1
|
Patel S, Porcari P, Coffee E, Kim N, Berishaj M, Peyear T, Zhang G, Keshari KR. Simultaneous noninvasive quantification of redox and downstream glycolytic fluxes reveals compartmentalized brain metabolism. SCIENCE ADVANCES 2024; 10:eadr2058. [PMID: 39705365 DOI: 10.1126/sciadv.adr2058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/14/2024] [Indexed: 12/22/2024]
Abstract
Brain metabolism across anatomic regions and cellular compartments plays an integral role in many aspects of neuronal function. Changes in key metabolic pathway fluxes, including oxidative and reductive energy metabolism, have been implicated in a wide range of brain diseases. Given the complex nature of the brain and the need for understanding compartmentalized metabolism noninvasively in vivo, new tools are required. Herein, using hyperpolarized (HP) magnetic resonance imaging coupled with in vivo isotope tracing, we develop a platform to simultaneously probe redox and energy metabolism in the murine brain. By combining HP dehydroascorbate and pyruvate, we are able to visualize increased lactate production in the white matter and increased redox capacity in the deep gray matter. Leveraging positional labeling, we show differences in compartmentalized tricarboxylic acid cycle entry versus downstream flux to glutamate. These findings lay the foundation for clinical translation of the proposed approach to probe brain metabolism.
Collapse
Affiliation(s)
- Saket Patel
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paola Porcari
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elizabeth Coffee
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nathaniel Kim
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marjan Berishaj
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thasin Peyear
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Guannan Zhang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kayvan R Keshari
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
2
|
Wang W, Cheng Y, Meng Q, Jia B, Yao D, Cheng Y. The additive interaction of healthy lifestyles and genetic susceptibility on colorectal cancer risk in prediabetes: a large population-based prospective cohort study. BMC Gastroenterol 2024; 24:462. [PMID: 39696080 DOI: 10.1186/s12876-024-03552-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVE We aimed to investigate the interrelationships among polygenic risk scores (PRS), healthy lifestyle factors (HLFs), and colorectal cancer (CRC) risk in individuals with prediabetes. To investigate whether adherence to HLFs influence CRC risk in those with elevated PRS within this specific population. METHODS Data from 22,408 prediabetes participants without CRC at baseline were analyzed from the UK Biobank. HLFs were graded using healthy lifestyle scores (HLSs) and classified as favorable, intermediate, or unfavorable, while the PRS for CRC was categorized as high, medium, or low. Cox proportional hazards models were used to calculate hazard ratios (HR) and 95% confidence intervals (CI) for CRC risk. RESULTS High PRS (HR: 2.36; 95% CI: 1.86-3.00) and medium PRS (HR: 1.42; 95% CI: 1.09-1.83) prediabetes were associated with increased CRC risk compared to those with low PRS. HLFs were linked to lower CRC risk in a dose-response manner, with never smoking (HR: 0.69; 95% CI: 0.57-0.84) and maintaining a healthy BMI (HR: 0.64; 95% CI: 0.49-0.82) associated with reduced CRC risk. Adherence to favorable HLFs may reduce the CRC risk in those with medium (HR: 0.51; 95% CI: 0.27-0.95) and high PRS (HR: 0.62; 95% CI: 0.39-0.99) over 15 years of follow-up. In participants with high PRS and unfavorable HLFs, the excess risk due to the additive interaction between PRS and HLFs was 1.41% (p < 0.01), especially for women (1.07%). CONCLUSIONS There is an additive interaction of PRS and HLFs on CRC risk in individuals with prediabetes. Adopting favorable HLFs should be integrated into the management of prediabetes individuals to reduce the risk of CRC.
Collapse
Affiliation(s)
- Wenchen Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yikang Cheng
- The First Clinical Institute, Zunyi Medical University, Zunyi, 563000, China
| | - Qingyu Meng
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Baoqing Jia
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Dawei Yao
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an, Jiaotong University Health Science Center, Xi'an, China.
| | - Yiping Cheng
- Department of Endocrinology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250021, China.
| |
Collapse
|
3
|
Saitoh Y, Takeda K, Okawachi K, Tanimura Y. High dose of ascorbic acid induces selective cell growth inhibition and cell death in human gastric signet-ring cell carcinoma-derived NUGC-4 cells. Biochim Biophys Acta Gen Subj 2024; 1869:130738. [PMID: 39675589 DOI: 10.1016/j.bbagen.2024.130738] [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: 10/21/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
Anticancer effects of high-dose vitamin C (VC) have been evaluated on many cancer cell lines, and its efficacy in clinical trials and in combination with anticancer drugs or radiation have been reported; however, its effect on gastric cancer and its mechanisms remain unclear. In the present study, the cell growth inhibitory/lethal effects of high-dose ascorbic acid (AsA), a reduced form of VC was examined on three gastric cancer cell lines. Of these, signet ring cell carcinoma NUGC-4 cells were the most sensitive, but the effects were small and limited in normal cells. Second, high-dose AsA was effective in NUGC-4 cells, whereas dehydroascorbic acid, an oxidized form of VC, was less effective. Third, high-dose AsA showed stronger cell growth inhibitory/lethal effects on floating cells than on adherent cells, and was effective even under hypoxic microenvironment conditions. A single 1-h treatment of high-dose AsA strongly inhibited cell growth, causing apoptosis-like cell death over 72 h after treatment, triggered by hydrogen peroxide generation, actin abnormality, DNA synthesis suppression, DNA damage induction, and ATP level decrease. The effects of high-dose AsA were inhibited either by adding or chelating iron ions, but was not affected via inhibiting AsA transport. Inhibition of glutathione synthesis enhanced the anticancer effects of high-dose AsA. These results indicate that a single high-dose of AsA induces cancer cell-selective, sustained cell growth inhibition and cell death, and these effects may be regulated by iron ion and/or intracellular oxidative stress levels in human gastric signet-ring cell carcinoma-derived NUGC-4 cells.
Collapse
Affiliation(s)
- Yasukazu Saitoh
- Program in Biological System Sciences Graduate School of Comprehensive Scientific Research, Prefectural University of Hiroshima, 5562, Nanatsuka, Shobara, Hiroshima 727-0023, Japan; Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, 5562, Nanatsuka, Shobara, Hiroshima 727-0023, Japan.
| | - Kaori Takeda
- Program in Biological System Sciences Graduate School of Comprehensive Scientific Research, Prefectural University of Hiroshima, 5562, Nanatsuka, Shobara, Hiroshima 727-0023, Japan
| | - Koichi Okawachi
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, 5562, Nanatsuka, Shobara, Hiroshima 727-0023, Japan
| | - Yusuke Tanimura
- Program in Biological System Sciences Graduate School of Comprehensive Scientific Research, Prefectural University of Hiroshima, 5562, Nanatsuka, Shobara, Hiroshima 727-0023, Japan
| |
Collapse
|
4
|
Cui Z, Wang H, Feng X, Wu C, Yi M, He R, Pan T, Gao R, Feng L, Zeng B, Huang G, Wang Y, Du Y, Zhang CJ, Xiao X, Wang C. MYO1F regulates T-cell activation and glycolytic metabolism by promoting the acetylation of GAPDH. Cell Mol Immunol 2024:10.1038/s41423-024-01247-6. [PMID: 39668163 DOI: 10.1038/s41423-024-01247-6] [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: 09/05/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024] Open
Abstract
Proper cellular metabolism in T cells is critical for a productive immune response. However, when dysregulated by intrinsic or extrinsic metabolic factors, T cells may contribute to a wide spectrum of diseases, such as cancers and autoimmune diseases. However, the metabolic regulation of T cells remains incompletely understood. Here, we show that MYO1F is required for human and mouse T-cell activation after TCR stimulation and that T-cell-specific Myo1f knockout mice exhibit an increased tumor burden and attenuated EAE severity due to impaired T-cell activation in vivo. Mechanistically, after TCR stimulation, MYO1F is phosphorylated by LCK at tyrosines 607 and 634, which is critical for glyceraldehyde-3-phosphate dehydrogenase (GAPDH) acetylation at Lys84, 86 and 227 mediated by α-TAT1, which is an acetyltransferase, and these processes are important for its activation, cellular glycolysis and thus the effector function of T cells. Importantly, we show that a fusion protein of VAV1-MYO1F, a recurrent peripheral T-cell lymphoma (PTCL)-associated oncogenic protein, promotes hyperacetylation of GAPDH and its activation, which leads to aberrant glycolysis and T-cell proliferation, and that inhibition of the activity of GAPDH significantly limits T-cell activation and proliferation and extends the survival of hVAV1-MYO1F knock-in mice. Moreover, hyperacetylation of GAPDH was confirmed in human PTCL patient samples containing the VAV1-MYO1F gene fusion. Overall, this study revealed not only the mechanisms by which MYO1F regulates T-cell metabolism and VAV1-MYO1F fusion-induced PTCL but also promising therapeutic targets for the treatment of PTCL.
Collapse
Affiliation(s)
- Zhihui Cui
- Key Laboratory of Molecular Biophysics of the Ministry of Education, National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074; Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Heping Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074; Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiong Feng
- Key Laboratory of Molecular Biophysics of the Ministry of Education, National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074; Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chuyu Wu
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ming Yi
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ruirui He
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ting Pan
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ru Gao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074; Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lingyun Feng
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Bo Zeng
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Guoling Huang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yuan Wang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yanyun Du
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China.
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| | - Cun-Jin Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Xue Xiao
- Department of Pathology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Chenhui Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China.
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| |
Collapse
|
5
|
O’Reilly A, Zhao W, Wickström S, Arnér ESJ, Kiessling R. Reactive oxygen species: Janus-faced molecules in the era of modern cancer therapy. J Immunother Cancer 2024; 12:e009409. [PMID: 39645234 PMCID: PMC11629020 DOI: 10.1136/jitc-2024-009409] [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: 10/05/2024] [Accepted: 11/11/2024] [Indexed: 12/09/2024] Open
Abstract
Oxidative stress, that is, an unbalanced increase in reactive oxygen species (ROS), contributes to tumor-induced immune suppression and limits the efficacy of immunotherapy. Cancer cells have inherently increased ROS production, intracellularly through metabolic perturbations and extracellularly through activation of NADPH oxidases, which promotes cancer progression. Further increased ROS production or impaired antioxidant systems, induced, for example, by chemotherapy or radiotherapy, can preferentially kill cancer cells over healthy cells. Inflammatory cell-derived ROS mediate immunosuppressive effects of myeloid-derived suppressor cells and activated granulocytes, hampering antitumor effector cells such as T cells and natural killer (NK) cells. Cancer therapies modulating ROS levels in tumors may thus have entirely different consequences when targeting cancer cells versus immune cells. Here we discuss the possibility of developing more efficient cancer therapies based on reduction-oxidation modulation, as either monotherapies or in combination with immunotherapy. Short-term, systemic administration of antioxidants or drugs blocking ROS production can boost the immune system and act in synergy with immunotherapy. However, prolonged use of antioxidants can instead enhance tumor progression. Alternatives to systemic antioxidant administration are under development where gene-modified or activated T cells and NK cells are shielded ex vivo against the harmful effects of ROS before the infusion to patients with cancer.
Collapse
Affiliation(s)
- Aine O’Reilly
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine, University College Cork, Cork, Ireland
- The Christie NHS Foundation Trust, Manchester, UK
| | - Wenchao Zhao
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Stina Wickström
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Patient area Head and Neck, Lung and Skin, Karolinska University Hospital, Stockholm, Sweden
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Selenoprotein Research and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
| | - Rolf Kiessling
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Patient area Head and Neck, Lung and Skin, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
6
|
Locasale JW, Goncalves MD, Di Tano M, Burgos-Barragan G. Diet and Cancer Metabolism. Cold Spring Harb Perspect Med 2024; 14:a041549. [PMID: 38621831 PMCID: PMC11610756 DOI: 10.1101/cshperspect.a041549] [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: 04/17/2024]
Abstract
Diet and exercise are modifiable lifestyle factors known to have a major influence on metabolism. Clinical practice addresses diseases of altered metabolism such as diabetes or hypertension by altering these factors. Despite enormous public interest, there are limited defined diet and exercise regimens for cancer patients. Nevertheless, the molecular basis of cancer has converged over the past 15 years on an essential role for altered metabolism in cancer. However, our understanding of the molecular mechanisms that underlie the impact of diet and exercise on cancer metabolism is in its very early stages. In this work, we propose conceptual frameworks for understanding the consequences of diet and exercise on cancer cell metabolism and tumor biology and also highlight recent developments. By advancing our mechanistic understanding, we also discuss actionable ways that such interventions could eventually reach the mainstay of both medical oncology and cancer control and prevention.
Collapse
Affiliation(s)
- Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, Durham, Norh Carolina 27710, USA
| | - Marcus D Goncalves
- Division of Endocrinology, Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | - Maira Di Tano
- Division of Endocrinology, Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | - Guillermo Burgos-Barragan
- Department of Pharmacology, Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10056, USA
| |
Collapse
|
7
|
Woubshete M, Chan LI, Diallinas G, Byrne B. The dimer of human SVCT1 is key for transport function. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184390. [PMID: 39369805 DOI: 10.1016/j.bbamem.2024.184390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/06/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Humans and other primates lack the ability to synthesize the essential nutrient, Vitamin C, which is derived exclusively from the diet. Crucial for effective vitamin C uptake are the Na+ dependent Vitamin C transporters, SVCT1 and SVCT2, members of the nucleobase ascorbate transporter (NAT) family. SVCT1 and 2 actively transport the reduced form of Vitamin C, ascorbic acid, into key tissues. The recent structure of the mouse SVCT1 revealed the molecular basis of substrate binding and that, like the other structurally characterised members of the NAT family, it exists as a closely associated dimer. SVCT1 is likely to function via the elevator mechanism with the core domain of each protomer able to bind substrate and move through the membrane carrying the substrate across the membrane. Here we explored the function of a range of variants of the human SVCT1, revealing a range of residues involved in substrate selection and binding, and confirming the importance of the C-terminus in membrane localisation. Furthermore, using a dominant negative mutant we show that the dimer is essential for transport function, as previously seen in the fungal homologue, UapA. In addition, we show that a localisation deficient C-terminal truncation of SVCT1 blocks correct localisation of co-expressed, associated wildtype SVCT1. These results clearly show the importance of the dimer in both correct SVCT1 trafficking and transport activity.
Collapse
Affiliation(s)
- Menebere Woubshete
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Lok I Chan
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - George Diallinas
- Department of Biology, National and Kapodistrian University of Athens, Panepistimioupolis, 15784 Athens, Greece; Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, 70013 Heraklion, Greece
| | - Bernadette Byrne
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK.
| |
Collapse
|
8
|
Zhang LL, Li L, Wang D, Hong Y, Tang K, Hong J, Chen Z, Yang W, Lu L, Duan LY. Rapid redox-response featured visual ascorbic acid sensor based on simple-assembled europium metal-organic framework. Food Chem 2024; 459:140339. [PMID: 38986206 DOI: 10.1016/j.foodchem.2024.140339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
A facile, fast and visible sensing platform for ascorbic acid (AA) detection has been developed based on self-assembled hydrangea-like europium metal-organic framework (HL-EuMOF). HL-EuMOF was synthesized through a simple one-step mixing process with Eu3+ and 1, 10-phenanthroline-2, 9-dicarboxylic acid at room temperature, which exhibited excellent properties including strong red fluorescence, long decay lifetime (548.623 μs) and good luminescent stability. Based on the specific redox reaction between Fe3+ and AA, the HL-EuMOF@Fe3+ was fabricated with "turn-off" response for AA, where the resulting Fe2+ displayed effective fluorescence quenching ability toward HL-EuMOF. The sensor demonstrated low detection limit (31.94 nM), rapid response time (30 s) and high selectivity. Integration of smartphone-assisted RGB analysis with HL-EuMOF@Fe3+ permitted convenient and visible quantitative determination of AA level. This approach also presented good detection performances in complex human serum and beverage samples, which could provide a valuable tool for AA detection in biomedical research and food industry.
Collapse
Affiliation(s)
- Lin-Lin Zhang
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Li Li
- Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Ministry of Education, Key Laboratory of Chemical Utilization of Plant Resources of Nanchang, College of Chemistry and Materials, College of Science, Jiangxi Agricultural University, Nanchang 330045, China
| | - Dan Wang
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Yanping Hong
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Kaijie Tang
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Jiaxin Hong
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Zeng Chen
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Wuying Yang
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Limin Lu
- Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Ministry of Education, Key Laboratory of Chemical Utilization of Plant Resources of Nanchang, College of Chemistry and Materials, College of Science, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Lu-Ying Duan
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
9
|
Jia X, Wang Y, Qiao Y, Jiang X, Li J. Nanomaterial-based regulation of redox metabolism for enhancing cancer therapy. Chem Soc Rev 2024; 53:11590-11656. [PMID: 39431683 DOI: 10.1039/d4cs00404c] [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: 10/22/2024]
Abstract
Altered redox metabolism is one of the hallmarks of tumor cells, which not only contributes to tumor proliferation, metastasis, and immune evasion, but also has great relevance to therapeutic resistance. Therefore, regulation of redox metabolism of tumor cells has been proposed as an attractive therapeutic strategy to inhibit tumor growth and reverse therapeutic resistance. In this respect, nanomedicines have exhibited significant therapeutic advantages as intensively reported in recent studies. In this review, we would like to summarize the latest advances in nanomaterial-assisted strategies for redox metabolic regulation therapy, with a focus on the regulation of redox metabolism-related metabolite levels, enzyme activity, and signaling pathways. In the end, future expectations and challenges of such emerging strategies have been discussed, hoping to enlighten and promote their further development for meeting the various demands of advanced cancer therapies. It is highly expected that these therapeutic strategies based on redox metabolism regulation will play a more important role in the field of nanomedicine.
Collapse
Affiliation(s)
- Xiaodan Jia
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yue Wang
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yue Qiao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Xiue Jiang
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Jinghong Li
- Beijing Institute of Life Science and Technology, Beijing 102206, P. R. China
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P. R. China.
| |
Collapse
|
10
|
Cheng M, Chu AKY, Li Z, Yang S, Smith MD, Zhang Q, Brown NG, Marzluff WF, Bardeesy N, Milner JJ, Welch JD, Xiong Y, Baldwin AS. TET2 promotes tumor antigen presentation and T cell IFN-γ, which is enhanced by vitamin C. JCI Insight 2024; 9:e175098. [PMID: 39388288 PMCID: PMC11601905 DOI: 10.1172/jci.insight.175098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/08/2024] [Indexed: 10/12/2024] Open
Abstract
Immune evasion by tumors is promoted by low T cell infiltration, ineffective T cell activity directed against the tumor, and reduced tumor antigen presentation. The TET2 DNA dioxygenase gene is frequently mutated in hematopoietic malignancies and loss of TET enzymatic activity is found in a variety of solid tumors. We showed previously that vitamin C (VC), a cofactor of TET2, enhances tumor-associated T cell recruitment and checkpoint inhibitor therapy responses in a TET2-dependent manner. Using single-cell RNA sequencing (scRNA-seq) analysis performed on B16-OVA melanoma tumors, we have shown here that an additional function for TET2 in tumors is to promote expression of certain antigen presentation machinery genes, which is potently enhanced by VC. Consistently, VC promoted antigen presentation in cell-based and tumor assays in a TET2-dependent manner. Quantifying intercellular signaling from the scRNA-seq dataset showed that T cell-derived IFN-γ-induced signaling within the tumor and tumor microenvironment requires tumor-associated TET2 expression, which is enhanced by VC treatment. Analysis of patient tumor samples indicated that TET activity directly correlates with antigen presentation gene expression and with patient outcomes. Our results demonstrate the importance of tumor-associated TET2 activity as a critical mediator of tumor immunity, which is augmented by high-dose VC therapy.
Collapse
Affiliation(s)
- Meng Cheng
- Curriculum in Genetics and Molecular Biology, and
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Angel Ka Yan Chu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Zhijun Li
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Biochemistry and Biophysics, and
| | - Shiyue Yang
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Matthew D. Smith
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Qi Zhang
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Biochemistry and Biophysics, and
| | - Nicholas G. Brown
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - William F. Marzluff
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Biochemistry and Biophysics, and
| | - Nabeel Bardeesy
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - J. Justin Milner
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joshua D. Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
- Department of Computer Science and Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Yue Xiong
- Cullgen, Inc., San Diego, California, USA
| | - Albert S. Baldwin
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
11
|
Jahan I, Islam MA, Harun-Ur-Rashid M, Sultana GNN. Cancer prevention at the microscopic level with the potent power of micronutrients. Heliyon 2024; 10:e39680. [PMID: 39553634 PMCID: PMC11564030 DOI: 10.1016/j.heliyon.2024.e39680] [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: 06/10/2024] [Revised: 10/13/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
Cancer remains a leading cause of morbidity and mortality worldwide, necessitating ongoing exploration of effective prevention strategies. Micronutrients, vital for maintaining cellular health, offer promising avenues for cancer prevention. This review delineates the critical roles of micronutrients in cancer prevention, elucidating their mechanisms at the cellular level. Focusing on essential vitamins and minerals like Vitamins A, C, D, E, selenium, and zinc, we explore their profound effects on fundamental cellular processes such as DNA repair, oxidative stress regulation, cellular proliferation, and immune surveillance. These nutrients, characterized by their antioxidative, anti-inflammatory, and immune-enhancing properties, have shown potential in reducing the risk of cancer. The article synthesizes outcomes from a broad spectrum of clinical trials, epidemiological studies, and systematic reviews to evaluate the efficacy of micronutrients in thwarting cancer development. This critical analysis explores significant trials, addresses controversies in nutrient efficacy, and highlights the implications for clinical practice and public health policy. The review underscores the importance of integrating nutritional strategies into comprehensive cancer prevention frameworks and suggests directions for future research to optimize the preventive potentials of micronutrients.
Collapse
Affiliation(s)
- Israt Jahan
- Genetic Engineering and Biotechnology Research Laboratory (GEBRL), Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, 1000, Bangladesh
| | - Md Aminul Islam
- Genetic Engineering and Biotechnology Research Laboratory (GEBRL), Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, 1000, Bangladesh
| | - Mohammad Harun-Ur-Rashid
- Department of Chemistry, International University of Business Agriculture and Technology (IUBAT), Dhaka, 1230, Bangladesh
| | - Gazi Nurun Nahar Sultana
- Genetic Engineering and Biotechnology Research Laboratory (GEBRL), Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, 1000, Bangladesh
| |
Collapse
|
12
|
Fox J, Batchelor DVB, Coletta PL, Valleley EM, Evans SD. Microbubble Enhanced Delivery of Vitamin C for Treatment of Colorectal Cancer. ACS OMEGA 2024; 9:45270-45278. [PMID: 39554410 PMCID: PMC11561761 DOI: 10.1021/acsomega.4c06779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/20/2024] [Accepted: 10/23/2024] [Indexed: 11/19/2024]
Abstract
During chemotherapy treatment for cancer, often only a fraction of the administered dose reaches the tumor site, with the remaining drug spreading throughout the body, producing unwanted side-effects and restricting how much drug can be safely administered. A potential solution to reduce this problem is the use of microbubbles. The interaction between microbubbles and ultrasound generates pores in the tumor cells, permitting enhanced drug uptake. This study investigates the delivery of the ascorbic acid derivative, palmitoyl ascorbate, to KRAS-mutated colorectal cancer cells in vitro. Ultrasound-triggered microbubbles enhanced the efficacy of liposomal palmitoyl ascorbate treatments by 1.7- and 2.2-fold in LS174T and HCT116 CRC cell lines, respectively. This enhancement was achieved without increasing the drug dosage, and the therapeutic effect was shown to be localized to the area that received the ultrasound pulse, aiding in the reduction of off-site toxicity.
Collapse
Affiliation(s)
- Joseph Fox
- Molecular
and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, U.K.
| | - Damien V. B. Batchelor
- Molecular
and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, U.K.
| | - Patricia Louise Coletta
- Leeds
Institute of Medical Research, St James’s
University Hospital, Wellcome
Trust Brenner Building, Leeds LS9 7TF, U.K.
| | - Elizabeth M.A. Valleley
- Leeds
Institute of Medical Research, St James’s
University Hospital, Wellcome
Trust Brenner Building, Leeds LS9 7TF, U.K.
| | - Stephen D. Evans
- Molecular
and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, U.K.
| |
Collapse
|
13
|
Chen A, Huang H, Fang S, Hang Q. ROS: A "booster" for chronic inflammation and tumor metastasis. Biochim Biophys Acta Rev Cancer 2024; 1879:189175. [PMID: 39218404 DOI: 10.1016/j.bbcan.2024.189175] [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: 05/09/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Reactive oxygen species (ROS) are a group of highly active molecules produced by normal cellular metabolism and play a crucial role in the human body. In recent years, researchers have increasingly discovered that ROS plays a vital role in the progression of chronic inflammation and tumor metastasis. The inflammatory tumor microenvironment established by chronic inflammation can induce ROS production through inflammatory cells. ROS can then directly damage DNA or indirectly activate cellular signaling pathways to promote tumor metastasis and development, including breast cancer, lung cancer, liver cancer, colorectal cancer, and so on. This review aims to elucidate the relationship between ROS, chronic inflammation, and tumor metastasis, explaining how chronic inflammation can induce tumor metastasis and how ROS can contribute to the evolution of chronic inflammation toward tumor metastasis. Interestingly, ROS can have a "double-edged sword" effect, promoting tumor metastasis in some cases and inhibiting it in others. This article also highlights the potential applications of ROS in inhibiting tumor metastasis and enhancing the precision of tumor-targeted therapy. Combining ROS with nanomaterials strategies may be a promising approach to enhance the efficacy of tumor treatment.
Collapse
Affiliation(s)
- Anqi Chen
- Medical College, Yangzhou University, Yangzhou 225009, China
| | - Haifeng Huang
- Department of Laboratory Medicine, The First People's Hospital of Yancheng, Yancheng 224006, China; Department of Laboratory Medicine, Yancheng Clinical Medical College of Jiangsu University, Yancheng 224006, China
| | - Sumeng Fang
- School of Mathematics, Tianjin University, Tianjin 300350, China
| | - Qinglei Hang
- Jiangsu Provincial Innovation and Practice Base for Postdoctors, Suining People's Hospital, Affiliated Hospital of Xuzhou Medical University, Suining 221200, China; Key Laboratory of Jiangsu Province University for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou 225009, China; Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
14
|
Bodeker KL, Smith BJ, Berg DJ, Chandrasekharan C, Sharif S, Fei N, Vollstedt S, Brown H, Chandler M, Lorack A, McMichael S, Wulfekuhle J, Wagner BA, Buettner GR, Allen BG, Caster JM, Dion B, Kamgar M, Buatti JM, Cullen JJ. A randomized trial of pharmacological ascorbate, gemcitabine, and nab-paclitaxel for metastatic pancreatic cancer. Redox Biol 2024; 77:103375. [PMID: 39369582 PMCID: PMC11491967 DOI: 10.1016/j.redox.2024.103375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/26/2024] [Accepted: 09/28/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Patients with metastatic pancreatic ductal adenocarcinoma (PDAC) have poor 5-year survival. Pharmacological ascorbate (P-AscH-, high dose, intravenous, vitamin C) has shown promise as an adjunct to chemotherapy. We hypothesized adding P-AscH- to gemcitabine and nab-paclitaxel would increase survival in patients with metastatic PDAC. METHODS Patients diagnosed with stage IV pancreatic cancer randomized 1:1 to gemcitabine and nab-paclitaxel only (SOC, control) or to SOC with concomitant P-AscH-, 75 g three times weekly (ASC, investigational). The primary outcome was overall survival with secondary objectives of determining progression-free survival and adverse event incidence. Quality of life and patient reported outcomes for common oncologic symptoms were captured as an exploratory objective. Thirty-six participants were randomized; of this 34 received their assigned study treatment. All analyses were based on data frozen on December 11, 2023. RESULTS Intravenous P-AscH- increased serum ascorbate levels from micromolar to millimolar levels. P-AscH- added to the gemcitabine + nab-paclitaxel (ASC) increased overall survival to 16 months compared to 8.3 months with gemcitabine + nab-paclitaxel (SOC) (HR = 0.46; 90 % CI 0.23, 0.92; p = 0.030). Median progression free survival was 6.2 (ASC) vs. 3.9 months (SOC) (HR = 0.43; 90 % CI 0.20, 0.92; p = 0.029). Adding P-AscH- did not negatively impact quality of life or increase the frequency or severity of adverse events. CONCLUSIONS P-AscH- infusions of 75 g three times weekly in patients with metastatic pancreatic cancer prolongs overall and progression free survival without detriment to quality of life or added toxicity (ClinicalTrials.gov number NCT02905578).
Collapse
Affiliation(s)
- Kellie L Bodeker
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Brian J Smith
- College of Public Health, The University of Iowa, Iowa City, IA, USA
| | - Daniel J Berg
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Chandrikha Chandrasekharan
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Saima Sharif
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Naomi Fei
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Sandy Vollstedt
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Heather Brown
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Meghan Chandler
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Amanda Lorack
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Stacy McMichael
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Jared Wulfekuhle
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Brett A Wagner
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Garry R Buettner
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Bryan G Allen
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Joseph M Caster
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Barbara Dion
- Medical College of Wisconsin Division of Hematology and Oncology, Milwaukee, WI, USA
| | - Mandana Kamgar
- Medical College of Wisconsin Division of Hematology and Oncology, Milwaukee, WI, USA
| | - John M Buatti
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Joseph J Cullen
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA; Department of Surgery, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
15
|
Swamynathan MM, Kuang S, Watrud KE, Doherty MR, Gineste C, Mathew G, Gong GQ, Cox H, Cheng E, Reiss D, Kendall J, Ghosh D, Reczek CR, Zhao X, Herzka T, Špokaitė S, Dessus AN, Kim ST, Klingbeil O, Liu J, Nowak DG, Alsudani H, Wee TL, Park Y, Minicozzi F, Rivera K, Almeida AS, Chang K, Chakrabarty RP, Wilkinson JE, Gimotty PA, Diermeier SD, Egeblad M, Vakoc CR, Locasale JW, Chandel NS, Janowitz T, Hicks JB, Wigler M, Pappin DJ, Williams RL, Cifani P, Tuveson DA, Laporte J, Trotman LC. Dietary pro-oxidant therapy by a vitamin K precursor targets PI 3-kinase VPS34 function. Science 2024; 386:eadk9167. [PMID: 39446948 DOI: 10.1126/science.adk9167] [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: 09/20/2023] [Accepted: 08/27/2024] [Indexed: 10/26/2024]
Abstract
Men taking antioxidant vitamin E supplements have increased prostate cancer (PC) risk. However, whether pro-oxidants protect from PC remained unclear. In this work, we show that a pro-oxidant vitamin K precursor [menadione sodium bisulfite (MSB)] suppresses PC progression in mice, killing cells through an oxidative cell death: MSB antagonizes the essential class III phosphatidylinositol (PI) 3-kinase VPS34-the regulator of endosome identity and sorting-through oxidation of key cysteines, pointing to a redox checkpoint in sorting. Testing MSB in a myotubular myopathy model that is driven by loss of MTM1-the phosphatase antagonist of VPS34-we show that dietary MSB improved muscle histology and function and extended life span. These findings enhance our understanding of pro-oxidant selectivity and show how definition of the pathways they impinge on can give rise to unexpected therapeutic opportunities.
Collapse
Affiliation(s)
- Manojit M Swamynathan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Shan Kuang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Kaitlin E Watrud
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Mary R Doherty
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Charlotte Gineste
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, Inserm U1258, Strasbourg University, Illkirch CEDEX 67404, France
| | - Grinu Mathew
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Grace Q Gong
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Hilary Cox
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Eileen Cheng
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - David Reiss
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, Inserm U1258, Strasbourg University, Illkirch CEDEX 67404, France
| | - Jude Kendall
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Diya Ghosh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Colleen R Reczek
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Xiang Zhao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Tali Herzka
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Saulė Špokaitė
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | | | - Seung Tea Kim
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Olaf Klingbeil
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Juan Liu
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC 27695, USA
| | - Dawid G Nowak
- Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
- Division of Hematology and Medical Oncology, Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Habeeb Alsudani
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Tse-Luen Wee
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Youngkyu Park
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | | | - Keith Rivera
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Ana S Almeida
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
- APC Microbiome Ireland and School of Microbiology, University College Cork, Cork T12 K8AF, Ireland
| | - Kenneth Chang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Ram P Chakrabarty
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - John E Wilkinson
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Phyllis A Gimotty
- Perelman School of Medicine, Division of Biostatistics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah D Diermeier
- Department of Biochemistry, University of Otago, Dunedin 9016, New Zealand
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
- School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Jason W Locasale
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC 27695, USA
| | - Navdeep S Chandel
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Tobias Janowitz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - James B Hicks
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
- Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Michael Wigler
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Darryl J Pappin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | | | - Paolo Cifani
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, Inserm U1258, Strasbourg University, Illkirch CEDEX 67404, France
| | - Lloyd C Trotman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| |
Collapse
|
16
|
Gorini F, Tonacci A. Vitamin C in the Management of Thyroid Cancer: A Highway to New Treatment? Antioxidants (Basel) 2024; 13:1242. [PMID: 39456495 PMCID: PMC11505632 DOI: 10.3390/antiox13101242] [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: 09/12/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Thyroid cancer (TC) is the most common endocrine malignancy, with an increased global incidence in recent decades, despite a substantially unchanged survival. While TC has an excellent overall prognosis, some types of TC are associated with worse patient outcomes, depending on the genetic setting. Furthermore, oxidative stress is related to more aggressive features of TC. Vitamin C, an essential nutrient provided with food or as a dietary supplement, is a well-known antioxidant and a scavenger of reactive oxygen species; however, at high doses, it can induce pro-oxidant effects, acting through multiple biological mechanisms that play a crucial role in killing cancer cells. Although experimental data and, less consistently, clinical studies, suggest the possibility of antineoplastic effects of vitamin C at pharmacological doses, the antitumor efficacy of this nutrient in TC remains at least partly unexplored. Therefore, this review discusses the current state of knowledge on the role of vitamin C, alone or in combination with other conventional therapies, in the management of TC, the mechanisms underlying this association, and the perspectives that may emerge in TC treatment strategies, and, also, in light of the development of novel functional foods useful to this extent, by implementing novel sensory analysis strategies.
Collapse
Affiliation(s)
- Francesca Gorini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy;
| | | |
Collapse
|
17
|
Joshi KR, Devkota HP, Al-Mutairi KA, Sugimura K, Yahara S, Khadka R, Thapa S, Shekh MU, Poudel S, Watanabe T. Therapeutic potential of Leea asiatica: Chemical isolation and validation of ethnomedicinal claims through in vitro and in silico assessment of antioxidant and anti-inflammatory properties. Heliyon 2024; 10:e38074. [PMID: 39386820 PMCID: PMC11462247 DOI: 10.1016/j.heliyon.2024.e38074] [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: 05/08/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Leea asiatica (L.) Ridsdale has been used by different ethnic communities to manage diseased conditions that can be traced to oxidative stress and cellular inflammations but scientific evidences to support the claim are scanty. The objective of this study was to isolate and identify the antioxidants present in the aerial parts of Leea asiatica, perform their molecular docking against proteins to inspect whether the traditional uses of the plant can be validated by an in-silico approach. Quercetin (1), gallic acid (2), kaempferol (3), methyl gallate (4), myricetin 3-O-α-L-rhamnopyranoside (5), (-)-epicatechin-3-O-gallate (6) and (-)-epigallocatechin-3-O-gallate (7) were isolated from the 70 % methanolic extract of the aerial parts. Compounds 2, 4, 6, and 7 are reported for the first time from Leea asiatica. Quercetin (1), gallic acid (2), (-)-epicatechin-3-O-gallate (6) and (-)-epigallocatechin-3-O-gallate (7) showed potent antioxidant activity against 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical. Molecular docking with NADPH oxidase and TNF-α revealed that epicatechin-3-O-gallate, epigallocatechin-3-O-gallate and quercetin bound with the least binding energy amongst the isolated compounds as well as standard (Trolox and Prednisolone). By molecular dynamics analysis, epicatechin-3-O-gallate maintained stable conformation with NADPH oxidase and TNF-α and was found to possess good ADMET profile thereby validating the ethnic use of the plant as a medicine in the management of inflammatory conditions by an in vitro and in silico approach.
Collapse
Affiliation(s)
- Khem Raj Joshi
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- School of Health and Allied Sciences, Faculty of Health Sciences, Pokhara University, Pokhara, 33700, Nepal
| | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Khalid Awadh Al-Mutairi
- University of Tabuk, Department of Biology, Faculty of Science, Tabuk, P.O. Box 741, Tabuk, 741, Saudi Arabia
| | - Koji Sugimura
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Shoji Yahara
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Ravindra Khadka
- School of Health and Allied Sciences, Faculty of Health Sciences, Pokhara University, Pokhara, 33700, Nepal
| | - Shankar Thapa
- Department of Pharmacy, Madan Bhandari Academy of Health Sciences, Hetauda, Nepal
| | - Mohammad Ujair Shekh
- School of Health and Allied Sciences, Faculty of Health Sciences, Pokhara University, Pokhara, 33700, Nepal
| | - Sandesh Poudel
- School of Health and Allied Sciences, Faculty of Health Sciences, Pokhara University, Pokhara, 33700, Nepal
| | - Takashi Watanabe
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| |
Collapse
|
18
|
Burgess ER, Praditi C, Phillips E, Vissers MCM, Robinson BA, Dachs GU, Wiggins GAR. Role of Sodium-Dependent Vitamin C Transporter-2 and Ascorbate in Regulating the Hypoxic Pathway in Cultured Glioblastoma Cells. J Cell Biochem 2024:e30658. [PMID: 39382087 DOI: 10.1002/jcb.30658] [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: 05/10/2024] [Revised: 07/21/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024]
Abstract
The most common and aggressive brain cancer, glioblastoma, is characterized by hypoxia and poor survival. The pro-tumour transcription factor, hypoxia-inducible factor (HIF), is regulated via HIF-hydroxylases that require ascorbate as cofactor. Decreased HIF-hydroxylase activity triggers the hypoxic pathway driving cancer progression. Tissue ascorbate accumulates via the sodium-dependent vitamin C transporter-2 (SVCT2). We hypothesize that glioblastoma cells rely on SVCT2 for ascorbate accumulation, and that knockout of this transporter would disrupt the regulation of the hypoxic pathway by ascorbate. Ascorbate uptake was measured in glioblastoma cell lines (U87MG, U251MG, T98G) by high-performance liquid chromatography. CRISPR/Cas9 was used to knockout SVCT2. Cells were treated with cobalt chloride, desferrioxamine or 5% oxygen, with/without ascorbate, and key hypoxic pathway proteins were measured using Western blot analysis. Ascorbate uptake was cell line dependent, ranging from 1.7 to 11.0 nmol/106 cells. SVCT2-knockout cells accumulated 90%-95% less intracellular ascorbate than parental cells. The hypoxic pathway was induced by all three stimuli, and ascorbate reduced this induction. In the SVCT2-knockout cells, ascorbate had limited effect on the hypoxic pathway. This study verifies that intracellular ascorbate is required to suppress the hypoxic pathway. As patient survival is related to an activated hypoxic pathway, increasing intra-tumoral ascorbate may be of clinical interest.
Collapse
Affiliation(s)
- Eleanor R Burgess
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
- Department of Immunobiochemistry, Mannheim Institute for Innate Immunology (MI3), Heidelberg University, Medical Faculty Mannheim, Mannheim, Germany
| | - Citra Praditi
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
- Mātai Hāora, Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Elisabeth Phillips
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Margreet C M Vissers
- Mātai Hāora, Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Bridget A Robinson
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
- Canterbury Regional Cancer and Haematology Service, Te Whatu Ora, Waitaha/Canterbury, Christchurch, New Zealand
| | - Gabi U Dachs
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - George A R Wiggins
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| |
Collapse
|
19
|
Wang Y, Wang Q, Zhang G, Li Y, Guo H, Zhou J, Wang T, Jia H, Zhu L. Masks As a New Hotspot for Antibiotic Resistance Gene Spread: Reveal the Contribution of Atmospheric Pollutants and Potential Risks. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:16100-16111. [PMID: 39137285 DOI: 10.1021/acs.est.4c03399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
The consumption of disposable surgical masks (DSMs) considerably increased during the coronavirus pandemic in 2019. Herein, we explored the spread of antibiotic resistance genes (ARGs) and the potential risks of antibiotic resistant bacteria (ARB) on DSMs. At environmentally relevant concentrations, the conjugate transfer frequency (CTF) of ARGs increased by 1.34-2.37 folds by 20 μg/m3 of atmospheric water-soluble inorganic ions (WSIIs), and it increased by 2.62-2.86 folds by 80 ng/m3 of polycyclic aromatic hydrocarbons (PAHs). Total suspended particulates (TSP) further promoted the CTF in combination with WSIIs or PAHs. Under WSII and PAH exposure, gene expression levels related to oxidative stress, cell membrane, and the adenosine triphosphate (ATP) were upregulated. WSIIs predominantly induced cellular contact, while PAHs triggered ATP formation and membrane damage. Molecular dynamics simulations showed that WSIIs and PAHs reduced membrane lipid fluidity and increased membrane permeability through interactions with the phosphatidylcholine bilayer. DSM filtering performance decreased, and the CTF of ARGs increased with the wearing time. The gut simulator test showed that ARB disrupted the human gut microbial community and increased total ARG abundance but did not change the ARG abundance carried by ARB themselves. A mathematical model showed that long-term WSII and PAH exposure accelerated ARG dissemination in DSMs.
Collapse
Affiliation(s)
- Yanjie Wang
- College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi Province 712100, PR China
- Key Laboratory of Plant Nutrition and the Agri-environment in Northwest China, Ministry of Agriculture, Yangling Shaanxi 712100, PR China
| | - Qi Wang
- College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi Province 712100, PR China
- Key Laboratory of Plant Nutrition and the Agri-environment in Northwest China, Ministry of Agriculture, Yangling Shaanxi 712100, PR China
| | - Guodong Zhang
- College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi Province 712100, PR China
- Key Laboratory of Plant Nutrition and the Agri-environment in Northwest China, Ministry of Agriculture, Yangling Shaanxi 712100, PR China
| | - Yutong Li
- College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi Province 712100, PR China
- Key Laboratory of Plant Nutrition and the Agri-environment in Northwest China, Ministry of Agriculture, Yangling Shaanxi 712100, PR China
| | - He Guo
- College of Biology and the Environment, Nanjing Forestry University, Nanjing 210037, China
| | - Jian Zhou
- College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi Province 712100, PR China
- Key Laboratory of Plant Nutrition and the Agri-environment in Northwest China, Ministry of Agriculture, Yangling Shaanxi 712100, PR China
| | - Tiecheng Wang
- College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi Province 712100, PR China
- Key Laboratory of Plant Nutrition and the Agri-environment in Northwest China, Ministry of Agriculture, Yangling Shaanxi 712100, PR China
| | - Hanzhong Jia
- College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi Province 712100, PR China
- Key Laboratory of Plant Nutrition and the Agri-environment in Northwest China, Ministry of Agriculture, Yangling Shaanxi 712100, PR China
| | - Lingyan Zhu
- College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| |
Collapse
|
20
|
Piotrowsky A, Burkard M, Hammerschmidt K, Ruple HK, Nonnenmacher P, Schumacher M, Leischner C, Berchtold S, Marongiu L, Kufer TA, Lauer UM, Renner O, Venturelli S. Analysis of High-Dose Ascorbate-Induced Cytotoxicity in Human Glioblastoma Cells and the Role of Dehydroascorbic Acid and Iron. Antioxidants (Basel) 2024; 13:1095. [PMID: 39334754 PMCID: PMC11429401 DOI: 10.3390/antiox13091095] [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: 07/25/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Several studies have demonstrated, both in vitro and in animal models, the anti-tumor efficacy of high-dose ascorbate treatment against a variety of tumor entities, including glioblastoma, the most common and aggressive primary malignant brain tumor. The aim of this study was to investigate the effects of high-dose ascorbate as well as dehydroascorbic acid on human glioblastoma cell lines and to evaluate different treatment conditions for the combined administration of ascorbate with magnesium (Mg2+) and iron (Fe3+). Intracellular levels of reactive oxygen species and the induction of cell death following ascorbate treatment were also investigated. We demonstrated high cytotoxicity and antiproliferative efficacy of high-dose ascorbate in human glioblastoma cells, whereas much weaker effects were observed for dehydroascorbic acid. Ascorbate-induced cell death was independent of apoptosis. Both the reduction in cell viability and the ascorbate-induced generation of intracellular reactive oxygen species could be significantly increased by incubating the cells with Fe3+ before ascorbate treatment. This work demonstrates, for the first time, an increase in ascorbate-induced intracellular ROS formation and cytotoxicity in human glioblastoma cells by pre-treatment of the tumor cells with ferric iron, as well as caspase-3 independence of cell death induced by high-dose ascorbate. Instead, the cell death mechanism caused by high-dose ascorbate in glioblastoma cells shows evidence of ferroptosis. The results of the present work provide insights into the efficacy and mode of action of pharmacological ascorbate for the therapy of glioblastoma, as well as indications for possible approaches to increase the effectiveness of ascorbate treatment.
Collapse
Affiliation(s)
- Alban Piotrowsky
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Markus Burkard
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Katharina Hammerschmidt
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Hannah K. Ruple
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Pia Nonnenmacher
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Monika Schumacher
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Christian Leischner
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Susanne Berchtold
- Department of Medical Oncology and Pneumology, Virotherapy Center Tuebingen (VCT), Medical University Hospital, 72076 Tuebingen, Germany
| | - Luigi Marongiu
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart, Germany
- HoLMiR-Hohenheim Center for Livestock Microbiome Research, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Thomas A. Kufer
- Department of Immunology, Institute of Nutritional Medicine, University of Hohenheim, Fruwirthstrasse 12, 70593 Stuttgart, Germany
| | - Ulrich M. Lauer
- Department of Medical Oncology and Pneumology, Virotherapy Center Tuebingen (VCT), Medical University Hospital, 72076 Tuebingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tuebingen, a Partnership between DKFZ and University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Olga Renner
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart, Germany
- Faculty of Food and Nutrition Sciences, Hochschule Niederrhein, University of Applied Sciences, Rheydter Strasse 277, 41065 Moenchengladbach, Germany
| | - Sascha Venturelli
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart, Germany
- Department of Vegetative and Clinical Physiology, Institute of Physiology, University of Tuebingen, Wilhelmstrasse 56, 72074 Tuebingen, Germany
| |
Collapse
|
21
|
Chang SH, Chuang KC, Li ZY, Chang MC, Liu KT, Hsu CS, Huang SW, Chung MC, Wang SC, Chen YJ, Shieh JJ. The Protective Effects of Mcl-1 on Mitochondrial Damage and Oxidative Stress in Imiquimod-Induced Cancer Cell Death. Cancers (Basel) 2024; 16:3060. [PMID: 39272918 PMCID: PMC11394135 DOI: 10.3390/cancers16173060] [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: 08/07/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Mitochondria, vital organelles that generate ATP, determine cell fate. Dysfunctional and damaged mitochondria are fragmented and removed through mitophagy, a mitochondrial quality control mechanism. The FDA-approved drug IMQ, a synthetic agonist of Toll-like receptor 7, exhibits antitumor activity against various skin malignancies. We previously reported that IMQ promptly reduced the level of the antiapoptotic Mcl-1 protein and that Mcl-1 overexpression attenuated IMQ-triggered apoptosis in skin cancer cells. Furthermore, IMQ profoundly disrupted mitochondrial function, promoted mitochondrial fragmentation, induced mitophagy, and caused cell death by generating high levels of ROS. However, whether Mcl-1 protects mitochondria from IMQ treatment is still unknown. In this study, we demonstrated that Mcl-1 overexpression induced resistance to IMQ-induced apoptosis and reduced both IMQ-induced ROS generation and oxidative stress in cancer cells. Mcl-1 overexpression maintained mitochondrial function and integrity and prevented mitophagy in IMQ-treated cancer cells. Furthermore, IL-6 protected against IMQ-induced apoptosis by increasing Mcl-1 expression and attenuating IMQ-induced mitochondrial fragmentation. Mcl-1 overexpression ameliorates IMQ-induced ROS generation and mitochondrial fragmentation, thereby increasing mitochondrial stability and ultimately attenuating IMQ-induced cell death. Investigating the roles of Mcl-1 in mitochondria is a potential strategy for cancer therapy development.
Collapse
Affiliation(s)
- Shu-Hao Chang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
| | - Kai-Cheng Chuang
- Department of Life Sciences, National Chung Hsing University, Taichung 402202, Taiwan
| | - Zheng-Yi Li
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
| | - Mao-Chia Chang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
| | - Kuang-Ting Liu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Pathology & Laboratory Medicine, Taoyuan Armed Forces General Hospital, Taoyuan 325208, Taiwan
| | - Chien-Sheng Hsu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
- Frontier Molecular Medical Research Center in Children, Changhua Christian Children Hospital, Changhua 500209, Taiwan
| | - Shi-Wei Huang
- Center for Cell Therapy and Translation Research, China Medical University Hospital, Taichung 404327, Taiwan
| | - Mu-Chi Chung
- Division of Nephrology, Department of Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- PhD Program in Translational Medicine, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Biotechnology, Asia University, Taichung 413305, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402202, Taiwan
| | - Shih-Chung Wang
- Division of Pediatric Hematology/Oncology, Changhua Christian Children Hospital, Changhua 500209, Taiwan
| | - Yi-Ju Chen
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Dermatology, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Jeng-Jer Shieh
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Education and Research, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| |
Collapse
|
22
|
Vaishampayan P, Lee Y. Redox-active vitamin C suppresses human osteosarcoma growth by triggering intracellular ROS-iron-calcium signaling crosstalk and mitochondrial dysfunction. Redox Biol 2024; 75:103288. [PMID: 39083898 PMCID: PMC11342202 DOI: 10.1016/j.redox.2024.103288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024] Open
Abstract
Pharmacological vitamin C (VC) has gained attention for its pro-oxidant characteristics and selective ability to induce cancer cell death. However, defining its role in cancer has been challenging due to its complex redox properties. In this study, using a human osteosarcoma (OS) model, we show that the redox-active property of VC is critical for inducing non-apoptotic cancer cell death via intracellular reactive oxygen species (ROS)-iron-calcium crosstalk and mitochondrial dysfunction. In both 2D and 3D OS cell culture models, only the oxidizable form of VC demonstrated potent dose-dependent cytotoxicity, while non-oxidizable and oxidized VC derivatives had minimal effects. Live-cell imaging showed that only oxidizable VC caused a surge in cytotoxic ROS, dependent on iron rather than copper. Inhibitors of ferroptosis, a form of iron-dependent cell death, along with classical apoptosis inhibitors, were unable to completely counteract the cytotoxic effects induced by VC. Further pharmacological and genetic inhibition analyses showed that VC triggers calcium release through inositol 1,4,5-trisphosphate receptors (IP3Rs), leading to mitochondrial ROS production and eventual cell death. RNA sequencing revealed down-regulation of genes involved in the mitochondrial electron transport chain and oxidative phosphorylation upon pharmacological VC treatment. Consistently, high-dose VC reduced mitochondrial membrane potential, oxidative phosphorylation, and ATP levels, with ATP reconstitution rescuing VC-induced cytotoxicity. In vivo OS xenograft studies demonstrated reduced tumor growth with high-dose VC administration, concomitant with the altered expression of mitochondrial ATP synthase (MT-ATP). These findings emphasize VC's potential clinical utility in osteosarcoma treatment by inducing mitochondrial metabolic dysfunction through a vicious intracellular ROS-iron-calcium cycle.
Collapse
Affiliation(s)
- Prajakta Vaishampayan
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA
| | - Yool Lee
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA; Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA; Sleep and Performance Research Center, Washington State University, Spokane, WA, 99202, USA; Steve Gleason Institute for Neuroscience, Washington State University, Spokane, WA, 99202, USA.
| |
Collapse
|
23
|
Kishimoto S, Crooks DR, Yasunori O, Kota Y, Yamamoto K, Linehan WM, Levine M, Krishna MC, Brender JR. Pharmacologic ascorbate induces transient hypoxia sensitizing pancreatic ductal adenocarcinoma to a hypoxia activated prodrug. Free Radic Biol Med 2024; 222:579-587. [PMID: 38992394 DOI: 10.1016/j.freeradbiomed.2024.07.008] [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] [Received: 05/14/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
Hypoxic tumor microenvironments pose a significant challenge in cancer treatment. Hypoxia-activated prodrugs like evofosfamide aim to specifically target and eliminate these resistant cells. However, their effectiveness is often limited by reoxygenation after cell death. We hypothesized that ascorbate's pro-oxidant properties could be harnessed to induce transient hypoxia, enhancing the efficacy of evofosfamide by overcoming reoxygenation. To test this hypothesis, we investigated the sensitivity of MIA Paca-2 and A549 cancer cells to ascorbate in vitro and in vivo. Ascorbate induced a cytotoxic effect at 5 mM that could be alleviated by endogenous administration of catalase, suggesting a role for hydrogen peroxide in its cytotoxic mechanism. In vitro, Seahorse experiments indicated that the generation of hydrogen peroxide consumes oxygen, which is offset at later time points by a reduction in oxygen consumption due to hydrogen peroxide's cytotoxic effect. In vivo, photoacoustic imaging showed pharmacologic ascorbate treatment at sublethal levels triggered a complex, multi-phasic response in tumor oxygenation across both cell lines. Initially, ascorbate generated transient hypoxia within minutes through hydrogen peroxide production, via reactions that consume oxygen. This initial hypoxic phase peaked at around 150 s and then gradually subsided. However, at longer time scales (approximately 300 s) a vasodilation effect triggered by ascorbate resulted in increased blood flow and subsequent reoxygenation. Combining sublethal levels of i. p. Ascorbate with evofosfamide significantly prolonged tumor doubling time in MIA Paca-2 and A549 xenografts compared to either treatment alone. This improvement, however, was only observed in a subpopulation of tumors, highlighting the complexity of the oxygenation response.
Collapse
Affiliation(s)
- Shun Kishimoto
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel R Crooks
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Clinical Cancer Metabolism Facility, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Otowa Yasunori
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yamashita Kota
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kazutoshi Yamamoto
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - W Marston Linehan
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Clinical Cancer Metabolism Facility, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark Levine
- Molecular and Clinical Nutrition Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Murali C Krishna
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Jeffrey R Brender
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
24
|
Cenigaonandia‐Campillo A, Garcia‐Bautista A, Rio‐Vilariño A, Cebrian A, del Puerto L, Pellicer JA, Gabaldón JA, Pérez‐Sánchez H, Carmena‐Bargueño M, Meroño C, Traba J, Fernandez‐Aceñero MJ, Baños‐Herraiz N, Mozas‐Vivar L, Núñez‐Delicado E, Garcia‐Foncillas J, Aguilera Ó. Vitamin-C-dependent downregulation of the citrate metabolism pathway potentiates pancreatic ductal adenocarcinoma growth arrest. Mol Oncol 2024; 18:2212-2233. [PMID: 38425123 PMCID: PMC11467799 DOI: 10.1002/1878-0261.13616] [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: 07/31/2023] [Revised: 01/17/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
In pancreatic ductal adenocarcinoma (PDAC), metabolic rewiring and resistance to standard therapy are closely associated. PDAC cells show enormous requirements for glucose-derived citrate, the first rate-limiting metabolite in the synthesis of new lipids. Both the expression and activity of citrate synthase (CS) are extraordinarily upregulated in PDAC. However, no previous relationship between gemcitabine response and citrate metabolism has been documented in pancreatic cancer. Here, we report for the first time that pharmacological doses of vitamin C are capable of exerting an inhibitory action on the activity of CS, reducing glucose-derived citrate levels. Moreover, ascorbate targets citrate metabolism towards the de novo lipogenesis pathway, impairing fatty acid synthase (FASN) and ATP citrate lyase (ACLY) expression. Lowered citrate availability was found to be directly associated with diminished proliferation and, remarkably, enhanced gemcitabine response. Moreover, the deregulated citrate-derived lipogenic pathway correlated with a remarkable decrease in extracellular pH through inhibition of lactate dehydrogenase (LDH) and overall reduced glycolytic metabolism. Modulation of citric acid metabolism in highly chemoresistant pancreatic adenocarcinoma, through molecules such as vitamin C, could be considered as a future clinical option to improve patient response to standard chemotherapy regimens.
Collapse
Affiliation(s)
| | - Ana Garcia‐Bautista
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - Anxo Rio‐Vilariño
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - Arancha Cebrian
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - Laura del Puerto
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - José Antonio Pellicer
- Molecular Recognition and Encapsulation Research Group (REM), Health Sciences DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - José Antonio Gabaldón
- Molecular Recognition and Encapsulation Research Group (REM), Health Sciences DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - Horacio Pérez‐Sánchez
- Bioinformatics and High‐Performance Computing Research Group (BIO‐HPC), Computer Engineering DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - Miguel Carmena‐Bargueño
- Bioinformatics and High‐Performance Computing Research Group (BIO‐HPC), Computer Engineering DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - Carolina Meroño
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones CientíficasUniversidad Autónoma de Madrid (CSIC‐UAM)Spain
- Instituto Universitario de Biología Molecular‐UAM (IUBM‐UAM), Departamento de Biología MolecularUniversidad Autónoma de MadridSpain
| | - Javier Traba
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones CientíficasUniversidad Autónoma de Madrid (CSIC‐UAM)Spain
- Instituto Universitario de Biología Molecular‐UAM (IUBM‐UAM), Departamento de Biología MolecularUniversidad Autónoma de MadridSpain
| | | | | | - Lorena Mozas‐Vivar
- Preclinical programe START Madrid‐FJD Hospital fundación Jiménez DíazSpain
| | - Estrella Núñez‐Delicado
- Molecular Recognition and Encapsulation Research Group (REM), Health Sciences DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - Jesús Garcia‐Foncillas
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - Óscar Aguilera
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
- Universidad Católica de Murcia (UCAM)Spain
| |
Collapse
|
25
|
Yuan L, Huang Y, Chen X, Gao Y, Ma X, Wang Z, Hu Y, He J, Han C, Li J, Li Z, Weng X, Huang R, Cui Y, Li L, Hu W. Improving both performance and stability of n-type organic semiconductors by vitamin C. NATURE MATERIALS 2024; 23:1268-1275. [PMID: 38937585 DOI: 10.1038/s41563-024-01933-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 05/28/2024] [Indexed: 06/29/2024]
Abstract
Organic semiconductors (OSCs) are one of the most promising candidates for flexible, wearable and large-area electronics. However, the development of n-type OSCs has been severely held back due to the poor stability of their most candidates, that is, the intrinsically high reactivity of negatively charged polarons to oxygen and water. Here we demonstrate a general strategy based on vitamin C to stabilize n-type OSCs, remarkably improving the performance and stability of their device, for example, organic field-effect transistors. Vitamin C scavenges reactive oxygen species and inhibits their generation by sacrificial oxidation and non-sacrificial triplet quenching in a cascade process, which not only lastingly prevents molecular structure from oxidation damage but also passivates the latent electron traps to stabilize electron transport. This study presents a way to overcome the long-standing stability problem of n-type OSCs and devices.
Collapse
Affiliation(s)
- Liqian Yuan
- Key Laboratory of Organic Integrated Circuit, Ministry of Education & Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, Tianjin, China
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, China
| | - Yinan Huang
- Key Laboratory of Organic Integrated Circuit, Ministry of Education & Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, Tianjin, China
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, China
| | - Xiaosong Chen
- Key Laboratory of Organic Integrated Circuit, Ministry of Education & Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, Tianjin, China
| | - Yixuan Gao
- Institute of Molecular Plus, Tianjin University, Tianjin, China
| | - Xiaonan Ma
- Institute of Molecular Plus, Tianjin University, Tianjin, China
| | - Zhongwu Wang
- Key Laboratory of Organic Integrated Circuit, Ministry of Education & Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, Tianjin, China
| | - Yongxu Hu
- Key Laboratory of Organic Integrated Circuit, Ministry of Education & Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, Tianjin, China
| | - Jinbo He
- Key Laboratory of Organic Integrated Circuit, Ministry of Education & Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, Tianjin, China
| | - Cheng Han
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, China
| | - Jing Li
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Zhiyun Li
- Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Xuefei Weng
- Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Rong Huang
- Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Yi Cui
- Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Liqiang Li
- Key Laboratory of Organic Integrated Circuit, Ministry of Education & Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, Tianjin, China.
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, China.
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, China.
| | - Wenping Hu
- Key Laboratory of Organic Integrated Circuit, Ministry of Education & Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, Tianjin, China.
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, China.
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, China.
| |
Collapse
|
26
|
Hei Z, Yang S, Ouyang G, Hanna J, Lepoivre M, Huynh T, Aguinaga L, Cassinat B, Maslah N, Bourge M, Golinelli-Cohen MP, Guittet O, Vallières C, Vernis L, Fenaux P, Huang ME. Targeting the redox vulnerability of acute myeloid leukaemia cells with a combination of auranofin and vitamin C. Br J Haematol 2024; 205:1017-1030. [PMID: 39087522 DOI: 10.1111/bjh.19680] [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: 05/07/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
Acute myeloid leukaemia (AML) is a heterogeneous disease characterized by complex molecular and cytogenetic abnormalities. Pro-oxidant cellular redox status is a common hallmark of AML cells, providing a rationale for redox-based anticancer strategy. We previously discovered that auranofin (AUF), initially used for the treatment of rheumatoid arthritis and repositioned for its anticancer activity, can synergize with a pharmacological concentration of vitamin C (VC) against breast cancer cell line models. In this study, we observed that this drug combination synergistically and efficiently killed cells of leukaemic cell lines established from different myeloid subtypes. In addition to an induced elevation of reactive oxygen species and ATP depletion, a rapid dephosphorylation of 4E-BP1 and p70S6K, together with a strong inhibition of protein synthesis were early events in response to AUF/VC treatment, suggesting their implication in AUF/VC-induced cytotoxicity. Importantly, a study on 22 primary AML specimens from various AML subtypes showed that AUF/VC combinations at pharmacologically achievable concentrations were effective to eradicate primary leukaemic CD34+ cells from the majority of these samples, while being less toxic to normal cord blood CD34+ cells. Our findings indicate that targeting the redox vulnerability of AML with AUF/VC combinations could present a potential anti-AML therapeutic approach.
Collapse
Affiliation(s)
- Zhiliang Hei
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Shujun Yang
- Department of Hematology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Guifang Ouyang
- Department of Hematology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jolimar Hanna
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Michel Lepoivre
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Tony Huynh
- Service d'Hématologie Séniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Lorea Aguinaga
- Service d'Hématologie Séniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Bruno Cassinat
- INSERM UMR 1131, Université Paris Cité, Hôpital Saint-Louis, IRSL, Paris, France
| | - Nabih Maslah
- INSERM UMR 1131, Université Paris Cité, Hôpital Saint-Louis, IRSL, Paris, France
| | - Mickaël Bourge
- Cytometry Facility, Imagerie-Gif, Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | | | - Olivier Guittet
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Cindy Vallières
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Laurence Vernis
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Pierre Fenaux
- Service d'Hématologie Séniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Meng-Er Huang
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| |
Collapse
|
27
|
Park Y, Jeong EM. Glutathione Dynamics in the Tumor Microenvironment: A Potential Target of Cancer Stem Cells and T Cells. Int J Stem Cells 2024; 17:270-283. [PMID: 38919125 PMCID: PMC11361844 DOI: 10.15283/ijsc24060] [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: 05/10/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 06/27/2024] Open
Abstract
Glutathione (GSH), the main cellular antioxidant, dynamically influences tumor growth, metastasis, and resistance to therapy in the tumor microenvironment (TME), which comprises cancer cells, immune cells, stromal cells, and non-cellular components, including the extracellular matrix, metabolites, hypoxia, and acidity. Cancer stem cells (CSCs) and T cells are minor but significant cell subsets of the TME. GSH dynamics influences the fate of CSCs and T cells. Here, we explored GSH dynamics in CSCs and T cells within the TME, as well as therapeutic approaches that could target these dynamics.
Collapse
Affiliation(s)
- Youngjun Park
- Jeju Research Institute of Pharmaceutical Sciences, College of Pharmacy, Jeju National University, Jeju, Korea
| | - Eui Man Jeong
- Jeju Research Institute of Pharmaceutical Sciences, College of Pharmacy, Jeju National University, Jeju, Korea
| |
Collapse
|
28
|
Tian S, Chen M. The mechanisms and drug therapies of colorectal cancer and epigenetics: bibliometrics and visualized analysis. Front Pharmacol 2024; 15:1466156. [PMID: 39268463 PMCID: PMC11391208 DOI: 10.3389/fphar.2024.1466156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Background Numerous studies have demonstrated a link between epigenetics and CRC. However, there has been no systematic analysis or visualization of relevant publications using bibliometrics. Methods 839 publications obtained from the Web of Science Core (WoSCC) were systematically analyzed using CiteSpace and VOSviewer software. Results The results show that the countries, institutions, and authors with the most published articles are the United States, Harvard University, and Ogino and Shuji, respectively. SEPT9 is a blood test for the early detection of colorectal cancer. Vitamin D and gut microbiota mediate colorectal cancer and epigenetics, and probiotics may reduce colorectal cancer-related symptoms. We summarize the specific epigenetic mechanisms of CRC and the current existence and potential epigenetic drugs associated with these mechanisms. It is closely integrated with clinical practice, and the possible research directions and challenges in the future are proposed. Conclusion This study reviews the current research trends and hotspots in CRC and epigenetics, which can promote the development of this field and provide references for researchers in this field.
Collapse
Affiliation(s)
- Siyu Tian
- School of Clinical Medicine, Chengdu University of TCM, Chengdu, China
| | - Min Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
29
|
Liao S, Wang Q, Chen S, Huang Q, Zhou L, Liu H, He S, Zhou Z. Mito-LND and (E)-Akt inhibitor-IV: novel compounds inducing endoplasmic reticulum stress and ROS accumulation against hepatocellular carcinoma. J Transl Med 2024; 22:792. [PMID: 39198815 PMCID: PMC11351498 DOI: 10.1186/s12967-024-05545-5] [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: 01/17/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related mortality. Although multi-kinase inhibitors can prolong the overall survival of late-stage HCC patients, the emergence of drug resistance diminishes these benefits, ultimately resulting in treatment failure. Therefore, there is an urgent need for novel and effective drugs to impede the progression of liver cancer. METHODS This study employed a concentration gradient increment method to establish acquired sorafenib or regorafenib-resistant SNU-449 cells. Cell viability was assessed using the cell counting kit-8 assay. A library of 793 bioactive small molecules related to metabolism screened compounds targeting both parental and drug-resistant cells. The screened compounds will be added to both the HCC parental cells and the drug-resistant cells, followed by a comprehensive assessment. Intracellular adenosine triphosphate (ATP) levels were quantified using kits. Flow cytometry was applied to assess cell apoptosis and reactive oxygen species (ROS). Real-time quantitative PCR studied relative gene expression, and western blot analysis assessed protein expression changes in HCC parental and drug-resistant cells. A xenograft model in vivo evaluated Mito-LND and (E)-Akt inhibitor-IV effects on liver tumors, with hematoxylin and eosin staining for tissue structure and immunohistochemistry staining for endoplasmic reticulum stress protein expression. RESULTS From the compound library, we screened out two novel compounds, Mito-LND and (E)-Akt inhibitor-IV, which could potently kill both parental cells and drug-resistant cells. Mito-LND could significantly suppress proliferation and induce apoptosis in HCC parental and drug-resistant cells by upregulating glycolytic intermediates and downregulating those of the tricarboxylic acid (TCA) cycle, thereby decreasing ATP production and increasing ROS. (E)-Akt inhibitor-IV achieved comparable results by reducing glycolytic intermediates, increasing TCA cycle intermediates, and decreasing ATP synthesis and ROS levels. Both compounds trigger apoptosis in HCC cells through the interplay of the AMPK/MAPK pathway and the endoplasmic reticulum stress response. In vivo assays also showed that these two compounds could significantly inhibit the growth of HCC cells and induce endoplasmic reticulum stress. CONCLUSION Through high throughput screening, we identified that Mito-LND and (E)-Akt inhibitor-IV are two novel compounds against both parental and drug-resistant HCC cells, which could offer new strategies for HCC patients.
Collapse
Affiliation(s)
- Siqi Liao
- The Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingliang Wang
- The Department of Pathology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Siyuan Chen
- The Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qixuan Huang
- The Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Zhou
- The Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongtao Liu
- The Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Song He
- The Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Zhihang Zhou
- The Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
30
|
Vernieri C, Ligorio F, Tripathy D, Longo VD. Cyclic fasting-mimicking diet in cancer treatment: Preclinical and clinical evidence. Cell Metab 2024; 36:1644-1667. [PMID: 39059383 DOI: 10.1016/j.cmet.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/03/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024]
Abstract
In preclinical tumor models, cyclic fasting and fasting-mimicking diets (FMDs) produce antitumor effects that become synergistic when combined with a wide range of standard anticancer treatments while protecting normal tissues from treatment-induced adverse events. More recently, results of phase 1/2 clinical trials showed that cyclic FMD is safe, feasible, and associated with positive metabolic and immunomodulatory effects in patients with different tumor types, thus paving the way for larger clinical trials to investigate FMD anticancer activity in different clinical contexts. Here, we review the tumor-cell-autonomous and immune-system-mediated mechanisms of fasting/FMD antitumor effects, and we critically discuss new metabolic interventions that could synergize with nutrient starvation to boost its anticancer activity and prevent or reverse tumor resistance while minimizing toxicity to patients. Finally, we highlight potential future applications of FMD approaches in combination with standard anticancer strategies as well as strategies to implement the design and conduction of clinical trials.
Collapse
Affiliation(s)
- Claudio Vernieri
- Medical Oncology and Hematology-Oncology Department, University of Milan, 20122 Milan, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, 20139 Milan, Italy.
| | - Francesca Ligorio
- Medical Oncology and Hematology-Oncology Department, University of Milan, 20122 Milan, Italy; Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Unit 1354, 1515 Holcombe Blvd, Houston, TX 77030-4009, USA
| | - Valter D Longo
- IFOM ETS, the AIRC Institute of Molecular Oncology, 20139 Milan, Italy; Longevity Institute, Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
31
|
Lan T, Arastu S, Lam J, Kim H, Wang W, Wang S, Bhatt V, Lopes EC, Hu Z, Sun M, Luo X, Ghergurovich JM, Su X, Rabinowitz JD, White E, Guo JY. Glucose-6-phosphate dehydrogenase maintains redox homeostasis and biosynthesis in LKB1-deficient KRAS-driven lung cancer. Nat Commun 2024; 15:5857. [PMID: 38997257 PMCID: PMC11245543 DOI: 10.1038/s41467-024-50157-8] [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: 10/04/2023] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
Cancer cells depend on nicotinamide adenine dinucleotide phosphate (NADPH) to combat oxidative stress and support reductive biosynthesis. One major NADPH production route is the oxidative pentose phosphate pathway (committed step: glucose-6-phosphate dehydrogenase, G6PD). Alternatives exist and can compensate in some tumors. Here, using genetically-engineered lung cancer mouse models, we show that G6PD ablation significantly suppresses KrasG12D/+;Lkb1-/- (KL) but not KrasG12D/+;P53-/- (KP) lung tumorigenesis. In vivo isotope tracing and metabolomics reveal that G6PD ablation significantly impairs NADPH generation, redox balance, and de novo lipogenesis in KL but not KP lung tumors. Mechanistically, in KL tumors, G6PD ablation activates p53, suppressing tumor growth. As tumors progress, G6PD-deficient KL tumors increase an alternative NADPH source from serine-driven one carbon metabolism, rendering associated tumor-derived cell lines sensitive to serine/glycine depletion. Thus, oncogenic driver mutations determine lung cancer dependence on G6PD, whose targeting is a potential therapeutic strategy for tumors harboring KRAS and LKB1 co-mutations.
Collapse
Affiliation(s)
- Taijin Lan
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | - Sara Arastu
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | - Jarrick Lam
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | - Hyungsin Kim
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | - Wenping Wang
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | - Samuel Wang
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | | | - Eduardo Cararo Lopes
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, 08854, USA
| | - Zhixian Hu
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | - Michael Sun
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | - Xuefei Luo
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | | | - Xiaoyang Su
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Joshua D Rabinowitz
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
- Department of Chemistry, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA
| | - Eileen White
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, 08854, USA
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, 08544, USA
| | - Jessie Yanxiang Guo
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA.
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
- Department of Chemical Biology, Rutgers Ernest Mario School of Pharmacy, Piscataway, NJ, 08854, USA.
| |
Collapse
|
32
|
Wang Y, He J, Lian S, Zeng Y, He S, Xu J, Luo L, Yang W, Jiang J. Targeting Metabolic-Redox Nexus to Regulate Drug Resistance: From Mechanism to Tumor Therapy. Antioxidants (Basel) 2024; 13:828. [PMID: 39061897 PMCID: PMC11273443 DOI: 10.3390/antiox13070828] [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: 05/22/2024] [Revised: 06/29/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Drug resistance is currently one of the biggest challenges in cancer treatment. With the deepening understanding of drug resistance, various mechanisms have been revealed, including metabolic reprogramming and alterations of redox balance. Notably, metabolic reprogramming mediates the survival of tumor cells in harsh environments, thereby promoting the development of drug resistance. In addition, the changes during metabolic pattern shift trigger reactive oxygen species (ROS) production, which in turn regulates cellular metabolism, DNA repair, cell death, and drug metabolism in direct or indirect ways to influence the sensitivity of tumors to therapies. Therefore, the intersection of metabolism and ROS profoundly affects tumor drug resistance, and clarifying the entangled mechanisms may be beneficial for developing drugs and treatment methods to thwart drug resistance. In this review, we will summarize the regulatory mechanism of redox and metabolism on tumor drug resistance and highlight recent therapeutic strategies targeting metabolic-redox circuits, including dietary interventions, novel chemosynthetic drugs, drug combination regimens, and novel drug delivery systems.
Collapse
Affiliation(s)
- Yuke Wang
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.H.); (S.L.); (Y.Z.); (S.H.); (J.X.)
| | - Jingqiu He
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.H.); (S.L.); (Y.Z.); (S.H.); (J.X.)
| | - Shan Lian
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.H.); (S.L.); (Y.Z.); (S.H.); (J.X.)
| | - Yan Zeng
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.H.); (S.L.); (Y.Z.); (S.H.); (J.X.)
| | - Sheng He
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.H.); (S.L.); (Y.Z.); (S.H.); (J.X.)
| | - Jue Xu
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.H.); (S.L.); (Y.Z.); (S.H.); (J.X.)
| | - Li Luo
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China;
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Wenyong Yang
- Department of Neurosurgery, Medical Research Center, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chong-Qing Medical University, Chengdu 610041, China
| | - Jingwen Jiang
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.H.); (S.L.); (Y.Z.); (S.H.); (J.X.)
| |
Collapse
|
33
|
Kabelitz D, Cierna L, Juraske C, Zarobkiewicz M, Schamel WW, Peters C. Empowering γδ T-cell functionality with vitamin C. Eur J Immunol 2024; 54:e2451028. [PMID: 38616772 DOI: 10.1002/eji.202451028] [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: 01/23/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
Vitamin C (ascorbic acid) is a potent antioxidant and a cofactor for various enzymes including histone demethylases and methylcytosine dioxygenases. Vitamin C also exerts direct cytotoxicity toward selected tumor cells including colorectal carcinoma. Moreover, vitamin C has been shown to impact immune cell differentiation at various levels including maturation and/or functionality of T cells and their progenitors, dendritic cells, B cells, and NK cells. γδ T cells have recently attracted great interest as effector cells for cell-based cancer immunotherapy, due to their HLA-independent recognition of a large variety of tumor cells. While γδ T cells can thus be also applied as an allogeneic off-the-shelf product, it is obvious that the effector function of γδ T cells needs to be optimized to ensure the best possible clinical efficacy. Here we review the immunomodulatory mechanisms of vitamin C with a special focus on how vitamin C enhances the effector function of γδ T cells. We also discuss future directions of how vitamin C can be used in the clinical setting to boost the efficacy of adoptive cell therapies.
Collapse
Affiliation(s)
- Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts University and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
- Institute of Immunology, UKSH Campus Kiel, Kiel, Germany
| | - Lea Cierna
- Institute of Immunology, Christian-Albrechts University and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Claudia Juraske
- Signalling Research Centres BIOSS and CIBSS, and Faculty of Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Michal Zarobkiewicz
- Institute of Immunology, Christian-Albrechts University and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Wolfgang W Schamel
- Signalling Research Centres BIOSS and CIBSS, and Faculty of Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Centre Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Peters
- Institute of Immunology, Christian-Albrechts University and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| |
Collapse
|
34
|
He MM, Wang K, Lo CH, Zhang Y, Polychronidis G, Knudsen MD, Zhong R, Ma Y, Wu K, Chan AT, Giovannucci EL, Ogino S, Ng K, Meyerhardt JA, Song M. Post-diagnostic multivitamin supplement use and colorectal cancer survival: A prospective cohort study. Cancer 2024; 130:2169-2179. [PMID: 38319287 PMCID: PMC11141725 DOI: 10.1002/cncr.35234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 12/01/2023] [Accepted: 01/04/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND Use of multivitamin supplements has been associated with lower incidence of colorectal cancer (CRC). However, its influence on CRC survival remains unknown. METHODS Among 2424 patients with stage I-III CRC who provided detailed information about multivitamin supplements in the Nurses' Health Study and Health Professionals Follow-up Study, the authors calculated multivariable hazard ratios (HRs) of multivitamin supplements for all-cause and CRC-specific mortality according to post-diagnostic use and dose of multivitamin supplements. RESULTS During a median follow-up of 11 years, the authors documented 1512 deaths, among which 343 were of CRC. Compared to non-users, post-diagnostic users of multivitamin supplements at a dose of 3-5 tablets/week had lower CRC-specific mortality (HR, 0.55; 95% confidence interval [CI], 0.36-0.83, p = .005), and post-diagnostic users at doses of 3-5 and 6-9 tablets/week had lower all-cause mortality (HR, 0.81; 95% CI, 0.67-0.99, p = .04; HR, 0.79; 95% CI, 0.70-0.88), p < .001). The dose-response analysis showed a curvilinear relationship for both CRC-specific (pnonlinearity < .001) and all-cause mortality (pnonlinearity = .004), with the maximum risk reduction observed at 3-5 tablets/week and no further reduction at higher doses. Compared to non-users in both pre- and post-diagnosis periods, new post-diagnostic users at dose of <10 tablets/week had a lower all-cause mortality (HR, 0.81; 95% CI, 0.71-0.94, p = .005), whereas new users at a dose of ≥10 tablets/week (HR, 1.58; 95% CI, 1.07-2.33) and discontinued users (HR, 1.35; 95% CI, 1.14-1.59) had a higher risk of mortality. CONCLUSIONS Use of multivitamin supplements at a moderate dose after a diagnosis of nonmetastatic CRC is associated with lower CRC-specific and overall mortality, whereas a high dose (≥10 tablets/week) use is associated with higher CRC-specific mortality.
Collapse
Affiliation(s)
- Ming-ming He
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kai Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Chun-Han Lo
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Yiwen Zhang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Georgios Polychronidis
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
- Study Centre of the German Surgical Society, University of Heidelberg, Heidelberg, Germany
| | - Markus D Knudsen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Section for Colorectal Cancer Screening, Cancer Registry of Norway, Oslo, Norway
- Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Norwegian PSC Research Center, Oslo University Hospital, Oslo, Norway
| | - Rong Zhong
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yuan Ma
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andrew T. Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Edward L. Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Shuji Ogino
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Jeffrey A. Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Mingyang Song
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
35
|
Ye J, Bao X, Wei J, Zhang Y, Liu Y, Xin L. Role of dietary nutrients and metabolism in colorectal cancer. Asia Pac J Clin Nutr 2024; 33:153-161. [PMID: 38794975 PMCID: PMC11170022 DOI: 10.6133/apjcn.202406_33(2).0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/05/2024] [Accepted: 01/23/2024] [Indexed: 05/27/2024]
Abstract
Colorectal cancer (CRC) is one of the most common malignancies and the leading causes of cancer related deaths worldwide. The development of CRC is driven by a combination of genetic and environmental factors. There is growing evidence that changes in dietary nutrition may modulate the CRC risk, and protective effects on the risk of developing CRC have been advocated for specific nutrients such as glucose, amino acids, lipid, vitamins, micronutrients and prebiotics. Metabolic crosstalk between tumor cells, tumor microenvironment components and intestinal flora further promote proliferation, invasion and metastasis of CRC cells and leads to treatment resistance. This review summarizes the research progress on CRC prevention, pathogenesis, and treatment by dietary supplementation or deficiency of glucose, amino acids, lipids, vitamins, micronutri-ents, and prebiotics, respectively. The roles played by different nutrients and dietary crosstalk in the tumor microenvironment and metabolism are discussed, and nutritional modulation is inspired to be beneficial in the prevention and treatment of CRC.
Collapse
Affiliation(s)
- Jinjun Ye
- Department of General Surgery, Longgang Central Hospital of Shenzhen, Longgang District, Shenzhen, Guangdong, China
| | - Xing Bao
- Department of General Surgery, Longgang Central Hospital of Shenzhen, Longgang District, Shenzhen, Guangdong, China
| | - Jiufeng Wei
- Department of General Surgery, Longgang Central Hospital of Shenzhen, Longgang District, Shenzhen, Guangdong, China
| | - Yuanpeng Zhang
- Department of General Surgery, Longgang Central Hospital of Shenzhen, Longgang District, Shenzhen, Guangdong, China
| | - Yu Liu
- Department of General Surgery, Longgang Central Hospital of Shenzhen, Longgang District, Shenzhen, Guangdong, China
| | - Le Xin
- Department of General Surgery, Longgang Central Hospital of Shenzhen, Longgang District, Shenzhen, Guangdong, China.
| |
Collapse
|
36
|
Chen XR, Zhou T, Zhou ZD, Fang ZH, Wang KB, Zhang C, Kong LY, Yang MH. The discovery of an anti-Candida xanthone with selective inhibition of Candida albicans GAPDH. Int J Antimicrob Agents 2024; 63:107172. [PMID: 38608845 DOI: 10.1016/j.ijantimicag.2024.107172] [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: 10/11/2023] [Revised: 03/26/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024]
Abstract
OBJECTIVES This study aimed to discover novel antifungals targeting Candida albicans glyceraldehyde-3-phosphate dehydrogenase (CaGAPDH), have an insight into inhibitory mode, and provide evidence supporting CaGAPDH as a target for new antifungals. METHODS Virtual screening was utilized to discover inhibitors of CaGAPDH. The inhibitory effect on cellular GAPDH was evaluated by determining the levels of ATP, NAD, NADH, etc., as well as examining GAPDH mRNA and protein expression. The role of GAPDH inhibition in C. albicans was supported by drug affinity responsive target stability and overexpression experiments. The mechanism of CaGAPDH inhibition was elucidated by Michaelis-Menten enzyme kinetics and site-specific mutagenesis based on docking. Chemical synthesis was used to produce an improved candidate. Different sources of GAPDH were used to evaluate inhibitory selectivity across species. In vitro and in vivo antifungal tests, along with anti-biofilm activity, were carried out to evaluate antifungal potential of GAPDH inhibitors. RESULTS A natural xanthone was identified as the first competitive inhibitor of CaGAPDH. It demonstrated in vitro anti-C. albicans potential but also caused hemolysis. XP-W, a synthetic side-chain-optimized xanthone, demonstrated a better safety profile, exhibiting a 50-fold selectivity for CaGAPDH over human GAPDH. XP-W also exhibited potent anti-biofilm activity and displayed broad-spectrum anti-Candida activities in vitro and in vivo, including multi-azole-resistant C. albicans. CONCLUSIONS These results demonstrate for the first time that CaGAPDH is a valuable target for antifungal drug discovery, and XP-W provides a promising lead.
Collapse
Affiliation(s)
- Xing-Ru Chen
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Tao Zhou
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Zhuo-Da Zhou
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Zhan-Hong Fang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Kai-Bo Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Chao Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Ling-Yi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China.
| | - Ming-Hua Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
37
|
Guo D, Liao Y, Na J, Wu L, Yin Y, Mi Z, Fang S, Liu X, Huang Y. The Involvement of Ascorbic Acid in Cancer Treatment. Molecules 2024; 29:2295. [PMID: 38792156 PMCID: PMC11123810 DOI: 10.3390/molecules29102295] [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: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Vitamin C (VC), also known as ascorbic acid, plays a crucial role as a water-soluble nutrient within the human body, contributing to a variety of metabolic processes. Research findings suggest that increased doses of VC demonstrate potential anti-tumor capabilities. This review delves into the mechanisms of VC absorption and its implications for cancer management. Building upon these foundational insights, we explore modern delivery systems for VC, evaluating its use in diverse cancer treatment methods. These include starvation therapy, chemodynamic therapy (CDT), photothermal/photodynamic therapy (PTT/PDT), electrothermal therapy, immunotherapy, cellular reprogramming, chemotherapy, radiotherapy, and various combination therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China; (D.G.); (Y.L.); (J.N.); (L.W.); (Y.Y.); (Z.M.); (S.F.)
| | - Yong Huang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China; (D.G.); (Y.L.); (J.N.); (L.W.); (Y.Y.); (Z.M.); (S.F.)
| |
Collapse
|
38
|
Jomova K, Alomar SY, Alwasel SH, Nepovimova E, Kuca K, Valko M. Several lines of antioxidant defense against oxidative stress: antioxidant enzymes, nanomaterials with multiple enzyme-mimicking activities, and low-molecular-weight antioxidants. Arch Toxicol 2024; 98:1323-1367. [PMID: 38483584 PMCID: PMC11303474 DOI: 10.1007/s00204-024-03696-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 01/31/2024] [Indexed: 03/27/2024]
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are well recognized for playing a dual role, since they can be either deleterious or beneficial to biological systems. An imbalance between ROS production and elimination is termed oxidative stress, a critical factor and common denominator of many chronic diseases such as cancer, cardiovascular diseases, metabolic diseases, neurological disorders (Alzheimer's and Parkinson's diseases), and other disorders. To counteract the harmful effects of ROS, organisms have evolved a complex, three-line antioxidant defense system. The first-line defense mechanism is the most efficient and involves antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx). This line of defense plays an irreplaceable role in the dismutation of superoxide radicals (O2•-) and hydrogen peroxide (H2O2). The removal of superoxide radicals by SOD prevents the formation of the much more damaging peroxynitrite ONOO- (O2•- + NO• → ONOO-) and maintains the physiologically relevant level of nitric oxide (NO•), an important molecule in neurotransmission, inflammation, and vasodilation. The second-line antioxidant defense pathway involves exogenous diet-derived small-molecule antioxidants. The third-line antioxidant defense is ensured by the repair or removal of oxidized proteins and other biomolecules by a variety of enzyme systems. This review briefly discusses the endogenous (mitochondria, NADPH, xanthine oxidase (XO), Fenton reaction) and exogenous (e.g., smoking, radiation, drugs, pollution) sources of ROS (superoxide radical, hydrogen peroxide, hydroxyl radical, peroxyl radical, hypochlorous acid, peroxynitrite). Attention has been given to the first-line antioxidant defense system provided by SOD, CAT, and GPx. The chemical and molecular mechanisms of antioxidant enzymes, enzyme-related diseases (cancer, cardiovascular, lung, metabolic, and neurological diseases), and the role of enzymes (e.g., GPx4) in cellular processes such as ferroptosis are discussed. Potential therapeutic applications of enzyme mimics and recent progress in metal-based (copper, iron, cobalt, molybdenum, cerium) and nonmetal (carbon)-based nanomaterials with enzyme-like activities (nanozymes) are also discussed. Moreover, attention has been given to the mechanisms of action of low-molecular-weight antioxidants (vitamin C (ascorbate), vitamin E (alpha-tocopherol), carotenoids (e.g., β-carotene, lycopene, lutein), flavonoids (e.g., quercetin, anthocyanins, epicatechin), and glutathione (GSH)), the activation of transcription factors such as Nrf2, and the protection against chronic diseases. Given that there is a discrepancy between preclinical and clinical studies, approaches that may result in greater pharmacological and clinical success of low-molecular-weight antioxidant therapies are also subject to discussion.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine The Philosopher University in Nitra, Nitra, 949 74, Slovakia
| | - Suliman Y Alomar
- Doping Research Chair, Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Saleh H Alwasel
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia.
| |
Collapse
|
39
|
Yang N, Li J, Yu S, Xia G, Li D, Yuan L, Wang Q, Ding L, Fan Z, Li J. Application of Nanomaterial-Based Sonodynamic Therapy in Tumor Therapy. Pharmaceutics 2024; 16:603. [PMID: 38794265 PMCID: PMC11125068 DOI: 10.3390/pharmaceutics16050603] [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: 03/28/2024] [Revised: 04/19/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Sonodynamic therapy (SDT) has attracted significant attention in recent years as it is an innovative approach to tumor treatment. It involves the utilization of sound waves or ultrasound (US) to activate acoustic sensitizers, enabling targeted drug release for precise tumor treatment. This review aims to provide a comprehensive overview of SDT, encompassing its underlying principles and therapeutic mechanisms, the applications of nanomaterials, and potential synergies with combination therapies. The review begins by introducing the fundamental principle of SDT and delving into the intricate mechanisms through which it facilitates tumor treatment. A detailed analysis is presented, outlining how SDT effectively destroys tumor cells by modulating drug release mechanisms. Subsequently, this review explores the diverse range of nanomaterials utilized in SDT applications and highlights their specific contributions to enhancing treatment outcomes. Furthermore, the potential to combine SDT with other therapeutic modalities such as photothermal therapy (PTT) and chemotherapy is discussed. These combined approaches aim to synergistically improve therapeutic efficacy while mitigating side effects. In conclusion, SDT emerges as a promising frontier in tumor treatment that offers personalized and effective treatment options with the potential to revolutionize patient care. As research progresses, SDT is poised to play a pivotal role in shaping the future landscape of oncology by providing patients with a broader spectrum of efficacious and tailored treatment options.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhongxiong Fan
- School of Pharmaceutical Sciences, Institute of Materia Medica, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Jinyao Li
- School of Pharmaceutical Sciences, Institute of Materia Medica, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| |
Collapse
|
40
|
Zhang L, Li G, Lin B, He H, Zhou R, Jiang W. Ascorbyl palmitate ameliorates inflammatory diseases by inhibition of NLRP3 inflammasome. Int Immunopharmacol 2024; 131:111915. [PMID: 38522141 DOI: 10.1016/j.intimp.2024.111915] [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: 01/25/2024] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
The aberrant activation of NLRP3 inflammasome contributes to pathogenesis of multiple inflammation-driven human diseases. However, the medications targeting NLRP3 inflammasome are not approved for clinic use to date. Here, we show that ascorbyl palmitate (AP), a lipophilic derivative of ascorbic acid (AA) and a safe food additive, is a potent inhibitor of NLRP3 inflammasome. Compared with AA, AP inhibited the activation of NLRP3 inflammasome with increased potency and specificity. Mechanistically, AP directly scavenged mitochondrial reactive oxygen species (mitoROS) by its antioxidant activity and blocked NLRP3-NEK7 interaction and NLRP3 inflammasome assembly. Moreover, AP showed more significant preventive effects than AA in LPS-induced systemic inflammation, dextran sulfate sodium (DSS)-induced colitis and experimental autoimmune encephalomyelitis (EAE). Thus, our results suggest that AP is a potential therapeutic combating NLRP3-driven diseases.
Collapse
Affiliation(s)
- Luchen Zhang
- Department of Geriatrics, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China, Hefei, China; Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Guoyang Li
- Department of Geriatrics, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China, Hefei, China; Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bolong Lin
- Department of Geriatrics, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China, Hefei, China; Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongbin He
- Department of Geriatrics, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China, Hefei, China; Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Rongbin Zhou
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Wei Jiang
- Department of Geriatrics, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China, Hefei, China; Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
41
|
Mangione R, Giallongo C, Duminuco A, La Spina E, Longhitano L, Giallongo S, Tibullo D, Lazzarino G, Saab MW, Sbriglione A, Palumbo GA, Graziani A, Alanazi AM, Di Pietro V, Tavazzi B, Amorini AM, Lazzarino G. Targeted Metabolomics Highlights Dramatic Antioxidant Depletion, Increased Oxidative/Nitrosative Stress and Altered Purine and Pyrimidine Concentrations in Serum of Primary Myelofibrosis Patients. Antioxidants (Basel) 2024; 13:490. [PMID: 38671937 PMCID: PMC11047794 DOI: 10.3390/antiox13040490] [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: 03/03/2024] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
To date, little is known concerning the circulating levels of biochemically relevant metabolites (antioxidants, oxidative/nitrosative stress biomarkers, purines, and pyrimidines) in patients with primary myelofibrosis (PMF), a rare form of myeloproliferative tumor causing a dramatic decrease in erythropoiesis and angiogenesis. In this study, using a targeted metabolomic approach, serum samples of 22 PMF patients and of 22 control healthy donors were analyzed to quantify the circulating concentrations of hypoxanthine, xanthine, uric acid (as representative purines), uracil, β-pseudouridine, uridine (as representative pyrimidines), reduced glutathione (GSH), ascorbic acid (as two of the main water-soluble antioxidants), malondialdehyde, nitrite, nitrate (as oxidative/nitrosative stress biomarkers) and creatinine, using well-established HPLC method for their determination. Results showed that PMF patients have dramatic depletions of both ascorbic acid and GSH (37.3- and 3.81-times lower circulating concentrations, respectively, than those recorded in healthy controls, p < 0.0001), accompanied by significant increases in malondialdehyde (MDA) and nitrite + nitrate (4.73- and 1.66-times higher circulating concentrations, respectively, than those recorded in healthy controls, p < 0.0001). Additionally, PMF patients have remarkable alterations of circulating purines, pyrimidines, and creatinine, suggesting potential mitochondrial dysfunctions causing energy metabolism imbalance and consequent increases in these cell energy-related compounds. Overall, these results, besides evidencing previously unknown serum metabolic alterations in PMF patients, suggest that the determination of serum levels of the aforementioned compounds may be useful to evaluate PMF patients on hospital admission for adjunctive therapies aimed at recovering their correct antioxidant status, as well as to monitor patients' status and potential pharmacological treatments.
Collapse
Affiliation(s)
- Renata Mangione
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Catholic University of the Sacred Heart of Rome, Largo F. Vito 1, 00168 Rome, Italy;
- Departmental Faculty of Medicine, UniCamillus—Saint Camillus International University of Health and Medical Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy; (A.G.); (G.L.)
| | - Cesarina Giallongo
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, Division of Hematology, University of Catania, Via S. Sofia 87, 95123 Catania, Italy; (C.G.); (S.G.); (G.A.P.)
| | - Andrea Duminuco
- Hematology Unit with BMT, A.O.U. Policlinico “G.Rodolico-San Marco”, Via S. Sofia 78, 95123 Catania, Italy;
| | - Enrico La Spina
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (E.L.S.); (L.L.); (D.T.); (G.L.); (M.W.S.); (A.S.)
| | - Lucia Longhitano
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (E.L.S.); (L.L.); (D.T.); (G.L.); (M.W.S.); (A.S.)
| | - Sebastiano Giallongo
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, Division of Hematology, University of Catania, Via S. Sofia 87, 95123 Catania, Italy; (C.G.); (S.G.); (G.A.P.)
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (E.L.S.); (L.L.); (D.T.); (G.L.); (M.W.S.); (A.S.)
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (E.L.S.); (L.L.); (D.T.); (G.L.); (M.W.S.); (A.S.)
| | - Miriam Wissam Saab
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (E.L.S.); (L.L.); (D.T.); (G.L.); (M.W.S.); (A.S.)
| | - Arianna Sbriglione
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (E.L.S.); (L.L.); (D.T.); (G.L.); (M.W.S.); (A.S.)
| | - Giuseppe A. Palumbo
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, Division of Hematology, University of Catania, Via S. Sofia 87, 95123 Catania, Italy; (C.G.); (S.G.); (G.A.P.)
| | - Andrea Graziani
- Departmental Faculty of Medicine, UniCamillus—Saint Camillus International University of Health and Medical Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy; (A.G.); (G.L.)
| | - Amer M. Alanazi
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Valentina Di Pietro
- Neurotrauma and Ophthalmology Research Group, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK;
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital, Edgbaston, Birmingham B15 2TH, UK
| | - Barbara Tavazzi
- Departmental Faculty of Medicine, UniCamillus—Saint Camillus International University of Health and Medical Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy; (A.G.); (G.L.)
| | - Angela Maria Amorini
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (E.L.S.); (L.L.); (D.T.); (G.L.); (M.W.S.); (A.S.)
| | - Giacomo Lazzarino
- Departmental Faculty of Medicine, UniCamillus—Saint Camillus International University of Health and Medical Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy; (A.G.); (G.L.)
| |
Collapse
|
42
|
Farhan K, Tariq MB, Saddiq A, Mansoor M, Ahmed A, Mohammed AS. From Antioxidant to Anticancer is Vitamin C Effective in Colorectal Cancer? Asian Pac J Cancer Prev 2024; 25:1107-1108. [PMID: 38679968 PMCID: PMC11162709 DOI: 10.31557/apjcp.2024.25.4.1107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/13/2024] [Indexed: 05/01/2024] Open
|
43
|
Shen X, Wang J, Deng B, Chen S, John C, Zhao Z, Sinha N, Haag J, Sun W, Kong W, Spasojevic I, Batinic-Haberle I, Secord AA, Zhou C, Bae-Jump VL. High-dose ascorbate exerts anti-tumor activities and improves inhibitory effect of carboplatin through the pro-oxidant function pathway in uterine serous carcinoma cell lines. Gynecol Oncol 2024; 183:93-102. [PMID: 38555710 DOI: 10.1016/j.ygyno.2024.03.021] [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: 01/16/2024] [Revised: 03/05/2024] [Accepted: 03/21/2024] [Indexed: 04/02/2024]
Abstract
OBJECTIVE Uterine serous carcinoma is a highly aggressive non-endometrioid subtype of endometrial cancer with poor survival rates overall, creating a strong need for new therapeutic strategies to improve outcomes. High-dose ascorbate (vitamin C) has been shown to inhibit cell proliferation and tumor growth in multiple preclinical models and has shown promising anti-tumor activity in combination with chemotherapy, with a favorable safety profile. We aimed to study the anti-tumor effects of ascorbate and its synergistic effect with carboplatin on uterine serous carcinoma cells. METHODS Cell proliferation was evaluated by MTT and colony formation assays in ARK1, ARK2 and SPEC2 cells. Cellular stress, antioxidant ability, cleaved caspase 3 activity and adhesion were measured by ELISA assays. Cell cycle was detected by Cellometer. Invasion was measured using a wound healing assay. Changes in protein expression were determined by Western immunoblotting. RESULTS High-dose ascorbate significantly inhibited cell proliferation, caused cell cycle arrest, induced cellular stress, and apoptosis, increased DNA damage, and suppressed cell invasion in ARK1 and SPEC2 cells. Treatment of both cells with 1 mM N-acetylcysteine reversed ascorbate-induced apoptosis and inhibition of cell proliferation. The combination of ascorbate and carboplatin produced significant synergistic effects in inhibiting cell proliferation and invasion, inducing cellular stress, causing DNA damage, and enhancing cleaved caspase 3 levels compared to each compound alone in both cells. CONCLUSIONS Ascorbate has potent antitumor activity and acts synergistically with carboplatin through its pro-oxidant effects. Clinical trials of ascorbate combined with carboplatin as adjuvant treatment of uterine serous carcinoma are worth exploring.
Collapse
Affiliation(s)
- Xiaochang Shen
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, PR China; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jiandong Wang
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, PR China
| | - Boer Deng
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, PR China; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shuning Chen
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, PR China; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Catherine John
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ziyi Zhao
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, PR China; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nikita Sinha
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer Haag
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Wenchuan Sun
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Weimin Kong
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, PR China
| | - Ivan Spasojevic
- Department of Medicine, Duke University School of Medicine, and PK/PD Core Laboratory, Duke Cancer Institute, Durham, NC 27710, USA
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Angeles Alvarez Secord
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Duke Cancer Institute, Duke University, Durham, NC 27710, USA
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Victoria L Bae-Jump
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
44
|
Wu K, El Zowalaty AE, Sayin VI, Papagiannakopoulos T. The pleiotropic functions of reactive oxygen species in cancer. NATURE CANCER 2024; 5:384-399. [PMID: 38531982 DOI: 10.1038/s43018-024-00738-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 01/19/2024] [Indexed: 03/28/2024]
Abstract
Cellular redox homeostasis is an essential, dynamic process that ensures the balance between reducing and oxidizing reactions within cells and thus has implications across all areas of biology. Changes in levels of reactive oxygen species can disrupt redox homeostasis, leading to oxidative or reductive stress that contributes to the pathogenesis of many malignancies, including cancer. From transformation and tumor initiation to metastatic dissemination, increasing reactive oxygen species in cancer cells can paradoxically promote or suppress the tumorigenic process, depending on the extent of redox stress, its spatiotemporal characteristics and the tumor microenvironment. Here we review how redox regulation influences tumorigenesis, highlighting therapeutic opportunities enabled by redox-related alterations in cancer cells.
Collapse
Affiliation(s)
- Katherine Wu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Ahmed Ezat El Zowalaty
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Volkan I Sayin
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Thales Papagiannakopoulos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA.
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
45
|
Man S, Bi J, Liu F, Xie W, Ma L. Vitamin C Inhibited Pulmonary Metastasis through Activating Nrf2/HO-1 Pathway. Mol Nutr Food Res 2024; 68:e2300706. [PMID: 38419398 DOI: 10.1002/mnfr.202300706] [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: 10/03/2023] [Revised: 12/12/2023] [Indexed: 03/02/2024]
Abstract
As an important nutritional component, vitamin C (Vc) shows good antitumor activity in a variety of cancer, but there are few studies in pulmonary metastasis. In order to verify its anticancer and antimetastatic effect, the study sets up H22 pulmonary metastasis mouse model. The results show that intraperitoneal injection of Vc inhibits pulmonary metastasis through up-regulating the expression of Nrf2, HO-1, cleaved caspases 3 and 9, and causing DNA damage and apoptosis which is similar to the pro-oxidant effect of Vc in p53 null cells (H1299 cells). Meanwhile, oral administration of Vc up-regulates the expression of p53, directly activates Nrf2/HO-1 pathway, increases expression of cleaved caspases 3 and 9, and ultimately inhibits pulmonary metastasis, which is the same as the antioxidant result of Vc in p53 wild-type cells. In addition, Vc inhibits the proliferation and migration of lung cancer cells in a concentration-dependent manner and has little cytotoxic effects on normal cells. Notably, the experiment further illustrates that besides intravenous Vc, oral Vc significantly inhibits the pulmonary metastasis in mice. All in all, these findings provide new clues for Vc-treated pulmonary metastasis in clinical research.
Collapse
Affiliation(s)
- Shuli Man
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Jingxian Bi
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Furui Liu
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Wenwen Xie
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| |
Collapse
|
46
|
Hernández-Prieto JH, Martini VP, Iulek J. Structure of glyceraldehyde-3-phosphate dehydrogenase from Paracoccidioides lutzii in complex with an aldonic sugar acid. Biochimie 2024; 218:20-33. [PMID: 37709188 DOI: 10.1016/j.biochi.2023.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 07/23/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
The pathogen Paracoccidioides lutzii (Pb01) is found in South America countries Colombia, Ecuador, Venezuela and Brazil, especially in the central, west, and north regions of the latter. It belongs to the Ajellomycetaceae family, Onygenales order, and is typically thermodimorphic, presenting yeast cells when it grows in animal tissues, but mycelia when in the environment, where it produces the infectious propagule. This fungus is one of the etiologic agents of Paracoccidioidomycosis (PCM), the most important endemic fungal infection in Latin America. Investigations on its genome have contributed to a better understanding about its metabolism and revealed the complexity of several metabolic glycolytic pathways. Glyceraldehyde-3-Phosphate Dehydrogenase from Paracoccidioides lutzii (PlGAPDH) is considered a moonlighting protein and participates in several biological processes of this pathogen. The enzyme was expressed and purified, as seen in SDS-PAGE gel, crystallized and had its three dimensional structure (3D) determined in complex with NAD+, a sulphate ion and d-galactonic acid, therefore, a type of 'GAA site'. It is the first GAPDH structure to show this chemical type in this site and how this protein can bind an acid derived from oxidation of a linear hexose.
Collapse
Affiliation(s)
| | | | - Jorge Iulek
- Department of Chemistry, State University of Ponta Grossa, Ponta Grossa, PR, 84030-900, Brazil.
| |
Collapse
|
47
|
Begimbetova D, Burska AN, Baltabekova A, Kussainova A, Kukanova A, Fazyl F, Ibragimova M, Manekenova K, Makishev A, Bersimbaev RI, Sarbassov DD. The Vitamin C Enantiomers Possess a Comparable Potency in the Induction of Oxidative Stress in Cancer Cells but Differ in Their Toxicity. Int J Mol Sci 2024; 25:2531. [PMID: 38473779 DOI: 10.3390/ijms25052531] [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: 12/26/2023] [Revised: 02/07/2024] [Accepted: 02/10/2024] [Indexed: 03/14/2024] Open
Abstract
The use of vitamin C (VC) in high doses demonstrates a potent tumor suppressive effect by mediating a glucose-dependent oxidative stress in Kirsten rat sarcoma (KRAS) mutant cancer cells. VC with arsenic trioxide (ATO) is a promising drug combination that might lead to the development of effective cancer therapeutics. Considering that a tumor suppressive effect of VC requires its high-dose administration, it is of interest to examine the toxicity of two enantiomers of VC (enantiomer d-optical isomer D-VC and natural l-optical isomer L-VC) in vitro and in vivo. We show that the combinations of L-VC with ATO and D-VC with ATO induced a similar cytotoxic oxidative stress in KrasG12D-expressing mutant cancer cells as indicated by a substantial increase in reactive oxidative species (ROS) production and depolarization of mitochondria. To examine the L-VC and D-VC toxicity effects, we administered high doses of D-VC and L-VC to CD1 mice and carried out an evaluation of their toxic effects. The daily injections of L-VC at a dose of 9.2 g/kg for 18 days were lethal to mice, while 80% of mice remained alive following the similar high-dose administration of D-VC. Following the drug injection courses and histopathological studies, we determined that a natural form of VC (L-VC) is more harmful and toxic to mice when compared to the effects caused by the similar doses of D-VC. Thus, our study indicates that the two enantiomers of VC have a similar potency in the induction of oxidative stress in cancer cells, but D-VC has a distinctive lower toxicity in mice compared to L-VC. While the mechanism of a distinctive toxicity between D-VC and L-VC is yet to be defined, our finding marks D-VC as a more preferable option compared to its natural enantiomer L-VC in clinical settings.
Collapse
Affiliation(s)
- Dinara Begimbetova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
| | - Agata N Burska
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
- School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan
| | | | - Assiya Kussainova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
| | - Assiya Kukanova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
- Department of Oncology, Astana Medical University, Astana 010000, Kazakhstan
| | - Fatima Fazyl
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
| | - Milana Ibragimova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
| | - Kenzhekyz Manekenova
- Department of Pathological Anatomy, Astana Medical University, Astana 010000, Kazakhstan
| | - Abay Makishev
- Department of Oncology, Astana Medical University, Astana 010000, Kazakhstan
| | - Rakhmetkazhi I Bersimbaev
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
| | - Dos D Sarbassov
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan
| |
Collapse
|
48
|
Wernhart S, Rassaf T. Exercise, cancer, and the cardiovascular system: clinical effects and mechanistic insights. Basic Res Cardiol 2024:10.1007/s00395-024-01034-4. [PMID: 38353711 DOI: 10.1007/s00395-024-01034-4] [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: 12/15/2023] [Revised: 01/21/2024] [Accepted: 01/21/2024] [Indexed: 03/05/2024]
Abstract
Cardiovascular diseases and cancer are the leading causes of death in the Western world and share common risk factors. Reduced cardiorespiratory fitness (CRF) is a major determinant of cardiovascular morbidity and cancer survival. In this review we discuss cancer- induced disturbances of parenchymal, cellular, and mitochondrial function, which limit CRF and may be antagonized and attenuated through exercise training. We show the impact of CRF on cancer survival and its attenuating effects on cardiotoxicity of cancer-related treatment. Tailored exercise programs are not yet available for each tumor entity as several trials were performed in heterogeneous populations without adequate cardiopulmonary exercise testing (CPET) prior to exercise prescription and with a wide variation of exercise modalities. There is emerging evidence that exercise may be a crucial pillar in cancer treatment and a tool to mitigate cardiotoxic treatment effects. We discuss modalities of aerobic exercise and resistance training and their potential to improve CRF in cancer patients and provide an example of a periodization model for exercise training in cancer.
Collapse
Affiliation(s)
- Simon Wernhart
- West German Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | - Tienush Rassaf
- West German Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| |
Collapse
|
49
|
Muchtaridi M, Az-Zahra F, Wongso H, Setyawati LU, Novitasari D, Ikram EHK. Molecular Mechanism of Natural Food Antioxidants to Regulate ROS in Treating Cancer: A Review. Antioxidants (Basel) 2024; 13:207. [PMID: 38397805 PMCID: PMC10885946 DOI: 10.3390/antiox13020207] [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: 12/02/2023] [Revised: 01/27/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Cancer is the second-highest mortality rate disease worldwide, and it has been estimated that cancer will increase by up to 20 million cases yearly by 2030. There are various options of treatment for cancer, including surgery, radiotherapy, and chemotherapy. All of these options have damaging adverse effects that can reduce the patient's quality of life. Cancer itself arises from a series of mutations in normal cells that generate the ability to divide uncontrollably. This cell mutation can happen as a result of DNA damage induced by the high concentration of ROS in normal cells. High levels of reactive oxygen species (ROS) can cause oxidative stress, which can initiate cancer cell proliferation. On the other hand, the cytotoxic effect from elevated ROS levels can be utilized as anticancer therapy. Some bioactive compounds from natural foods such as fruit, vegetables, herbs, honey, and many more have been identified as a promising source of natural antioxidants that can prevent oxidative stress by regulating the level of ROS in the body. In this review, we have highlighted and discussed the benefits of various natural antioxidant compounds from natural foods that can regulate reactive oxygen species through various pathways.
Collapse
Affiliation(s)
- Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.A.-Z.); (L.U.S.); (D.N.)
- Research Collaboration Centre for Radiopharmaceuticals Theranostic, National Research and Innovation Agency (BRIN), Jln. Raya Bandung Sumedang Km. 21, Jatinangor 45363, Indonesia;
| | - Farhah Az-Zahra
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.A.-Z.); (L.U.S.); (D.N.)
| | - Hendris Wongso
- Research Collaboration Centre for Radiopharmaceuticals Theranostic, National Research and Innovation Agency (BRIN), Jln. Raya Bandung Sumedang Km. 21, Jatinangor 45363, Indonesia;
- Research Center for Radioisotope, Radiopharmaceutical and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency (BRIN), Jl. Puspiptek, Kota Tangerang 15314, Indonesia
| | - Luthfi Utami Setyawati
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.A.-Z.); (L.U.S.); (D.N.)
- Research Collaboration Centre for Radiopharmaceuticals Theranostic, National Research and Innovation Agency (BRIN), Jln. Raya Bandung Sumedang Km. 21, Jatinangor 45363, Indonesia;
| | - Dhania Novitasari
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.A.-Z.); (L.U.S.); (D.N.)
| | - Emmy Hainida Khairul Ikram
- Integrated Nutrition Science and Therapy Research Group (INSPIRE), Faculty of Health Sciences, Universiti Teknologi MARA Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam 42300, Malaysia;
| |
Collapse
|
50
|
Fan H, Xu Z, Yao K, Zheng B, Zhang Y, Wang X, Zhang T, Li X, Hu H, Yue B, Hu Z, Zheng H. Osteoclast Cancer Cell Metabolic Cross-talk Confers PARP Inhibitor Resistance in Bone Metastatic Breast Cancer. Cancer Res 2024; 84:449-467. [PMID: 38038966 DOI: 10.1158/0008-5472.can-23-1443] [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: 05/14/2023] [Revised: 10/11/2023] [Accepted: 11/28/2023] [Indexed: 12/02/2023]
Abstract
The majority of patients with late-stage breast cancer develop distal bone metastases. The bone microenvironment can affect response to therapy, and uncovering the underlying mechanisms could help identify improved strategies for treating bone metastatic breast cancer. Here, we observed that osteoclasts reduced the sensitivity of breast cancer cells to DNA damaging agents, including cisplatin and the PARP inhibitor (PARPi) olaparib. Metabolic profiling identified elevated glutamine production by osteoclasts. Glutamine supplementation enhanced the survival of breast cancer cells treated with DNA damaging agents, while blocking glutamine uptake increased sensitivity and suppressed bone metastasis. GPX4, the critical enzyme responsible for glutathione oxidation, was upregulated in cancer cells following PARPi treatment through stress-induced ATF4-dependent transcriptional programming. Increased glutamine uptake and GPX4 upregulation concertedly enhanced glutathione metabolism in cancer cells to help neutralize oxidative stress and generate PARPi resistance. Analysis of paired patient samples of primary breast tumors and bone metastases revealed significant induction of GPX4 in bone metastases. Combination therapy utilizing PARPi and zoledronate, which blocks osteoclast activity and thereby reduces the microenvironmental glutamine supply, generated a synergistic effect in reducing bone metastasis. These results identify a role for glutamine production by bone-resident cells in supporting metastatic cancer cells to overcome oxidative stress and develop resistance to DNA-damaging therapies. SIGNIFICANCE Metabolic interaction between osteoclasts and tumor cells contributes to resistance to DNA-damaging agents, which can be blocked by combination treatment with PARP and osteoclast inhibitors to reduce bone metastatic burden.
Collapse
Affiliation(s)
- Huijuan Fan
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Zhanao Xu
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Ke Yao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Bingxin Zheng
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yuan Zhang
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Xuxiang Wang
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Tengjiang Zhang
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Xuan Li
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Haitian Hu
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Bin Yue
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Hanqiu Zheng
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|