via in vitro examination. Utilizing multiple machine learning algorithms, a stable prognostic model (NRS) has been developed, capable of predicting survival and responses to immune therapy. In addition, targeted drugs have been identified for both high and low scoring groups. Laboratory experiments have revealed that NME1 can inhibit the proliferation and invasion of CRC tumor cells. CONCLUSION Our study elucidates the potential pro-tumor mechanism of NUhighepi and the role of NME1 in inhibiting metastasis, further deepening the understanding of the role of nucleotide metabolism in colorectal cancer, and providing valuable targets for disrupting its properties.
Collapse
Affiliation(s)
- Junzhi Liu
- The Second Affiliated Hospital of Zhejiang University School Medicine, Hang Zhou, 310000, China
| | - Huimin Li
- The Second Affiliated Hospital of Zhejiang University School Medicine, Hang Zhou, 310000, China
| | - Lantian Wang
- The Second Affiliated Hospital of Zhejiang University School Medicine, Hang Zhou, 310000, China.
| | - Shurui Wang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
| | - Qiang Tang
- The Second Affiliated Hospital of Zhejiang University School Medicine, Hang Zhou, 310000, China.
| |
Collapse
|
26
|
Kinslow CJ, Ll MB, Cai Y, Yan J, Lorkiewicz PK, Al-Attar A, Tan J, Higashi RM, Lane AN, Fan TWM. Stable isotope-resolved metabolomics analyses of metabolic phenotypes reveal variable glutamine metabolism in different patient-derived models of non-small cell lung cancer from a single patient. Metabolomics 2024; 20:87. [PMID: 39068202 PMCID: PMC11317205 DOI: 10.1007/s11306-024-02126-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 05/02/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION Stable isotope tracers have been increasingly used in preclinical cancer model systems, including cell culture and mouse xenografts, to probe the altered metabolism of a variety of cancers, such as accelerated glycolysis and glutaminolysis and generation of oncometabolites. Comparatively little has been reported on the fidelity of the different preclinical model systems in recapitulating the aberrant metabolism of tumors. OBJECTIVES We have been developing several different experimental model systems for systems biochemistry analyses of non-small cell lung cancer (NSCLC1) using patient-derived tissues to evaluate appropriate models for metabolic and phenotypic analyses. METHODS To address the issue of fidelity, we have carried out a detailed Stable Isotope-Resolved Metabolomics study of freshly resected tissue slices, mouse patient derived xenografts (PDXs), and cells derived from a single patient using both 13C6-glucose and 13C5,15N2-glutamine tracers. RESULTS Although we found similar glucose metabolism in the three models, glutamine utilization was markedly higher in the isolated cell culture and in cell culture-derived xenografts compared with the primary cancer tissue or direct tissue xenografts (PDX). CONCLUSIONS This suggests that caution is needed in interpreting cancer biochemistry using patient-derived cancer cells in vitro or in xenografts, even at very early passage, and that direct analysis of patient derived tissue slices provides the optimal model for ex vivo metabolomics. Further research is needed to determine the generality of these observations.
Collapse
Affiliation(s)
- Connor J Kinslow
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, 622 West 168th Street, BNH B-11, New York, NY, 10032, USA
| | - Michael Bousamra Ll
- Department of Cardiovascular and Thoracic Surgery, University of Louisville, Louisville, KY, 40202, USA
- AMG Cardiothoracic Surgical Associates SE MI, 22201 Moross Rd. #352, Detroit, MI, 48236, USA
| | - Yihua Cai
- Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
- Center for Cellular Engineering, Department of Transfusion Medicine, NIH Clinical Center, Bethesda, MD, 20892, USA
| | - Jun Yan
- Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
- Division of Immunotherapy, The Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville, Louisville, KY, 40202, USA
| | - Pawel K Lorkiewicz
- Department of Chemistry, University of Louisville, Louisville, KY, 40202, USA
| | - Ahmad Al-Attar
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
- Dept. Pathology, U. Mass Memorial Medical Center, University of Massachusetts, Worcester, MA, 01605, USA
| | - Jinlian Tan
- The Department of Oral Immunology and Infection Disease, School of Dentistry, University of Louisville, 501 South Preston, St. Louisville, KY, 40202, USA
| | - Richard M Higashi
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Andrew N Lane
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA.
| | - Teresa W-M Fan
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
27
|
da Silva Costa SM, Ito MT, da Cruz PRS, De Souza BB, Rios VM, Bertozzo VDHE, Camargo ACL, Viturino MGM, Lanaro C, de Albuquerque DM, do Canto AM, Saad STO, Ospina-Prieto S, Ozelo MC, Costa FF, de Melo MB. The molecular mechanism responsible for HbSC retinopathy may depend on the action of the angiogenesis-related genes ROBO1 and SLC38A5. Exp Biol Med (Maywood) 2024; 249:10070. [PMID: 39114443 PMCID: PMC11303203 DOI: 10.3389/ebm.2024.10070] [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: 12/06/2023] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
HbSC disease, a less severe form of sickle cell disease, affects the retina more frequently and patients have higher rates of proliferative retinopathy that can progress to vision loss. This study aimed to identify differences in the expression of endothelial cell-derived molecules associated with the pathophysiology of proliferative sickle cell retinopathy (PSCR). RNAseq was used to compare the gene expression profile of circulating endothelial colony-forming cells from patients with SC hemoglobinopathy and proliferative retinopathy (n = 5), versus SC patients without retinopathy (n = 3). Real-time polymerase chain reaction (qRT-PCR) was used to validate the RNAseq results. A total of 134 differentially expressed genes (DEGs) were found. DEGs were mainly associated with vasodilatation, type I interferon signaling, innate immunity and angiogenesis. Among the DEGs identified, we highlight the most up-regulated genes ROBO1 (log2FoldChange = 4.32, FDR = 1.35E-11) and SLC38A5 (log2FoldChange = 3.36 FDR = 1.59E-07). ROBO1, an axon-guided receptor, promotes endothelial cell migration and contributes to the development of retinal angiogenesis and pathological ocular neovascularization. Endothelial SLC38A5, an amino acid (AA) transporter, regulates developmental and pathological retinal angiogenesis by controlling the uptake of AA nutrient, which may serve as metabolic fuel for the proliferation of endothelial cells (ECs) and consequent promotion of angiogenesis. Our data provide an important step towards elucidating the molecular pathophysiology of PSCR that may explain the differences in ocular manifestations between individuals with hemoglobinopathies and afford insights for new alternative strategies to inhibit pathological angiogenesis.
Collapse
Affiliation(s)
| | - Mirta Tomie Ito
- Center for Molecular Biology and Genetic Engineering, State University of Campinas—UNICAMP, Campinas, Brazil
| | | | - Bruno Batista De Souza
- Center for Molecular Biology and Genetic Engineering, State University of Campinas—UNICAMP, Campinas, Brazil
| | - Vinicius Mandolesi Rios
- Center for Molecular Biology and Genetic Engineering, State University of Campinas—UNICAMP, Campinas, Brazil
| | - Victor de Haidar e Bertozzo
- Center for Molecular Biology and Genetic Engineering, State University of Campinas—UNICAMP, Campinas, Brazil
| | - Ana Carolina Lima Camargo
- Center for Molecular Biology and Genetic Engineering, State University of Campinas—UNICAMP, Campinas, Brazil
| | | | - Carolina Lanaro
- Centro de Hematologia e Hemoterapia, Universidade Estadual de Campinas—UNICAMP, Campinas, Brazil
| | | | - Amanda Morato do Canto
- Departamento de Medicina Translacional, Faculdade de Ciências Médicas, Universidade Estadual de Campinas—UNICAMP, Campinas, Brazil
| | | | - Stephanie Ospina-Prieto
- Centro de Hematologia e Hemoterapia, Universidade Estadual de Campinas—UNICAMP, Campinas, Brazil
| | - Margareth Castro Ozelo
- Centro de Hematologia e Hemoterapia, Universidade Estadual de Campinas—UNICAMP, Campinas, Brazil
| | - Fernando Ferreira Costa
- Centro de Hematologia e Hemoterapia, Universidade Estadual de Campinas—UNICAMP, Campinas, Brazil
| | - Mônica Barbosa de Melo
- Center for Molecular Biology and Genetic Engineering, State University of Campinas—UNICAMP, Campinas, Brazil
| |
Collapse
|
28
|
Selarka K, Shravage BV. Illuminating intercellular autophagy: A comprehensive review of cell non-autonomous autophagy. Biochem Biophys Res Commun 2024; 716:150024. [PMID: 38701555 DOI: 10.1016/j.bbrc.2024.150024] [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: 03/31/2024] [Accepted: 04/26/2024] [Indexed: 05/05/2024]
Abstract
Macro-autophagy (autophagy hereafter) is an evolutionarily conserved cellular process that has long been recognized as an intracellular mechanism for maintaining cellular homeostasis. It involves the formation of a membraned structure called the autophagosome, which carries cargo that includes toxic protein aggregates and dysfunctional organelles to the lysosome for degradation and recycling. Autophagy is primarily considered and studied as a cell-autonomous mechanism. However, recent studies have illuminated an underappreciated facet of autophagy, i.e., non-autonomously regulated autophagy. Non-autonomously regulated autophagy involves the degradation of autophagic components, including organelles, cargo, and signaling molecules, and is induced in neighboring cells by signals from primary adjacent or distant cells/tissues/organs. This review provides insight into the complex molecular mechanisms governing non-autonomously regulated autophagy, highlighting the dynamic interplay between cells within tissue/organ or distinct cell types in different tissues/organs. Emphasis is placed on modes of intercellular communication that include secreted molecules, including microRNAs, and their regulatory roles in orchestrating this phenomenon. Furthermore, we explore the multidimensional roles of non-autonomously regulated autophagy in various physiological contexts, spanning tissue development and aging, as well as its importance in diverse pathological conditions, including cancer and neurodegeneration. By studying the complexities of non-autonomously regulated autophagy, we hope to gain insights into the sophisticated intercellular dynamics within multicellular organisms, including mammals. These studies will uncover novel avenues for therapeutic intervention to modulate intercellular autophagic pathways in altered human physiology.
Collapse
Affiliation(s)
- Karan Selarka
- Developmental Biology Group, MACS-Agharkar Research Institute, Pune, India; Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Bhupendra V Shravage
- Developmental Biology Group, MACS-Agharkar Research Institute, Pune, India; Department of Biotechnology, Savitribai Phule Pune University, Pune, India; Department of Zoology, Savitribai Phule Pune University, Pune, India.
| |
Collapse
|
29
|
De los Santos-Jiménez J, Campos-Sandoval JA, Alonso FJ, Márquez J, Matés JM. GLS and GLS2 Glutaminase Isoenzymes in the Antioxidant System of Cancer Cells. Antioxidants (Basel) 2024; 13:745. [PMID: 38929183 PMCID: PMC11200642 DOI: 10.3390/antiox13060745] [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: 05/25/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
A pathway frequently altered in cancer is glutaminolysis, whereby glutaminase (GA) catalyzes the main step as follows: the deamidation of glutamine to form glutamate and ammonium. There are two types of GA isozymes, named GLS and GLS2, which differ considerably in their expression patterns and can even perform opposing roles in cancer. GLS correlates with tumor growth and proliferation, while GLS2 can function as a context-dependent tumor suppressor. However, both isoenzymes have been described as essential molecules handling oxidant stress because of their involvement in glutathione production. We reviewed the literature to highlight the critical roles of GLS and GLS2 in restraining ROS and regulating both cellular signaling and metabolic stress due to their function as indirect antioxidant enzymes, as well as by modulating both reductive carboxylation and ferroptosis. Blocking GA activity appears to be a potential strategy in the dual activation of ferroptosis and inhibition of cancer cell growth in a ROS-mediated mechanism.
Collapse
Affiliation(s)
- Juan De los Santos-Jiménez
- Canceromics Lab, Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.D.l.S.-J.); (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29590 Málaga, Spain
| | - José A. Campos-Sandoval
- Canceromics Lab, Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.D.l.S.-J.); (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29590 Málaga, Spain
| | - Francisco J. Alonso
- Canceromics Lab, Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.D.l.S.-J.); (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29590 Málaga, Spain
| | - Javier Márquez
- Canceromics Lab, Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.D.l.S.-J.); (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29590 Málaga, Spain
| | - José M. Matés
- Canceromics Lab, Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.D.l.S.-J.); (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29590 Málaga, Spain
| |
Collapse
|
30
|
Patel R, Alfarsi LH, El-Ansari R, Masisi BK, Erkan B, Fakroun A, Ellis IO, Rakha EA, Green AR. ATF4 as a Prognostic Marker and Modulator of Glutamine Metabolism in Oestrogen Receptor-Positive Breast Cancer. Pathobiology 2024; 91:411-421. [PMID: 38861938 PMCID: PMC11614298 DOI: 10.1159/000539564] [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: 11/20/2023] [Accepted: 05/25/2024] [Indexed: 06/13/2024] Open
Abstract
INTRODUCTION ATF4, a stress-responsive transcription factor that upregulates adaptive genes, is a potential prognostic marker and modulator of glutamine metabolism in breast cancer. However, its exact role remains to be elucidated. METHODS ATF4 expression was evaluated at genomic and transcriptomic levels using METABRIC (n = 1,980), GeneMiner (n = 4,712), and KM-Plotter datasets. Proteomic expression was assessed via immunohistochemistry (n = 2,225) in the Nottingham Primary Breast Cancer Series. ATF4 genomic copy number (CN) variation and mRNA/protein in association with clinicopathological parameters, amino acid transporters (AATs), and patient outcome were investigated. RESULTS Genomic, transcriptomic, and proteomic overexpression of ATF4 was associated with more aggressive ER-negative tumours. ATF4 mRNA and protein expression were significantly associated with increased expression of glutamine related AATs including SLC1A5 (p < 0.01) and SLC7A11 (p < 0.02). High ATF4 and SLC1A5 protein expression was significantly associated with shorter breast cancer-specific survival (p < 0.01), especially in ER+ tumours (p < 0.01), while high ATF4 and SLC7A11 protein expression was associated with shorter survival (p < 0.01). CONCLUSION These findings suggest a complex interplay between ATF4 and AATs in breast cancer biology and underscore the potential role for ATF4 as a prognostic marker in ER+ breast cancer, offering a unique opportunity for risk stratification and personalized treatment strategies.
Collapse
Affiliation(s)
- Roshni Patel
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Lutfi H. Alfarsi
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Rokaya El-Ansari
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Brendah K. Masisi
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Busra Erkan
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Ali Fakroun
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Ian O. Ellis
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
- Cellular Pathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Emad A. Rakha
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
- Cellular Pathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Andrew R. Green
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| |
Collapse
|
31
|
Chen X, Xu Y, Ju Y, Gu P. Metabolic Regulation of Endothelial Cells: A New Era for Treating Wet Age-Related Macular Degeneration. Int J Mol Sci 2024; 25:5926. [PMID: 38892113 PMCID: PMC11172501 DOI: 10.3390/ijms25115926] [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: 04/16/2024] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Wet age-related macular degeneration (wet AMD) is a primary contributor to visual impairment and severe vision loss globally, but the prevailing treatments are often unsatisfactory. The development of conventional treatment strategies has largely been based on the understanding that the angiogenic switch of endothelial cells (ECs) is mainly dictated by angiogenic growth factors. Even though treatments targeting vascular endothelial growth factor (VEGF), like ranibizumab, are widely administered, more than half of patients still exhibit inadequate or null responses, suggesting the involvement of other pathogenic mechanisms. With advances in research in recent years, it has become well recognized that EC metabolic regulation plays an active rather than merely passive responsive role in angiogenesis. Disturbances of these metabolic pathways may lead to excessive neovascularization in angiogenic diseases such as wet AMD, therefore targeted modulation of EC metabolism represents a promising therapeutic strategy for wet AMD. In this review, we comprehensively discuss the potential applications of EC metabolic regulation in wet AMD treatment from multiple perspectives, including the involvement of ECs in wet AMD pathogenesis, the major endothelial metabolic pathways, and novel therapeutic approaches targeting metabolism for wet AMD.
Collapse
Affiliation(s)
- Xirui Chen
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (X.C.)
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Yang Xu
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (X.C.)
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Yahan Ju
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (X.C.)
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Ping Gu
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (X.C.)
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| |
Collapse
|
32
|
de Zeeuw P, Treps L, García-Caballero M, Harjes U, Kalucka J, De Legher C, Brepoels K, Peeters K, Vinckier S, Souffreau J, Bouché A, Taverna F, Dehairs J, Talebi A, Ghesquière B, Swinnen J, Schoonjans L, Eelen G, Dewerchin M, Carmeliet P. The gluconeogenesis enzyme PCK2 has a non-enzymatic role in proteostasis in endothelial cells. Commun Biol 2024; 7:618. [PMID: 38783087 PMCID: PMC11116505 DOI: 10.1038/s42003-024-06186-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 04/11/2024] [Indexed: 05/25/2024] Open
Abstract
Endothelial cells (ECs) are highly glycolytic, but whether they generate glycolytic intermediates via gluconeogenesis (GNG) in glucose-deprived conditions remains unknown. Here, we report that glucose-deprived ECs upregulate the GNG enzyme PCK2 and rely on a PCK2-dependent truncated GNG, whereby lactate and glutamine are used for the synthesis of lower glycolytic intermediates that enter the serine and glycerophospholipid biosynthesis pathways, which can play key roles in redox homeostasis and phospholipid synthesis, respectively. Unexpectedly, however, even in normal glucose conditions, and independent of its enzymatic activity, PCK2 silencing perturbs proteostasis, beyond its traditional GNG role. Indeed, PCK2-silenced ECs have an impaired unfolded protein response, leading to accumulation of misfolded proteins, which due to defective proteasomes and impaired autophagy, results in the accumulation of protein aggregates in lysosomes and EC demise. Ultimately, loss of PCK2 in ECs impaired vessel sprouting. This study identifies a role for PCK2 in proteostasis beyond GNG.
Collapse
Affiliation(s)
- Pauline de Zeeuw
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium
- Droia Ventures, Zaventem, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium
- CNRS, Nantes, France
| | - Melissa García-Caballero
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium
- Dept. Molecular Biology and Biochemistry, Fac. Science, University of Malaga, Malaga, Spain
| | - Ulrike Harjes
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium
| | - Joanna Kalucka
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium
- Aarhus Institute of Advanced Studies (AIAS), Department of Biomedicine, Aarhus University, Aarhus, 8000, Denmark
| | - Carla De Legher
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium
| | - Katleen Brepoels
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium
| | - Kristel Peeters
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium
| | - Stefan Vinckier
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium
| | - Joris Souffreau
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium
| | - Ann Bouché
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium
| | - Federico Taverna
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium
- Novartis Ireland, Dublin, Ireland
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
| | - Ali Talebi
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
| | - Bart Ghesquière
- Metabolomics Core Facility, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
- Metabolomics Core Facility, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium
| | - Johan Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
| | - Luc Schoonjans
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium
| | - Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium.
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium.
- Metaptys NV/Droia Labs, Leuven, Belgium.
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium.
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium.
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium.
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, B-3000, Belgium.
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
33
|
Usart M, Hansen N, Stetka J, Almeida Fonseca T, Guy A, Kimmerlin Q, Rai S, Hao-Shen H, Roux J, Dirnhofer S, Skoda RC. The glutaminase inhibitor CB-839 targets metabolic dependencies of JAK2-mutant hematopoiesis in MPN. Blood Adv 2024; 8:2312-2325. [PMID: 38295283 PMCID: PMC11117009 DOI: 10.1182/bloodadvances.2023010950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 01/04/2024] [Accepted: 01/21/2024] [Indexed: 02/02/2024] Open
Abstract
ABSTRACT Hyperproliferation of myeloid and erythroid cells in myeloproliferative neoplasms (MPN) driven by the JAK2-V617F mutation is associated with altered metabolism. Given the central role of glutamine in anabolic and catabolic pathways, we examined the effects of pharmacologically inhibiting glutaminolysis, that is, the conversion of glutamine (Gln) to glutamate (Glu), using CB-839, a small molecular inhibitor of the enzyme glutaminase (GLS). We show that CB-839 strongly reduced the mitochondrial respiration rate of bone marrow cells from JAK2-V617F mutant (VF) mice, demonstrating a marked dependence of these cells on Gln-derived ATP production. Consistently, in vivo treatment with CB-839 normalized blood glucose levels, reduced splenomegaly and decreased erythrocytosis in VF mice. These effects were more pronounced when CB-839 was combined with the JAK1/2 inhibitor ruxolitinib or the glycolysis inhibitor 3PO, indicating possible synergies when cotargeting different metabolic and oncogenic pathways. Furthermore, we show that the inhibition of glutaminolysis with CB-839 preferentially lowered the proportion of JAK2-mutant hematopoietic stem cells (HSCs). The total number of HSCs was decreased by CB-839, primarily by reducing HSCs in the G1 phase of the cell cycle. CB-839 in combination with ruxolitinib also strongly reduced myelofibrosis at later stages of MPN. In line with the effects shown in mice, proliferation of CD34+ hematopoietic stem and progenitor cells from polycythemia vera patients was inhibited by CB-839 at nanomolar concentrations. These data suggest that inhibiting GLS alone or in combination with inhibitors of glycolysis or JAK2 inhibitors represents an attractive new therapeutic approach to MPN.
Collapse
Affiliation(s)
- Marc Usart
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Nils Hansen
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Jan Stetka
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Tiago Almeida Fonseca
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Alexandre Guy
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- UMR1034, Inserm, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France
| | - Quentin Kimmerlin
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Shivam Rai
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Hui Hao-Shen
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Julien Roux
- Bioinformatics core facility, Department of Biomedicine, University of Basel, Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Stefan Dirnhofer
- Department of Pathology, University Hospital Basel, Basel, Switzerland
| | - Radek C. Skoda
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
34
|
Váchová L, Plocek V, Maršíková J, Rešetárová S, Hatáková L, Palková Z. Differential stability of Gcn4p controls its cell-specific activity in differentiated yeast colonies. mBio 2024; 15:e0068924. [PMID: 38624209 PMCID: PMC11077963 DOI: 10.1128/mbio.00689-24] [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: 03/07/2024] [Accepted: 03/22/2024] [Indexed: 04/17/2024] Open
Abstract
Gcn4p belongs to conserved AP-1 transcription factors involved in many cellular processes, including cell proliferation, stress response, and nutrient availability in yeast and mammals. AP-1 activities are regulated at different levels, such as translational activation or protein degradation, which increases the variability of regulation under different conditions. Gcn4p activity in unstructured yeast liquid cultures increases upon amino acid deficiency and is rapidly eliminated upon amino acid excess. Gcn2p kinase is the major described regulator of Gcn4p that enables GCN4 mRNA translation via the uORFs mechanism. Here, we show that Gcn4p is specifically active in U cells in the upper regions and inactive in L cells in the lower regions of differentiated colonies. Using in situ microscopy in combination with analysis of mutants and strains with GFP at different positions in the translational regulatory region of Gcn4p, we show that cell-specific Gcn4p activity is independent of Gcn2p or other translational or transcriptional regulation. Genetically, biochemically, and microscopically, we identified cell-specific proteasomal degradation as a key mechanism that diversifies Gcn4p function between U and L cells. The identified regulation leading to active Gcn4p in U cells with amino acids and efficient degradation in starved L cells differs from known regulations of Gcn4p in yeast but shows similarities to the activity of AP-1 ATF4 in mammals during insulin signaling. These findings may open new avenues for understanding the parallel activities of Gcn4p/ATF4 and reveal a novel biological role for cell type-specific regulation of proteasome-dependent degradation.IMPORTANCEIn nature, microbes usually live in spatially structured communities and differentiate into precisely localized, functionally specialized cells. The coordinated interplay of cells and their response to environmental changes, such as starvation, followed by metabolic adaptation, is critical for the survival of the entire community. Transcription factor Gcn4p is responsible for yeast adaptation under amino acid starvation in liquid cultures, and its activity is regulated mainly at the level of translation involving Gcn2p kinase. Whether Gcn4p functions in structured communities was unknown. We show that translational regulation of Gcn4p plays no role in the development of colony subpopulations; the main regulation occurs at the level of stabilization of the Gcn4p molecule in the cells of one subpopulation and its proteasomal degradation in the other. This regulation ensures specific spatiotemporal activity of Gcn4p in the colony. Our work highlights differences in regulatory networks in unorganized populations and organized structures of yeast, which in many respects resemble multicellular organisms.
Collapse
Affiliation(s)
- Libuše Váchová
- Institute of Microbiology of the Czech Academy of Sciences, BIOCEV, Prague, Czech Republic
| | - Vítězslav Plocek
- Faculty of Science, Charles University, BIOCEV, Prague, Czech Republic
| | - Jana Maršíková
- Faculty of Science, Charles University, BIOCEV, Prague, Czech Republic
| | - Stanislava Rešetárová
- Institute of Microbiology of the Czech Academy of Sciences, BIOCEV, Prague, Czech Republic
| | | | - Zdena Palková
- Faculty of Science, Charles University, BIOCEV, Prague, Czech Republic
| |
Collapse
|
35
|
Liu Y, Wu Z, Li Y, Chen Y, Zhao X, Wu M, Xia Y. Metabolic reprogramming and interventions in angiogenesis. J Adv Res 2024:S2090-1232(24)00178-4. [PMID: 38704087 DOI: 10.1016/j.jare.2024.05.001] [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: 03/15/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Endothelial cell (EC) metabolism plays a crucial role in the process of angiogenesis. Intrinsic metabolic events such as glycolysis, fatty acid oxidation, and glutamine metabolism, support secure vascular migration and proliferation, energy and biomass production, as well as redox homeostasis maintenance during vessel formation. Nevertheless, perturbation of EC metabolism instigates vascular dysregulation-associated diseases, especially cancer. AIM OF REVIEW In this review, we aim to discuss the metabolic regulation of angiogenesis by EC metabolites and metabolic enzymes, as well as prospect the possible therapeutic opportunities and strategies targeting EC metabolism. KEY SCIENTIFIC CONCEPTS OF REVIEW In this work, we discuss various aspects of EC metabolism considering normal and diseased vasculature. Of relevance, we highlight that the implications of EC metabolism-targeted intervention (chiefly by metabolic enzymes or metabolites) could be harnessed in orchestrating a spectrum of pathological angiogenesis-associated diseases.
Collapse
Affiliation(s)
- Yun Liu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zifang Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yikun Li
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yating Chen
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Xuan Zhao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Miaomiao Wu
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Yaoyao Xia
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
36
|
Masci D, Puxeddu M, Silvestri R, La Regina G. Metabolic Rewiring in Cancer: Small Molecule Inhibitors in Colorectal Cancer Therapy. Molecules 2024; 29:2110. [PMID: 38731601 PMCID: PMC11085455 DOI: 10.3390/molecules29092110] [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: 03/15/2024] [Revised: 04/16/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Alterations in cellular metabolism, such as dysregulation in glycolysis, lipid metabolism, and glutaminolysis in response to hypoxic and low-nutrient conditions within the tumor microenvironment, are well-recognized hallmarks of cancer. Therefore, understanding the interplay between aerobic glycolysis, lipid metabolism, and glutaminolysis is crucial for developing effective metabolism-based therapies for cancer, particularly in the context of colorectal cancer (CRC). In this regard, the present review explores the complex field of metabolic reprogramming in tumorigenesis and progression, providing insights into the current landscape of small molecule inhibitors targeting tumorigenic metabolic pathways and their implications for CRC treatment.
Collapse
Affiliation(s)
- Domiziana Masci
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy;
| | - Michela Puxeddu
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (R.S.)
| | - Romano Silvestri
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (R.S.)
| | - Giuseppe La Regina
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (R.S.)
| |
Collapse
|
37
|
Zhu G, Guan F, Li S, Zhang Q, Zhang X, Qin Y, Sun Z, Peng S, Cheng J, Li Y, Ren R, Fan T, Liu H. Glutaminase potentiates the glycolysis in esophageal squamous cell carcinoma by interacting with PDK1. Mol Carcinog 2024; 63:897-911. [PMID: 38353358 DOI: 10.1002/mc.23696] [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: 11/08/2023] [Revised: 12/14/2023] [Accepted: 01/24/2024] [Indexed: 04/13/2024]
Abstract
Increasing evidence has demonstrated that glutaminase (GLS) as a key mitochondrial enzyme plays a pivotal role in glutaminolysis, which widely participates in glutamine metabolism serving as main energy sources and building blocks for tumor growth. However, the roles and molecular mechanisms of GLS in esophageal squamous cell carcinoma (ESCC) remains unknown. Here, we found that GLS was highly expressed in ESCC tissues and cells. GLS inhibitor CB-839 significantly suppressed cell proliferation, colony formation, migration and invasion of ESCC cells, whereas GLS overexpression displayed the opposite effects. In addition, CB-839 markedly suppressed glucose consumption and lactate production, coupled with the downregulation of glycolysis-related proteins HK2, PFKM, PKM2 and LDHA, whereas GLS overexpression exhibited the adverse results. In vivo animal experiment revealed that CB-839 dramatically suppressed tumor growth, whereas GLS overexpression promoted tumor growth in ESCC cells xenografted nude mice. Mechanistically, GLS was localized in mitochondria of ESCC cells, which interacted with PDK1 protein. CB-839 attenuated the interaction of GLS and PDK1 in ESCC cells by suppressing PDK1 expression, which further evoked the downregulation of p-PDHA1 (s293), however, GLS overexpression markedly enhanced the level of p-PDHA1 (s293). These findings suggest that interaction of GLS with PDK1 accelerates the glycolysis of ESCC cells by inactivating PDH enzyme, and thus targeting GLS may be a novel therapeutic approach for ESCC patients.
Collapse
Affiliation(s)
- Guangzhao Zhu
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shenglei Li
- Department of Pathology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qing Zhang
- The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Translational Medicine Research Center Zhengzhou, Henan, China
| | - Xueying Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yue Qin
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhangzhan Sun
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shaohua Peng
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jiexing Cheng
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yiyang Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ruili Ren
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Tianli Fan
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Hongtao Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
38
|
He Q, Li J, Tao C, Zeng C, Liu C, Zheng Z, Mou S, Liu W, Zhang B, Yu X, Zhai Y, Wang J, Zhang Q, Zhang Y, Zhang D, Zhao J, Ge P. High glutamine increases stroke risk by inducing the endothelial-to-mesenchymal transition in moyamoya disease. MedComm (Beijing) 2024; 5:e525. [PMID: 38628905 PMCID: PMC11018113 DOI: 10.1002/mco2.525] [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: 07/09/2023] [Revised: 02/04/2024] [Accepted: 02/26/2024] [Indexed: 04/19/2024] Open
Abstract
At present, there is limited research on the mechanisms underlying moyamoya disease (MMD). Herein, we aimed to determine the role of glutamine in MMD pathogenesis, and 360 adult patients were prospectively enrolled. Human brain microvascular endothelial cells (HBMECs) were subjected to Integrin Subunit Beta 4 (ITGB4) overexpression or knockdown and atorvastatin. We assessed factors associated with various signaling pathways in the context of the endothelial-to-mesenchymal transition (EndMT), and the expression level of related proteins was validated in the superficial temporal arteries of patients. We found glutamine levels were positively associated with a greater risk of stroke (OR = 1.599, p = 0.022). After treatment with glutamine, HBMECs exhibited enhanced proliferation, migration, and EndMT, all reversed by ITGB4 knockdown. In ITGB4-transfected HBMECs, the MAPK-ERK-TGF-β/BMP pathway was activated, with Smad4 knockdown reversing the EndMT. Furthermore, atorvastatin suppressed the EndMT by inhibiting Smad1/5 phosphorylation and promoting Smad4 ubiquitination in ITGB4-transfected HBMECs. We also found the protein level of ITGB4 was upregulated in the superficial temporal arteries of patients with MMD. In conclusion, our study suggests that glutamine may be an independent risk factor for hemorrhage or infarction in patients with MMD and targeting ITGB4 could potentially be therapeutic approaches for MMD.
Collapse
Affiliation(s)
- Qiheng He
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Junsheng Li
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Chuming Tao
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Chaofan Zeng
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Chenglong Liu
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Zhiyao Zheng
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- Research Unit of Accurate Diagnosis, Treatment, and Translational Medicine of Brain TumorsChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of NeurosurgeryPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Siqi Mou
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Wei Liu
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Bojian Zhang
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Xiaofan Yu
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Yuanren Zhai
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Jia Wang
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- 3D Printing Center in Clinical NeuroscienceChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Qian Zhang
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Yan Zhang
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Dong Zhang
- Department of NeurosurgeryBeijing HospitalBeijingChina
| | - Jizong Zhao
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- 3D Printing Center in Clinical NeuroscienceChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Peicong Ge
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
39
|
Hasan Bou Issa L, Fléchon L, Laine W, Ouelkdite A, Gaggero S, Cozzani A, Tilmont R, Chauvet P, Gower N, Sklavenitis-Pistofidis R, Brinster C, Thuru X, Touil Y, Quesnel B, Mitra S, Ghobrial IM, Kluza J, Manier S. MYC dependency in GLS1 and NAMPT is a therapeutic vulnerability in multiple myeloma. iScience 2024; 27:109417. [PMID: 38510131 PMCID: PMC10952034 DOI: 10.1016/j.isci.2024.109417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/26/2023] [Accepted: 03/01/2024] [Indexed: 03/22/2024] Open
Abstract
Multiple myeloma (MM) is an incurable hematological malignancy in which MYC alterations contribute to the malignant phenotype. Nevertheless, MYC lacks therapeutic druggability. Here, we leveraged large-scale loss-of-function screens and conducted a small molecule screen to identify genes and pathways with enhanced essentiality correlated with MYC expression. We reported a specific gene dependency in glutaminase (GLS1), essential for the viability and proliferation of MYC overexpressing cells. Conversely, the analysis of isogenic models, as well as cell lines dataset (CCLE) and patient datasets, revealed GLS1 as a non-oncogenic dependency in MYC-driven cells. We functionally delineated the differential modulation of glutamine to maintain mitochondrial function and cellular biosynthesis in MYC overexpressing cells. Furthermore, we observed that pharmaceutical inhibition of NAMPT selectively affects MYC upregulated cells. We demonstrate the effectiveness of combining GLS1 and NAMPT inhibitors, suggesting that targeting glutaminolysis and NAD synthesis may be a promising strategy to target MYC-driven MM.
Collapse
Affiliation(s)
- Lama Hasan Bou Issa
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Léa Fléchon
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - William Laine
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Aicha Ouelkdite
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Silvia Gaggero
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Adeline Cozzani
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Remi Tilmont
- Department of Hematology, CHU Lille, 59000 Lille, France
| | - Paul Chauvet
- Department of Hematology, CHU Lille, 59000 Lille, France
| | - Nicolas Gower
- Department of Hematology, CHU Lille, 59000 Lille, France
| | | | - Carine Brinster
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Xavier Thuru
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Yasmine Touil
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Bruno Quesnel
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
- Department of Hematology, CHU Lille, 59000 Lille, France
| | - Suman Mitra
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Irene M. Ghobrial
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Jérôme Kluza
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Salomon Manier
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
- Department of Hematology, CHU Lille, 59000 Lille, France
| |
Collapse
|
40
|
Loopmans S, Tournaire G, Stockmans I, Stegen S, Carmeliet G. Hypoxia rewires glucose and glutamine metabolism in different sources of skeletal stem and progenitor cells similarly, except for pyruvate. J Bone Miner Res 2024; 39:150-160. [PMID: 38477776 DOI: 10.1093/jbmr/zjad016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 11/23/2023] [Accepted: 12/07/2023] [Indexed: 03/14/2024]
Abstract
Skeletal stem and progenitor cells (SSPCs) are crucial for bone development, homeostasis, and repair. SSPCs are considered to reside in a rather hypoxic niche in the bone, but distinct SSPC niches have been described in different skeletal regions, and they likely differ in oxygen and nutrient availability. Currently it remains unknown whether the different SSPC sources have a comparable metabolic profile and respond in a similar manner to hypoxia. In this study, we show that cell proliferation of all SSPCs was increased in hypoxia, suggesting that SSPCs can indeed function in a hypoxic niche in vivo. In addition, low oxygen tension increased glucose consumption and lactate production, but affected pyruvate metabolism cell-specifically. Hypoxia decreased tricarboxylic acid (TCA) cycle anaplerosis and altered glucose entry into the TCA cycle from pyruvate dehydrogenase to pyruvate carboxylase and/or malic enzyme. Finally, a switch from glutamine oxidation to reductive carboxylation was observed in hypoxia, as well as cell-specific adaptations in the metabolism of other amino acids. Collectively, our findings show that SSPCs from different skeletal locations proliferate adequately in hypoxia by rewiring glucose and amino acid metabolism in a cell-specific manner.
Collapse
Affiliation(s)
- Shauni Loopmans
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven 3000, Belgium
| | - Guillaume Tournaire
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven 3000, Belgium
| | - Ingrid Stockmans
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven 3000, Belgium
| | - Steve Stegen
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven 3000, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
41
|
Linehan A, O’Reilly M, McDermott R, O’Kane GM. Targeting KRAS mutations in pancreatic cancer: opportunities for future strategies. Front Med (Lausanne) 2024; 11:1369136. [PMID: 38576709 PMCID: PMC10991798 DOI: 10.3389/fmed.2024.1369136] [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: 01/11/2024] [Accepted: 02/22/2024] [Indexed: 04/06/2024] Open
Abstract
Targeting the RAS pathway remains the holy grail of precision oncology. In the case of pancreatic ductal adenocarcinomas (PDAC), 90-92% harbor mutations in the oncogene KRAS, triggering canonical MAPK signaling. The smooth structure of the altered KRAS protein without a binding pocket and its affinity for GTP have, in the past, hampered drug development. The emergence of KRASG12C covalent inhibitors has provided renewed enthusiasm for targeting KRAS. The numerous pathways implicated in RAS activation do, however, lead to the development of early resistance. In addition, the dense stromal niche and immunosuppressive microenvironment dictated by oncogenic KRAS can influence treatment responses, highlighting the need for a combination-based approach. Given that mutations in KRAS occur early in PDAC tumorigenesis, an understanding of its pleiotropic effects is key to progress in this disease. Herein, we review current perspectives on targeting KRAS with a focus on PDAC.
Collapse
Affiliation(s)
- Anna Linehan
- Department of Medical Oncology, St Vincent’s University Hospital, Dublin, Ireland
| | - Mary O’Reilly
- Department of Medical Oncology, St Vincent’s University Hospital, Dublin, Ireland
| | - Ray McDermott
- Department of Medical Oncology, St Vincent’s University Hospital, Dublin, Ireland
| | - Grainne M. O’Kane
- Department of Medical Oncology, St James’s Hospital, Dublin, Ireland
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
42
|
Ma Y, Tang R, Huang P, Li D, Liao M, Gao S. Mitochondrial energy metabolism-related gene signature as a prognostic indicator for pancreatic adenocarcinoma. Front Pharmacol 2024; 15:1332042. [PMID: 38572434 PMCID: PMC10987750 DOI: 10.3389/fphar.2024.1332042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/29/2024] [Indexed: 04/05/2024] Open
Abstract
Background: Pancreatic adenocarcinoma (PAAD) is a highly malignant gastrointestinal tumor and is associated with an unfavorable prognosis worldwide. Considering the effect of mitochondrial metabolism on the prognosis of pancreatic cancer has rarely been investigated, we aimed to establish prognostic gene markers associated with mitochondrial energy metabolism for the prediction of survival probability in patients with PAAD. Methods: Gene expression data were obtained from The Cancer Genome Atlas and Gene Expression Omnibus databases, and the mitochondrial energy metabolism-related genes were obtained from the GeneCards database. Based on mitochondrial energy metabolism score (MMs), differentially expressed MMRGs were established for MMs-high and MMs-low groups using ssGSEA. After the univariate Cox and least absolute and selection operator (LASSO) analyses, a prognostic MMRG signature was used in the multivariate Cox proportional regression model. Survival and immune cell infiltration analyses were performed. In addition, a nomogram based on the risk model was used to predict the survival probability of patients with PAAD. Finally, the expression of key genes was verified using quantitative polymerase chain reaction and immunohistochemical staining. Intro cell experiments were performed to evaluated the proliferation and invasion of pancreatic cancer cells. Results: A prognostic signature was constructed consisting of two mitochondrial energy metabolism-related genes (MMP11, COL10A1). Calibration and receiver operating characteristic (ROC) curves verified the good predictability performance of the risk model for the survival rate of patients with PAAD. Finally, immune-related analysis explained the differences in immune status between the two subgroups based on the risk model. The high-risk score group showed higher estimate, immune, and stromal scores, expression of eight checkpoint genes, and infiltration of M0 macrophages, which might indicate a beneficial response to immunotherapy. The qPCR results confirmed high expression of MMP11 in pancreatic cancer cell lines, and IHC also verified high expression of MMP11 in clinical pancreatic ductal adenocarcinoma tissues. In vitro cell experiments also demonstrated the role of MMP11 in cell proliferation and invasion. Conclusion: Our study provides a novel two-prognostic gene signature-based on MMRGs-that accurately predicted the survival of patients with PAAD and could be used for mitochondrial energy metabolism-related therapies in the future.
Collapse
Affiliation(s)
- Yu Ma
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Ronghao Tang
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Peilin Huang
- School of Medicine, Southeast University, Nanjing, China
| | - Danhua Li
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Meijian Liao
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Shoucui Gao
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
43
|
Clemente-Suárez VJ, Peris-Ramos HC, Redondo-Flórez L, Beltrán-Velasco AI, Martín-Rodríguez A, David-Fernandez S, Yáñez-Sepúlveda R, Tornero-Aguilera JF. Personalizing Nutrition Strategies: Bridging Research and Public Health. J Pers Med 2024; 14:305. [PMID: 38541047 PMCID: PMC10970995 DOI: 10.3390/jpm14030305] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 11/11/2024] Open
Abstract
In recent years, although life expectancy has increased significantly, non-communicable diseases (NCDs) continue to pose a significant threat to the health of the global population. Therefore, eating habits have been recognized as key modifiable factors that influence people's health and well-being. For this reason, it is interesting to study dietary patterns, since the human diet is a complex mixture of macronutrients, micronutrients, and bioactive compounds, and can modulate multiple physiological processes, including immune function, the metabolism, and inflammation. To ensure that the data we acquired were current and relevant, we searched primary and secondary sources, including scientific journals, bibliographic indexes, and databases in the last 15 years with the most relevant articles. After this search, we observed that all the recent research on NCDs suggests that diet is a critical factor in shaping an individual's health outcomes. Thus, cardiovascular, metabolic, mental, dental, and visual health depends largely on the intake, habits and patterns, and nutritional behaviors. A diet high in processed and refined foods, added sugars, and saturated fats can increase the risk of developing chronic diseases. On the other hand, a diet rich in whole, nutrient-dense foods, such as vegetables, fruits, nuts, legumes, and a high adherence to Mediterranean diet can improve health's people.
Collapse
Affiliation(s)
- Vicente Javier Clemente-Suárez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (V.J.C.-S.); (J.F.T.-A.)
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| | - Helia Carmen Peris-Ramos
- Faculty of Biomedical and Health Sciences, Clinical Odontology Department, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (H.C.P.-R.); (S.D.-F.)
| | - Laura Redondo-Flórez
- Department of Health Sciences, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Tajo Street, s/n, Villaviciosa de Odón, 28670 Madrid, Spain;
| | | | - Alexandra Martín-Rodríguez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (V.J.C.-S.); (J.F.T.-A.)
| | - Susana David-Fernandez
- Faculty of Biomedical and Health Sciences, Clinical Odontology Department, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (H.C.P.-R.); (S.D.-F.)
| | - Rodrigo Yáñez-Sepúlveda
- Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 2520000, Chile;
| | | |
Collapse
|
44
|
Muranaka H, Akinsola R, Billet S, Pandol SJ, Hendifar AE, Bhowmick NA, Gong J. Glutamine Supplementation as an Anticancer Strategy: A Potential Therapeutic Alternative to the Convention. Cancers (Basel) 2024; 16:1057. [PMID: 38473414 PMCID: PMC10930819 DOI: 10.3390/cancers16051057] [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: 02/09/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
Glutamine, a multifaceted nonessential/conditionally essential amino acid integral to cellular metabolism and immune function, holds pivotal importance in the landscape of cancer therapy. This review delves into the intricate dynamics surrounding both glutamine antagonism strategies and glutamine supplementation within the context of cancer treatment, emphasizing the critical role of glutamine metabolism in cancer progression and therapy. Glutamine antagonism, aiming to disrupt tumor growth by targeting critical metabolic pathways, is challenged by the adaptive nature of cancer cells and the complex metabolic microenvironment, potentially compromising its therapeutic efficacy. In contrast, glutamine supplementation supports immune function, improves gut integrity, alleviates treatment-related toxicities, and improves patient well-being. Moreover, recent studies highlighted its contributions to epigenetic regulation within cancer cells and its potential to bolster anti-cancer immune functions. However, glutamine implementation necessitates careful consideration of potential interactions with ongoing treatment regimens and the delicate equilibrium between supporting normal cellular function and promoting tumorigenesis. By critically assessing the implications of both glutamine antagonism strategies and glutamine supplementation, this review aims to offer comprehensive insights into potential therapeutic strategies targeting glutamine metabolism for effective cancer management.
Collapse
Affiliation(s)
- Hayato Muranaka
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (R.A.); (S.B.); (S.J.P.); (A.E.H.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Rasaq Akinsola
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (R.A.); (S.B.); (S.J.P.); (A.E.H.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sandrine Billet
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (R.A.); (S.B.); (S.J.P.); (A.E.H.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Stephen J. Pandol
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (R.A.); (S.B.); (S.J.P.); (A.E.H.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Andrew E. Hendifar
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (R.A.); (S.B.); (S.J.P.); (A.E.H.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Neil A. Bhowmick
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (R.A.); (S.B.); (S.J.P.); (A.E.H.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Research, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Jun Gong
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (R.A.); (S.B.); (S.J.P.); (A.E.H.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
45
|
Papadakos SP, Argyrou A, Lekakis V, Arvanitakis K, Kalisperati P, Stergiou IE, Konstantinidis I, Schizas D, Koufakis T, Germanidis G, Theocharis S. Metformin in Esophageal Carcinoma: Exploring Molecular Mechanisms and Therapeutic Insights. Int J Mol Sci 2024; 25:2978. [PMID: 38474224 DOI: 10.3390/ijms25052978] [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: 02/10/2024] [Revised: 02/29/2024] [Accepted: 03/03/2024] [Indexed: 03/14/2024] Open
Abstract
Esophageal cancer (EC) remains a formidable malignancy with limited treatment options and high mortality rates, necessitating the exploration of innovative therapeutic avenues. Through a systematic analysis of a multitude of studies, we synthesize the diverse findings related to metformin's influence on EC. This review comprehensively elucidates the intricate metabolic pathways and molecular mechanisms through which metformin may exert its anti-cancer effects. Key focus areas include its impact on insulin signaling, AMP-activated protein kinase (AMPK) activation, and the mTOR pathway, which collectively contribute to its role in mitigating esophageal cancer progression. This review critically examines the body of clinical and preclinical evidence surrounding the potential role of metformin, a widely prescribed anti-diabetic medication, in EC management. Our examination extends to the modulation of inflammation, oxidative stress and angiogenesis, revealing metformin's potential as a metabolic intervention in esophageal cancer pathogenesis. By consolidating epidemiological and clinical data, we assess the evidence that supports metformin's candidacy as an adjuvant therapy for esophageal cancer. By summarizing clinical and preclinical findings, our review aims to enhance our understanding of metformin's role in EC management, potentially improving patient care and outcomes.
Collapse
Affiliation(s)
- Stavros P Papadakos
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Goudi, 11527 Athens, Greece
| | - Alexandra Argyrou
- Academic Department of Gastroenterology, Laikon General Hospital, Athens University Medical School, 11527 Athens, Greece
| | - Vasileios Lekakis
- Academic Department of Gastroenterology, Laikon General Hospital, Athens University Medical School, 11527 Athens, Greece
| | - Konstantinos Arvanitakis
- First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
- Basic and Translational Research Unit (BTRU), Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Polyxeni Kalisperati
- Pathophysiology Department, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioanna E Stergiou
- Pathophysiology Department, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | | | - Dimitrios Schizas
- First Department of Surgery, Laikon General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Theocharis Koufakis
- Second Propaedeutic Department of Internal Medicine, General Hospital "Hippokration", Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Georgios Germanidis
- First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
- Basic and Translational Research Unit (BTRU), Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Goudi, 11527 Athens, Greece
| |
Collapse
|
46
|
Zhao M, Wang K, Lin R, Mu F, Cui J, Tao X, Weng Y, Wang J. Influence of glutamine metabolism on diabetes Development:A scientometric review. Heliyon 2024; 10:e25258. [PMID: 38375272 PMCID: PMC10875382 DOI: 10.1016/j.heliyon.2024.e25258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
Objective "Metabolism affects function" is the consensus of researchers at present. It has potential clinical application value to study the effects of regulating glutamine (Gln) metabolism on diabetes physiology or pathology. Our research aimed to summarize the latest research progress, frontier hot topics and future development trends in this field from the perspective of scientometrics. Methods Relevant literatures and reviews were obtained from the Web of Science (WoS) between January 1, 2001 and May 31, 2022. An online analysis platform of bibliometrics, CiteSpace, and VOS viewer software were used to generate visual knowledge network graphs, including publication countries, institutions and authors partnership analysis, co-occurrence analysis, co-citation analysis, as well as citations and keywords burst detection to acquire research trends and hotspots. Results Our results showed that a total of 945 publications in the WoS database met the analysis requirements, with articles being the main type. The overall characteristics showed an increasing trend in the number of publications and citations. The United States was leading the way in this research and was a hub for aggregating collaborations across countries. Vanderbilt University delivered high-quality impact with the most published articles. DeBerardinis, RJ in this field was the most representative author and his main research contents were Gln metabolism and mitochondrial glutaminolysis. Significantly, there was a relative lack of collaboration between institutions and authors. In addition, "type 2 diabetes", "glutamine", "metabolism", "gene expression" and "metabolomics" were the keywords categories with high frequency in co-citation references and co-occurrence cluster keywords. Analysis of popular keywords burst detection showed that "branched chain", "oxidative phosphorylation", "kinase", "insulin sensitivity", "tca cycle", "magnetic resonance spectroscopy" and "flux analysis" were new research directions and emerging methods to explore the link between Gln metabolism and diabetes. Overall, exploring Gln metabolism showed a gradual upward trend in the field of diabetes. Conclusion This comprehensive scientometric study identified the general outlook for the field and provided valuable guidance for ongoing research. Strategies to regulate Gln metabolism hold promise as a novel target to treat diabetes, as well as integration and intersection of multidisciplinary provides cooperation strategies and technical guarantees for the development of this field.
Collapse
Affiliation(s)
- Meina Zhao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032 Shannxi Province, China
| | - Kaiyan Wang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, 710032 Shannxi Province, China
| | - Rui Lin
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032 Shannxi Province, China
| | - Fei Mu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032 Shannxi Province, China
| | - Jia Cui
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032 Shannxi Province, China
| | - Xingru Tao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032 Shannxi Province, China
| | - Yan Weng
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032 Shannxi Province, China
| | - Jingwen Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032 Shannxi Province, China
| |
Collapse
|
47
|
Lazen P, Lima Cardoso P, Sharma S, Cadrien C, Roetzer-Pejrimovsky T, Furtner J, Strasser B, Hingerl L, Lipka A, Preusser M, Marik W, Bogner W, Widhalm G, Rössler K, Trattnig S, Hangel G. A Comparison of 7 Tesla MR Spectroscopic Imaging and 3 Tesla MR Fingerprinting for Tumor Localization in Glioma Patients. Cancers (Basel) 2024; 16:943. [PMID: 38473305 DOI: 10.3390/cancers16050943] [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: 12/08/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
This paper investigated the correlation between magnetic resonance spectroscopic imaging (MRSI) and magnetic resonance fingerprinting (MRF) in glioma patients by comparing neuro-oncological markers obtained from MRSI to T1/T2 maps from MRF. Data from 12 consenting patients with gliomas were analyzed by defining hotspots for T1, T2, and various metabolic ratios, and comparing them using Sørensen-Dice similarity coefficients (DSCs) and the distances between their centers of intensity (COIDs). The median DSCs between MRF and the tumor segmentation were 0.73 (T1) and 0.79 (T2). The DSCs between MRSI and MRF were the highest for Gln/tNAA (T1: 0.75, T2: 0.80, tumor: 0.78), followed by Gly/tNAA (T1: 0.57, T2: 0.62, tumor: 0.54) and tCho/tNAA (T1: 0.61, T2: 0.58, tumor: 0.45). The median values in the tumor hotspot were T1 = 1724 ms, T2 = 86 ms, Gln/tNAA = 0.61, Gly/tNAA = 0.28, Ins/tNAA = 1.15, and tCho/tNAA = 0.48, and, in the peritumoral region, were T1 = 1756 ms, T2 = 102 ms, Gln/tNAA = 0.38, Gly/tNAA = 0.20, Ins/tNAA = 1.06, and tCho/tNAA = 0.38, and, in the NAWM, were T1 = 950 ms, T2 = 43 ms, Gln/tNAA = 0.16, Gly/tNAA = 0.07, Ins/tNAA = 0.54, and tCho/tNAA = 0.20. The results of this study constitute the first comparison of 7T MRSI and 3T MRF, showing a good correspondence between these methods.
Collapse
Affiliation(s)
- Philipp Lazen
- High-Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
- Department for Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
- Christian Doppler Laboratory for MR Imaging Biomarkers, 1090 Vienna, Austria
| | - Pedro Lima Cardoso
- High-Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Sukrit Sharma
- High-Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Cornelius Cadrien
- High-Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
- Department for Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Roetzer-Pejrimovsky
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Julia Furtner
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
- Research Center for Medical Image Analysis and Artificial Intelligence (MIAAI), Danube Private University, 3500 Krems, Austria
| | - Bernhard Strasser
- High-Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Lukas Hingerl
- High-Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Alexandra Lipka
- High-Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Internal Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Wolfgang Marik
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Wolfgang Bogner
- High-Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
- Christian Doppler Laboratory for MR Imaging Biomarkers, 1090 Vienna, Austria
| | - Georg Widhalm
- Department for Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Karl Rössler
- Department for Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
- Christian Doppler Laboratory for MR Imaging Biomarkers, 1090 Vienna, Austria
| | - Siegfried Trattnig
- High-Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
- Christian Doppler Laboratory for MR Imaging Biomarkers, 1090 Vienna, Austria
- Institute for Clinical Molecular MRI, Karl Landsteiner Society, 3100 St. Pölten, Austria
| | - Gilbert Hangel
- High-Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
- Department for Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
- Christian Doppler Laboratory for MR Imaging Biomarkers, 1090 Vienna, Austria
| |
Collapse
|
48
|
Guz W, Podgórski R, Bober Z, Aebisher D, Truszkiewicz A, Olek M, Machorowska Pieniążek A, Kawczyk-Krupka A, Bartusik-Aebisher D. In Vitro MRS of Cells Treated with Trastuzumab at 1.5 Tesla. Int J Mol Sci 2024; 25:1719. [PMID: 38338997 PMCID: PMC10855746 DOI: 10.3390/ijms25031719] [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: 12/30/2023] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
The aim of the study was to investigate the effect of Trastuzumab on the MCF-7 and CRL-2314 breast cancer cell lines. Additionally, an attempt was made to optimize magnetic resonance spectroscopy (MRS) for cell culture studies, with particular emphasis on the impact of treatment with Trastuzumab. The research materials included MCF-7 and CRL-2314 breast cancer cell lines. The study examined the response of these cell lines to treatment with Trastuzumab. The clinical magnetic resonance imaging (MRI) system, OPTIMA MR360 manufactured by GEMS, with a magnetic field induction of 1.5 T, was used. Due to the nature of the tested objects, their size and shape, it was necessary to design and manufacture additional receiving coils. They were used to image the tested cell cultures and record the spectroscopic signal. The spectra obtained by MRS were confirmed by NMR using a 300 MHz NMR Fourier 300 with the TopSpin 3.1 system from Bruker. The designed receiving coils allowed for conducting experiments with the cell lines in a satisfactory manner. These tests would not be possible using factory-delivered coils due to their parameters and the size of the test objects, whose volume did not exceed 1 mL. MRS studies revealed an increase in the metabolite at 1.9 ppm, which indicates the induction of histone acetylation. Changes in histone acetylation play a very important role in both cell development and differentiation processes. The use of Trastuzumab therapy in breast cancer cells increases the levels of acetylated histones. MRS studies and spectra obtained from the 300 MHz NMR system are consistent with the specificity inherent in both systems.
Collapse
Affiliation(s)
- Wiesław Guz
- Department of Diagnostic Imaging and Nuclear Medicine, Medical College of Rzeszów University, 35-959 Rzeszów, Poland;
| | - Rafal Podgórski
- Department of Biochemistry and General Chemistry, Medical College of Rzeszów University, 35-959 Rzeszów, Poland; (R.P.); (D.B.-A.)
| | - Zuzanna Bober
- Department of Photomedicine and Physical Chemistry, Medical College of Rzeszów University, 35-959 Rzeszów, Poland; (Z.B.); (A.T.)
| | - David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of Rzeszów University, 35-959 Rzeszów, Poland; (Z.B.); (A.T.)
| | - Adrian Truszkiewicz
- Department of Photomedicine and Physical Chemistry, Medical College of Rzeszów University, 35-959 Rzeszów, Poland; (Z.B.); (A.T.)
| | - Marcin Olek
- Department of Densitry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland (A.M.P.)
| | - Agnieszka Machorowska Pieniążek
- Department of Densitry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland (A.M.P.)
| | - Aleksandra Kawczyk-Krupka
- Department of Internal Medicine, Angiology and Physical Medicine, Center for Laser Diagnostics and Therapy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
| | - Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of Rzeszów University, 35-959 Rzeszów, Poland; (R.P.); (D.B.-A.)
| |
Collapse
|
49
|
Shi H, Yuan X, Yang X, Huang R, Fan W, Liu G. A novel diabetic foot ulcer diagnostic model: identification and analysis of genes related to glutamine metabolism and immune infiltration. BMC Genomics 2024; 25:125. [PMID: 38287255 PMCID: PMC10826017 DOI: 10.1186/s12864-024-10038-2] [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: 11/07/2023] [Accepted: 01/22/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Diabetic foot ulcer (DFU) is one of the most common and severe complications of diabetes, with vascular changes, neuropathy, and infections being the primary pathological mechanisms. Glutamine (Gln) metabolism has been found to play a crucial role in diabetes complications. This study aims to identify and validate potential Gln metabolism biomarkers associated with DFU through bioinformatics and machine learning analysis. METHODS We downloaded two microarray datasets related to DFU patients from the Gene Expression Omnibus (GEO) database, namely GSE134431, GSE68183, and GSE80178. From the GSE134431 dataset, we obtained differentially expressed Gln-metabolism related genes (deGlnMRGs) between DFU and normal controls. We analyzed the correlation between deGlnMRGs and immune cell infiltration status. We also explored the relationship between GlnMRGs molecular clusters and immune cell infiltration status. Notably, WGCNA to identify differentially expressed genes (DEGs) within specific clusters. Additionally, we conducted GSVA to annotate enriched genes. Subsequently, we constructed and screened the best machine learning model. Finally, we validated the predictions' accuracy using a nomogram, calibration curves, decision curve analysis (DCA), and the GSE134431, GSE68183, and GSE80178 dataset. RESULTS In both the DFU and normal control groups, we confirmed the presence of deGlnMRGs and an activated immune response. From the GSE134431 dataset, we obtained 20 deGlnMRGs, including CTPS1, NAGS, SLC7A11, GGT1, GCLM, RIMKLA, ARG2, ASL, ASNS, ASNSD1, PPAT, GLS2, GLUD1, MECP2, ASS1, PRODH, CTPS2, ALDH5A1, DGLUCY, and SLC25A12. Furthermore, two clusters were identified in DFU. Immune infiltration analysis indicated the presence of immune heterogeneity in these two clusters. Additionally, we established a Support Vector Machine (SVM) model based on 5 genes (R3HCC1, ZNF562, MFN1, DRAM1, and PTGDS), which exhibited excellent performance on the external validation datasetGSE134431, GSE68183, and GSE80178 (AUC = 0.929). CONCLUSION This study has identified five Gln metabolism genes associated with DFU, revealing potential novel biomarkers and therapeutic targets for DFU. Additionally, the infiltration of immune-inflammatory cells plays a crucial role in the progression of DFU.
Collapse
Affiliation(s)
- Hongshuo Shi
- Department of Peripheral Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Yuan
- Department of Peripheral Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao Yang
- Department of Peripheral Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Renyan Huang
- Department of Peripheral Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Weijing Fan
- Department of Peripheral Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Guobin Liu
- Department of Peripheral Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Guangming Traditional Chinese Medicine Hospital Pudong New Area, Shanghai, China.
| |
Collapse
|
50
|
Peinado RDS, Martins LG, Pacca CC, Saivish MV, Borsatto KC, Nogueira ML, Tasic L, Arni RK, Eberle RJ, Coronado MA. HR-MAS NMR Metabolomics Profile of Vero Cells under the Influence of Virus Infection and nsP2 Inhibitor: A Chikungunya Case Study. Int J Mol Sci 2024; 25:1414. [PMID: 38338694 PMCID: PMC10855909 DOI: 10.3390/ijms25031414] [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: 12/16/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 02/12/2024] Open
Abstract
The arbovirus Chikungunya (CHIKV) is transmitted by Aedes mosquitoes in urban environments, and in humans, it triggers debilitating symptoms involving long-term complications, including arthritis and Guillain-Barré syndrome. The development of antiviral therapies is relevant, as no efficacious vaccine or drug has yet been approved for clinical application. As a detailed map of molecules underlying the viral infection can be obtained from the metabolome, we validated the metabolic signatures of Vero E6 cells prior to infection (CC), following CHIKV infection (CV) and also upon the inclusion of the nsP2 protease inhibitor wedelolactone (CWV), a coumestan which inhibits viral replication processes. The metabolome groups evidenced significant changes in the levels of lactate, myo-inositol, phosphocholine, glucose, betaine and a few specific amino acids. This study forms a preliminary basis for identifying metabolites through HR-MAS NMR (High Resolution Magic Angle Spinning Nuclear Magnetic Ressonance Spectroscopy) and proposing the affected metabolic pathways of cells following viral infection and upon incorporation of putative antiviral molecules.
Collapse
Affiliation(s)
- Rafaela dos S. Peinado
- Multiuser Center for Biomolecular Innovation, Department of Physics, Institute of Biosciences, Languages and Exact Sciences (Ibilce—UNESP), Sao Jose do Rio Preto, Sao Paulo 15054000, Brazil; (R.d.S.P.); (K.C.B.); (R.K.A.)
| | - Lucas G. Martins
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas 13083862, Brazil; (L.G.M.); (L.T.)
| | - Carolina C. Pacca
- Virology Research Laboratory, Medical School of Sao Jose do Rio Preto (FAMERP), Sao Paulo 15090000, Brazil; (C.C.P.); (M.V.S.); (M.L.N.)
| | - Marielena V. Saivish
- Virology Research Laboratory, Medical School of Sao Jose do Rio Preto (FAMERP), Sao Paulo 15090000, Brazil; (C.C.P.); (M.V.S.); (M.L.N.)
| | - Kelly C. Borsatto
- Multiuser Center for Biomolecular Innovation, Department of Physics, Institute of Biosciences, Languages and Exact Sciences (Ibilce—UNESP), Sao Jose do Rio Preto, Sao Paulo 15054000, Brazil; (R.d.S.P.); (K.C.B.); (R.K.A.)
| | - Maurício L. Nogueira
- Virology Research Laboratory, Medical School of Sao Jose do Rio Preto (FAMERP), Sao Paulo 15090000, Brazil; (C.C.P.); (M.V.S.); (M.L.N.)
| | - Ljubica Tasic
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas 13083862, Brazil; (L.G.M.); (L.T.)
| | - Raghuvir K. Arni
- Multiuser Center for Biomolecular Innovation, Department of Physics, Institute of Biosciences, Languages and Exact Sciences (Ibilce—UNESP), Sao Jose do Rio Preto, Sao Paulo 15054000, Brazil; (R.d.S.P.); (K.C.B.); (R.K.A.)
| | - Raphael J. Eberle
- Institute of Biological Information Processing IBI-7: Structural Biochemistry, Forschungszentrum Jülich, 52428 Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Mônika A. Coronado
- Institute of Biological Information Processing IBI-7: Structural Biochemistry, Forschungszentrum Jülich, 52428 Jülich, Germany
| |
Collapse
|